Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genome Res ; 34(1): 47-56, 2024 02 07.
Article in English | MEDLINE | ID: mdl-38290979

ABSTRACT

Oxidative stress-induced DNA damage and its repair systems are related to cancer etiology; however, the molecular basis triggering tumorigenesis is not well understood. Here, we aimed to explore the causal relationship between oxidative stress, somatic mutations in pre-tumor-initiated normal tissues, and tumor incidence in the small intestines of MUTYH-proficient and MUTYH-deficient mice. MUTYH is a base excision repair enzyme associated with human colorectal cancer. Mice were administered different concentrations of potassium bromate (KBrO3; an oxidizing agent)-containing water for 4 wk for mutagenesis studies or 16 wk for tumorigenesis studies. All Mutyh -/- mice treated with >0.1% KBrO3 developed multiple tumors, and the average tumor number increased dose dependently. Somatic mutation analysis of Mutyh -/-/rpsL transgenic mice revealed that G:C  > T:A transversion was the only mutation type correlated positively with KBrO3 dose and tumor incidence. These mutations preferentially occurred at 5'G in GG and GAA sequences in rpsL This characteristic mutation pattern was also observed in the genomic region of Mutyh -/- tumors using whole-exome sequencing. It closely corresponded to signature 18 and SBS36, typically caused by 8-oxo-guanine (8-oxoG). 8-oxoG-induced mutations were sequence context dependent, yielding a biased amino acid change leading to missense and stop-gain mutations. These mutations frequently occurred in critical amino acid codons of known cancer drivers, Apc or Ctnnb1, known for activating Wnt signal pathway. Our results indicate that oxidative stress contributes to increased tumor incidence by elevating the likelihood of gaining driver mutations by increasing 8-oxoG-mediated mutagenesis, particularly under MUTYH-deficient conditions.


Subject(s)
Guanine/analogs & derivatives , Neoplasms , Oxidative Stress , Humans , Mice , Animals , Oxidative Stress/genetics , Mutagenesis , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Mutation , Mice, Transgenic , Neoplasms/genetics , Amino Acids/genetics , DNA Repair
2.
Genes Cells ; 29(9): 746-756, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38964745

ABSTRACT

An autism-associated gene Shank3 encodes multiple splicing isoforms, Shank3a-f. We have recently reported that Shank3a/b-knockout mice were more susceptible to kainic acid-induced seizures than wild-type mice at 4 weeks of age. Little is known, however, about how the N-terminal and ankyrin repeat domains (NT-Ank) of Shank3a