Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Carcinogenesis ; 41(1): 36-43, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31058919

ABSTRACT

Reactive oxygen species (ROS) generated during cellular respiration oxidize various cellular constituents, which cause carcinogenesis. Because most studies on the role of ROS in carcinogenesis have mainly been performed using tumor-derived cell lines, which harbor various types of mutation, it has been difficult to determine the molecular details that lead to cancer formation. To overcome this difficulty, we established human-induced pluripotent stem cell lines in which the intracellular ROS levels are controlled at various differentiation stages by manipulating the ROS-yielding mitochondria. By introducing a specific amino acid substitution (I69E) into the succinate dehydrogenase complex, subunit C protein, a component of mitochondrial respiratory chain complex II, the ROS level increased considerably. When ROS-overproducing cells at the early stage of endoderm differentiation were subcutaneously inoculated into the backs of nude mice, we observed tumor formation. These tumor-initiating cells were subjected to a comprehensive analysis by RNA sequencing. It was revealed that tumor-initiating cells showed 27 upregulated transcripts compared with control cells. The newly identified genes include those coding for PAX8 and FOSB (transcription factors) as well as FGF22, whose expressions are known to increase in developing embryos. These results suggest that these genes may play a pivotal role in cancer formation at the very early stages of cell differentiation.


Subject(s)
Cell Transformation, Neoplastic/pathology , Induced Pluripotent Stem Cells/pathology , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Reactive Oxygen Species/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Female , Fibroblast Growth Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasms/genetics , Oxidation-Reduction , PAX8 Transcription Factor/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, RNA , Up-Regulation , Xenograft Model Antitumor Assays
2.
Cereb Cortex ; 24(9): 2476-88, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23595620

ABSTRACT

Diabetes mellitus (DM) is considered to be a risk factor for dementia including Alzheimer's disease (AD). However, the molecular mechanism underlying this risk is not well understood. We examined gene expression profiles in postmortem human brains donated for the Hisayama study. Three-way analysis of variance of microarray data from frontal cortex, temporal cortex, and hippocampus was performed with the presence/absence of AD and vascular dementia, and sex, as factors. Comparative analyses of expression changes in the brains of AD patients and a mouse model of AD were also performed. Relevant changes in gene expression identified by microarray analysis were validated by quantitative real-time reverse-transcription polymerase chain reaction and western blotting. The hippocampi of AD brains showed the most significant alteration in gene expression profile. Genes involved in noninsulin-dependent DM and obesity were significantly altered in both AD brains and the AD mouse model, as were genes related to psychiatric disorders and AD. The alterations in the expression profiles of DM-related genes in AD brains were independent of peripheral DM-related abnormalities. These results indicate that altered expression of genes related to DM in AD brains is a result of AD pathology, which may thereby be exacerbated by peripheral insulin resistance or DM.


Subject(s)
Alzheimer Disease/metabolism , Frontal Lobe/metabolism , Hippocampus/metabolism , Temporal Lobe/metabolism , Animals , Dementia, Vascular/metabolism , Diabetes Mellitus/genetics , Disease Models, Animal , Female , Gene Expression , Humans , Male , Mice, Transgenic
3.
Am J Pathol ; 181(4): 1378-86, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22841817

ABSTRACT

Retinitis pigmentosa (RP) is a genetically heterogenous group of inherited retinal degenerative diseases resulting from photoreceptor cell death and affecting >1 million persons globally. Although oxidative stress has been implicated in the pathogenesis of RP, the mechanisms by which oxidative stress mediates photoreceptor cell death are largely unknown. Here, we show that oxidation of nucleic acids is a key component in the initiation of death-signaling pathways in rd10 mice, a model of RP. Accumulation of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG) increased in photoreceptor cells, and especially within their nuclei, in rd10 mice as well as in Royal College of Surgeons rats, another model of RP caused by different genetic mutations. Vitreous samples from humans with RP contained higher levels of 8-oxo-dG excreted than samples from nondegenerative controls. Transgenic overexpression of human MutT homolog-1, which hydrolyzes oxidized purine nucleoside triphosphates in the nucleotide pool, significantly attenuated 8-oxo-dG accumulation in nuclear DNA and photoreceptor cell death in rd10 mice, in addition to suppressing DNA single-strand break formation, poly(ADP-ribose) polymerase activation, and nuclear translocation of apoptosis-inducing factor. These findings indicate that oxidative DNA damage is an important process for the triggering of photoreceptor cell death in rd10 mice and suggest that stimulation of DNA repair enzymes may be a novel therapeutic approach to attenuate photoreceptor cell loss in RP.


Subject(s)
DNA Damage , DNA Repair Enzymes/metabolism , Inheritance Patterns/genetics , Phosphoric Monoester Hydrolases/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Animals , Apoptosis Inducing Factor/metabolism , Calpain/metabolism , Cell Death , Cell Nucleus/metabolism , DNA Breaks, Single-Stranded , Disease Models, Animal , Enzyme Activation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Protein Transport , Rats , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Signal Transduction
4.
EMBO J ; 27(2): 421-32, 2008 Jan 23.
Article in English | MEDLINE | ID: mdl-18188152

ABSTRACT

Oxidative base lesions, such as 8-oxoguanine (8-oxoG), accumulate in nuclear and mitochondrial DNAs under oxidative stress, resulting in cell death. However, it is not known which form of DNA is involved, whether nuclear or mitochondrial, nor is it known how the death order is executed. We established cells which selectively accumulate 8-oxoG in either type of DNA by expression of a nuclear or mitochondrial form of human 8-oxoG DNA glycosylase in OGG1-null mouse cells. The accumulation of 8-oxoG in nuclear DNA caused poly-ADP-ribose polymerase (PARP)-dependent nuclear translocation of apoptosis-inducing factor, whereas that in mitochondrial DNA caused mitochondrial dysfunction and Ca2+ release, thereby activating calpain. Both cell deaths were triggered by single-strand breaks (SSBs) that had accumulated in the respective DNAs, and were suppressed by knockdown of adenine DNA glycosylase encoded by MutY homolog, thus indicating that excision of adenine opposite 8-oxoG lead to the accumulation of SSBs in each type of DNA. SSBs in nuclear DNA activated PARP, whereas those in mitochondrial DNA caused their depletion, thereby initiating the two distinct pathways of cell death.


Subject(s)
Cell Nucleus/genetics , DNA Damage , DNA, Mitochondrial/metabolism , Signal Transduction , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis Inducing Factor/metabolism , Blotting, Western , Calcium/metabolism , Caspases/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Death/physiology , Cell Line , Cell Nucleus/metabolism , Comet Assay , DNA Breaks, Single-Stranded , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Humans , Mice , Mutation , Oxidation-Reduction , Oxidative Stress/drug effects , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/genetics , Transfection , Vitamin K 3/pharmacology
5.
Cell Death Discov ; 8(1): 150, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35365611

ABSTRACT

Oxidative stress plays a pivotal role in the differentiation and proliferation of cells and programmed cell death. However, studies on the role of oxidative stress in differentiation have mainly employed the detection of reactive oxygen species (ROS) during differentiation or generated by ROS inducers. Therefore, it is difficult to clarify the significance of endogenous ROS production in the differentiation of human cells. We developed a system to control the intracellular level of ROS in the initial stage of differentiation in human iPS cells. By introducing a specific substitution (I69E) into the SDHC protein, a component of the mitochondrial respiratory chain complex, the endogenous ROS level increased. This caused impaired endoderm differentiation of iPS cells, and this impairment was reversed by overproduction of mitochondrial-targeted catalase, an anti-oxidant enzyme. Expression of tumor-related FOXC1 transcription factor increased transiently as early as 4 h after ROS-overproduction in the initial stage of differentiation. Knockdown of FOXC1 markedly improved impaired endoderm differentiation, suggesting that endogenous ROS production in the early differentiation state suppresses endoderm differentiation via transient FOXC1 expression.

6.
Cancer Sci ; 102(4): 677-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21235684

ABSTRACT

8-oxoguanine is a major base lesion in DNA or in nucleotides caused by oxidative stress, and is highly mutagenic because it can pair with adenine as well as cytosine. Adenine DNA glycosylase, encoded by the human mutY homolog gene, MUTYH, excises adenine in the nascent strand when inserted opposite 8-oxoguanine in template DNA, and thus suppresses mutagenesis caused by 8-oxoguanine that has accumulated in DNA due to oxidative stress. Several germ-line mutations in MUTYH are predisposed to MUTYH-associated polyposis, an autosomal recessive disorder characterized by multiple colorectal adenomas and carcinomas. Loss of function of MUTYH leads to an accumulation of somatic mutations in APC and KRAS genes, resulting in the development of adenomas/carcinomas. We recently demonstrated that accumulation of 8-oxoguanine in nuclear and mitochondrial DNA triggers two distinct cell death pathways that are independent of each other. Both pathways are initiated by the accumulation of MUTYH-generated single-strand breaks (SSBs) in nuclear or mitochondrial DNA. Our findings indicate that MUTYH-induced cell death due to oxidative stress results in an efficient elimination of mutagenic cells that have accumulated high levels of 8-oxoguanine in their DNAs. It is most likely that loss of function of MUTYH in stem or progenitor cells in the intestinal epithelium of MUTYH-associated polyposis patients results in escape from programmed cell death; however, accumulated 8-oxoguanine causes various mutations in APC or KRAS genes in these proliferative cells, thereby promoting tumorigenesis. We thus propose that MUTYH suppresses tumorigenesis under conditions of oxidative stress by inducing cell death and by suppressing mutagenesis.


Subject(s)
Apoptosis , Cell Transformation, Neoplastic , DNA Glycosylases/physiology , Neoplasms/etiology , Neoplasms/prevention & control , Oxidative Stress , Germ-Line Mutation/genetics , Humans , Neoplasms/enzymology
7.
Sci Rep ; 11(1): 5819, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33758207

ABSTRACT

8-Oxoguanine (8-oxoG), a major oxidative base lesion, is highly accumulated in Alzheimer's disease (AD) brains during the pathogenic process. MTH1 hydrolyzes 8-oxo-dGTP to 8-oxo-dGMP, thereby avoiding 8-oxo-dG incorporation into DNA. 8-OxoG DNA glycosylase-1 (OGG1) excises 8-oxoG paired with cytosine in DNA, thereby minimizing 8-oxoG accumulation in DNA. Levels of MTH1 and OGG1 are significantly reduced in the brains of sporadic AD cases. To understand how 8-oxoG accumulation in the genome is involved in AD pathogenesis, we established an AD mouse model with knockout of Mth1 and Ogg1 genes in a 3xTg-AD background. MTH1 and OGG1 deficiency increased 8-oxoG accumulation in nuclear and, to a lesser extent, mitochondrial genomes, causing microglial activation and neuronal loss with impaired cognitive function at 4-5 months of age. Furthermore, minocycline, which inhibits microglial activation and reduces neuroinflammation, markedly decreased the nuclear accumulation of 8-oxoG in microglia, and inhibited microgliosis and neuronal loss. Gene expression profiling revealed that MTH1 and OGG1 efficiently suppress progression of AD by inducing various protective genes against AD pathogenesis initiated by Aß/Tau accumulation in 3xTg-AD brain. Our findings indicate that efficient suppression of 8-oxoG accumulation in brain genomes is a new approach for prevention and treatment of AD.


Subject(s)
Alzheimer Disease/genetics , DNA Glycosylases/genetics , Guanine/analogs & derivatives , Phosphoric Monoester Hydrolases/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , DNA Damage/drug effects , DNA Repair/drug effects , Disease Progression , Gene Expression Profiling , Guanine/metabolism , Guanine/toxicity , Humans , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Oxidative Stress/drug effects
8.
Mutat Res ; 703(1): 51-8, 2010 Nov 28.
Article in English | MEDLINE | ID: mdl-20542142

ABSTRACT

Accumulation of oxidized bases such as 8-oxoguanine in either nuclear or mitochondrial DNA triggers various cellular dysfunctions including mutagenesis, and programmed cell death or senescence. Recent studies have revealed that oxidized nucleoside triphosphates such as 8-oxo-dGTP in the nucleotide pool are the main source of oxidized bases accumulating in the DNA of cells under oxidative stress. To counteract such deleterious effects of nucleotide pool damage, mammalian cells possess MutT homolog-1 (MTH1) with oxidized purine nucleoside triphosphatase and related enzymes, thus minimizing the accumulation of oxidized bases in cellular DNA. Depletion or increased expression of the MTH1 protein have revealed its significant roles in avoiding programmed cell death or senescence as well as mutagenesis, and accumulating evidences indicate that MTH1 is involved in suppression of degenerative disorders such as neurodegeneration.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Apoptosis , DNA Damage , DNA Repair Enzymes/metabolism , Deoxyguanine Nucleotides/metabolism , Nucleoside-Triphosphatase/metabolism , Phosphoric Monoester Hydrolases/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA, Mitochondrial/metabolism , Guanine/analogs & derivatives , Guanine/metabolism , Humans , Nerve Degeneration , Neurons/metabolism , Oxidative Stress/genetics , Purine Nucleosides/metabolism , Reactive Oxygen Species/metabolism
9.
DNA Repair (Amst) ; 7(3): 418-30, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18155646

ABSTRACT

Human MutT homolog (hMTH1) hydrolyzes oxidized purine nucleoside triphosphates to monophosphates, thereby avoiding incorporation of such oxidized purines into DNA or RNA. We examined whether hMTH1 prevents cellular dysfunction induced by sodium nitroprusside, a spontaneous NO donor. Exposure to sodium nitroprusside caused an 8-oxoguanine (8-oxoG) buildup in DNA of proliferating MTH1-null cells which underwent mitochondrial degeneration and subsequently died. Quiescent MTH1-null cells also died with 8-oxoG buildup but only when the buildup affected mitochondrial and not nuclear DNA. In both proliferative and quiescent conditions, the accumulation of 8-oxoG in DNA and cell death was effectively prevented by hMTH1. Knockdown of MUTYH in quiescent MTH1-null cells significantly prevented the cell death, suggesting that 8-oxoG incorporated into mitochondrial DNA is a main cause of this form of cell death. To verify this possibility, an artificially modified hMTH1, namely mTP-EGFP-hMTH1, which localizes exclusively in mitochondria, was expressed in MTH1-null cells. mTP-EGFP-hMTH1 selectively prevented buildup of 8-oxoG in mitochondrial but not nuclear DNA after exposure of proliferating cells to sodium nitroprusside, and also efficiently prevented cell death. We thus concluded that exposure of cells to sodium nitroprusside causes oxidation of mitochondrial deoxynucleotide pools, and that buildup of oxidized bases in mitochondrial DNA initiates cell death.


Subject(s)
Cell Death/drug effects , Mitochondria/drug effects , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Glycosylases/antagonists & inhibitors , DNA Repair Enzymes/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemistry , Deoxyguanosine/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Green Fluorescent Proteins/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Nitric Oxide/metabolism , Oxidation-Reduction , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/physiology
10.
Methods Mol Biol ; 554: 199-212, 2009.
Article in English | MEDLINE | ID: mdl-19513676

ABSTRACT

8-Oxoguanine (8-oxoG), an oxidized form of guanine, is one of the major mutagenic lesions generated under oxidative stress. Oxidative damage in mitochondrial DNA has been implicated as a causative factor for a wide variety of degenerative diseases as well as for cancer during aging. We established a quantitative method for in situ detection of 8-oxoG in mitochondrial DNA in a single-cell level using a monoclonal antibody. Specific detection of 8-oxoG in mitochondrial DNA was confirmed by pre-treatment of samples with DNase I or MutM, the latter excising 8-oxoG opposite C in DNA. We then analyzed 8-oxoG dynamics in mitochondrial DNA of the wild-type and 8-oxoG DNA glycosylase (OGG1)-deficient mouse cells after exposure to hydrogen peroxide. Intensities for the 8-oxoG immunoreactivity in mitochondrial DNA were increased immediately after the exposure to hydrogen peroxide in both types of cells. The increased intensities returned to basal levels within a few hours only in wild-type cells, but not in OGG1-deficient cells which exhibited the increased intensities even 24 h after the exposure. These results indicate that OGG1 is a major enzyme for excision repair of 8-oxoG in mitochondrial DNA in mouse cells, and that our method described here is appropriate to study 8-oxoG dynamics in mitochondrial DNA.


Subject(s)
DNA, Mitochondrial/chemistry , Fluorescent Antibody Technique/methods , Guanine/analogs & derivatives , Guanine/analysis , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Cells, Cultured , DNA Glycosylases/physiology , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Guanine/chemistry , Hydrogen Peroxide/pharmacology , Mice , Mice, Knockout , Mitochondria/metabolism , NIH 3T3 Cells , Oxidants/pharmacology , Oxidation-Reduction
11.
Methods Mol Biol ; 554: 251-64, 2009.
Article in English | MEDLINE | ID: mdl-19513679

ABSTRACT

Oxidative base lesions, such as 8-oxoguanine, accumulate in nuclear and mitochondrial DNAs under oxidative stress, resulting in cell death. However, it is not known whether only oxidative lesion accumulated in mitochondrial DNA is involved in such cell death. By introducing human cDNA encoding a nuclear form of 8-oxoG DNA glycosylase (hOGG1-1a) into immortalized mouse embryo fibroblasts lacking Ogg1 gene, we established a cell line which selectively accumulates 8-oxoguanine in mitochondrial DNA under oxidative stress. Selective accumulation of 8-oxoguanine in mitochondrial DNA in this cell line causes degradation of mitochondrial DNA followed by ATP depletion, mitochondrial membrane permeability transition, and Ca(2+) efflux, which in turn activates calpains to execute cell death. Knockdown of MUTYH which excises adenine opposite 8-oxoG in DNA prevents degradation of mitochondrial DNA and activation of calpain, thus suppressing the cell death induced by menadione.


Subject(s)
Cell Nucleus/genetics , DNA Damage , DNA Glycosylases/physiology , DNA Repair , DNA, Mitochondrial/genetics , Adenosine Triphosphate/metabolism , Animals , Blotting, Southern , Calcium/metabolism , Calpain/metabolism , Cell Nucleus/metabolism , Cell Survival , Cells, Cultured , DNA, Mitochondrial/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Guanine/analogs & derivatives , Guanine/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress
12.
Mech Ageing Dev ; 161(Pt A): 95-104, 2017 01.
Article in English | MEDLINE | ID: mdl-27233446

ABSTRACT

In normal brain, neurons in the cortex and hippocampus produce insulin, which modulates glucose metabolism and cognitive functions. It has been shown that insulin resistance impairs glucose metabolism and mitochondrial function, thus increasing production of reactive oxygen species. Recent progress in Alzheimer's disease (AD) research revealed that insulin production and signaling are severely impaired in AD brain, thereby resulting in mitochondrial dysfunction and increased oxidative stress. Among possible oxidative DNA lesions, 8-oxoguanine (8-oxoG) is highly accumulated in the brain of AD patients. Previously we have shown that incorporating 8-oxoG in nuclear and mitochondrial DNA promotes MUTYH (adenine DNA glycosylase) dependent neurodegeneration. Moreover, cortical neurons prepared from MTH1 (8-oxo-dGTPase)/OGG1 (8-oxoG DNA glycosylase)-double deficient adult mouse brains is shown to exhibit significantly poor neuritogenesis in vitro with increased 8-oxoG accumulation in mitochondrial DNA in the absence of antioxidants. Therefore, 8-oxoG can be considered involved in the neurodegenerative process in AD brain. In mild cognitive impairment, mitochondrial dysfunction and oxidative damage may induce synaptic dysfunction due to energy failures in neurons thus resulting in impaired cognitive function. If such abnormality lasts long, it can lead to vicious cycles of oxidative damage, which may then trigger the neurodegenerative process seen in Alzheimer type dementia.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Cortex/metabolism , DNA Damage , DNA, Mitochondrial/metabolism , Glucose/metabolism , Mitochondria/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA, Mitochondrial/genetics , Glucose/genetics , Humans , Mitochondria/genetics , Mitochondria/pathology
13.
DNA Repair (Amst) ; 58: 13-20, 2017 10.
Article in English | MEDLINE | ID: mdl-28843610

ABSTRACT

OGG1 (8-oxoguanine-DNA glycosylase) is the major DNA repair glycosylase removing the premutagenic DNA base modification 8-oxo-7,8-dihydroguanine (8-oxoG) from the genome of mammalian cells. In addition, there is accumulating evidence that OGG1 and its substrate 8-oxoG might function in the regulation of certain genes, which could account for an attenuated immune response observed in Ogg1-/- mice in several settings. Indications for at least two different mechanisms have been obtained. Thus, OGG1 could either act as an ancillary transcription factor cooperating with the lysine-specific demethylase LSD1 or as an activator of small GTPases. Here, we analysed the activation by lipopolysaccaride (LPS) of primary splenocytes obtained from two different Ogg1-/- mouse strains. We found that the induction of TNF-α expression was reduced in splenocytes (in particular macrophages) of both Ogg1-/- strains. Notably, an inhibitor of LSD1, OG-L002, reduced the induction of TNF-α mRNA in splenocytes from wild-type mice to the level observed in splenocytes from Ogg1-/- mice and had no influence in the latter cells. In contrast, inhibitors of the MAP kinases p38 and JNK as well as the antioxidant N-acetylcysteine attenuated the LPS-stimulated TNF-α expression both in the absence and presence of OGG1. The free base 8-oxo-7,8-dihydroguanine had no influence on the TNF-α expression in the splenocytes. The data demonstrate that OGG1 plays a role in an LSD1-dependent pathway of LPS-induced macrophage activation in mice.


Subject(s)
DNA Glycosylases/immunology , Spleen/immunology , Tumor Necrosis Factor-alpha/genetics , Animals , DNA/metabolism , DNA Damage , DNA Glycosylases/metabolism , DNA Glycosylases/physiology , DNA Repair , Gene Expression Regulation , Guanine/analogs & derivatives , Guanine/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Spleen/cytology , Spleen/metabolism , Transcription Factors/immunology , Transcription Factors/physiology
14.
Sci Rep ; 6: 22086, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26912170

ABSTRACT

Oxidative stress and mitochondrial dysfunction are implicated in aging-related neurodegenerative disorders. 8-Oxoguanine (8-oxoG), a common oxidised base lesion, is often highly accumulated in brains from patients with neurodegenerative disorders. MTH1 hydrolyses 8-oxo-2'-deoxyguanosine triphosphate (8-oxo-dGTP) to 8-oxo-dGMP and pyrophosphate in nucleotide pools, while OGG1 excises 8-oxoG paired with cytosine in DNA, thereby minimising the accumulation of 8-oxoG in DNA. Mth1/Ogg1-double knockout (TO-DKO) mice are highly susceptible to neurodegeneration under oxidative conditions and show increased accumulation of 8-oxoG in mitochondrial DNA (mtDNA) in neurons, suggesting that 8-oxoG accumulation in mtDNA causes mitochondrial dysfunction. Here, we evaluated the contribution of MTH1 and OGG1 to the prevention of mitochondrial dysfunction during neuritogenesis in vitro. We isolated cortical neurons from adult wild-type and TO-DKO mice and maintained them with or without antioxidants for 2 to 5 days and then examined neuritogenesis. In the presence of antioxidants, both TO-DKO and wild-type neurons exhibited efficient neurite extension and arborisation. However, in the absence of antioxidants, the accumulation of 8-oxoG in mtDNA of TO-DKO neurons was increased resulting in mitochondrial dysfunction. Cells also exhibited poor neurite outgrowth with decreased complexity of neuritic arborisation, indicating that MTH1 and OGG1 are essential for neuritogenesis under oxidative conditions.


Subject(s)
Cerebral Cortex/metabolism , DNA, Mitochondrial/metabolism , Guanine/analogs & derivatives , Mitochondria/metabolism , Neurites/metabolism , Animals , Cells, Cultured , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA, Mitochondrial/genetics , Guanine/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Oxidation-Reduction , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism
15.
Sci Rep ; 6: 37889, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27897204

ABSTRACT

In the mitochondria-mediated vicious cycle of Alzheimer's disease (AD), intracellular amyloid ß (Aß) induces mitochondrial dysfunction and reactive oxygen species, which further accelerate Aß accumulation. This vicious cycle is thought to play a pivotal role in the development of AD, although the molecular mechanism remains unclear. Here, we examined the effects of human mitochondrial transcriptional factor A (hTFAM) on the pathology of a mouse model of AD (3xTg-AD), because TFAM is known to protect mitochondria from oxidative stress through maintenance of mitochondrial DNA (mtDNA). Expression of hTFAM significantly improved cognitive function, reducing accumulation of both 8-oxoguanine, an oxidized form of guanine, in mtDNA and intracellular Aß in 3xTg-AD mice and increasing expression of transthyretin, known to inhibit Aß aggregation. Next, we found that AD model neurons derived from human induced pluripotent stem cells carrying a mutant PSEN1(P117L) gene, exhibited mitochondrial dysfunction, accumulation of 8-oxoguanine and single-strand breaks in mtDNA, and impaired neuritogenesis with a decreased expression of transthyretin, which is known to be downregulated by oxidative stress. Extracellular treatment with recombinant hTFAM effectively suppressed these deleterious outcomes. Moreover, the treatment increased expression of transthyretin, accompanied by reduction of intracellular Aß. These results provide new insights into potential novel therapeutic targets.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/psychology , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Transcription Factors/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Cognition , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Oxidative Stress , Prealbumin/metabolism , Presenilin-1/genetics , Reactive Oxygen Species/metabolism
16.
J Invest Dermatol ; 134(10): 2610-2619, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24732403

ABSTRACT

Reactive oxygen species produced in response to UVR are important in skin tumor development. We have previously reported that deficiency of the Ogg1 gene, encoding the repair enzyme for 8-oxo-7,8-dihydroguanine (8-oxoG), increases skin tumor incidence in mice upon repetitive UVB exposure and modulation of UVB-induced inflammatory response. Spirulina platensis is used as a human food supplement because it contains abundant nutritional and antioxidant components. Therefore, we investigated the inhibitory effects of S. platensis on UVB-induced skin tumor development in Ogg1 knockout-(KO) mice and the wild-type (WT) counterpart. Dietary S. platensis suppressed tumor induction and development in both genotypes compared with our previous data without S. platensis. Induction of erythema and ear swelling, one of the hallmarks of UVB-induced inflammatory responses, was suppressed in the skin of Ogg1-KO mice and albino hairless mice fed with dietary S. platensis. Compared with untreated mice, S. platensis-administered mice showed significantly reduced 8-oxoG formation in the skin after UVB exposure. Moreover, we found that S. platensis effectively downregulated the signal proteins p38 mitogen-activated protein kinase, stress-activated protein kinase/c-Jun N-terminal kinase, and extracellular signal-regulated kinase after UVB exposure especially in Ogg1-KO mice. Our results suggest that S. platensis exerts antitumor effects against UVB irradiation in the skin through its anti-inflammatory and antioxidant effects.


Subject(s)
Dietary Supplements , Neoplasms, Radiation-Induced/prevention & control , Plant Extracts/therapeutic use , Radiodermatitis/prevention & control , Skin Neoplasms/prevention & control , Spirulina , Ultraviolet Rays , Animals , DNA Glycosylases/deficiency , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Disease Models, Animal , Female , Genotype , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/radiation effects , Male , Mice , Mice, Hairless , Mice, Knockout , Mitogen-Activated Protein Kinase 8/metabolism , Neoplasms, Radiation-Induced/metabolism , Neoplasms, Radiation-Induced/pathology , Plant Extracts/pharmacology , Radiodermatitis/metabolism , Radiodermatitis/pathology , Skin/metabolism , Skin/pathology , Skin/radiation effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Radiat Res ; 54(3): 453-66, 2013 May.
Article in English | MEDLINE | ID: mdl-23297316

ABSTRACT

Changes in the thymic microenvironment lead to radiation-induced thymic lymphomagenesis, but the phenomena are not fully understood. Here we show that radiation-induced chromosomal instability and bystander effects occur in thymocytes and are involved in lymphomagenesis in C57BL/6 mice that have been irradiated four times with 1.8-Gy γ-rays. Reactive oxygen species (ROS) were generated in descendants of irradiated thymocytes during recovery from radiation-induced thymic atrophy. Concomitantly, descendants of irradiated thymocytes manifested DNA lesions as revealed by γ-H2AX foci, chromosomal instability, aneuploidy with trisomy 15 and bystander effects on chromosomal aberration induction in co-cultured ROS-sensitive mutant cells, suggesting that the delayed generation of ROS is a primary cause of these phenomena. Abolishing the bystander effect of post-irradiation thymocytes by superoxide dismutase and catalase supports ROS involvement. Chromosomal instability in thymocytes resulted in the generation of abnormal cell clones bearing trisomy 15 and aberrant karyotypes in the thymus. The emergence of thymic lymphomas from the thymocyte population containing abnormal cell clones indicated that clones with trisomy 15 and altered karyotypes were prelymphoma cells with the potential to develop into thymic lymphomas. The oncogene Notch1 was rearranged after the prelymphoma cells were established. Thus, delayed nontargeted radiation effects drive thymic lymphomagenesis through the induction of characteristic changes in intrathymic immature T cells and the generation of prelymphoma cells.


Subject(s)
Bystander Effect/radiation effects , Carcinogenesis/radiation effects , Chromosomal Instability/radiation effects , Lymphoma/metabolism , Radiation Injuries/metabolism , Reactive Oxygen Species/metabolism , Thymus Neoplasms/metabolism , Animals , Carcinogenesis/genetics , Cells, Cultured , Chromosomal Instability/genetics , Dose Fractionation, Radiation , Dose-Response Relationship, Radiation , Female , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Precancerous Conditions , Radiation Dosage , Radiation Injuries/genetics , Radiation Injuries/pathology , Thymocytes/metabolism , Thymocytes/radiation effects , Thymus Neoplasms/genetics , Thymus Neoplasms/pathology
18.
J Clin Invest ; 122(12): 4344-61, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143307

ABSTRACT

8-Oxoguanine (8-oxoG), a common DNA lesion caused by reactive oxygen species, is associated with carcinogenesis and neurodegeneration. Although the mechanism by which 8-oxoG causes carcinogenesis is well understood, the mechanism by which it causes neurodegeneration is unknown. Here, we report that neurodegeneration is triggered by MUTYH-mediated excision repair of 8-oxoG-paired adenine. Mutant mice lacking 8-oxo-2'-deoxyguanosine triphosphate-depleting (8-oxo-dGTP-depleting) MTH1 and/or 8-oxoG-excising OGG1 exhibited severe striatal neurodegeneration, whereas mutant mice lacking MUTYH or OGG1/MUTYH were resistant to neurodegeneration under conditions of oxidative stress. These results indicate that OGG1 and MTH1 are protective, while MUTYH promotes neurodegeneration. We observed that 8-oxoG accumulated in the mitochondrial DNA of neurons and caused calpain-dependent neuronal loss, while delayed nuclear accumulation of 8-oxoG in microglia resulted in PARP-dependent activation of apoptosis-inducing factor and exacerbated microgliosis. These results revealed that neurodegeneration is a complex process caused by 8-oxoG accumulation in the genomes of neurons and microglia. Different signaling pathways were triggered by the accumulation of single-strand breaks in each type of DNA generated during base excision repair initiated by MUTYH, suggesting that suppression of MUTYH may protect the brain under conditions of oxidative stress.


Subject(s)
DNA Glycosylases/physiology , DNA Repair , Guanine/analogs & derivatives , Neurodegenerative Diseases/metabolism , Oxidative Stress , Animals , Apoptosis Inducing Factor/metabolism , Benzamides/pharmacology , Calpain/antagonists & inhibitors , Calpain/metabolism , Cell Nucleus/metabolism , Corpus Striatum/pathology , DNA Breaks, Single-Stranded , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA, Mitochondrial/genetics , Dipeptides/pharmacology , Guanine/metabolism , Guanine/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Mitochondria/metabolism , Motor Activity , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Nitro Compounds , Phosphoric Monoester Hydrolases/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Propionates
SELECTION OF CITATIONS
SEARCH DETAIL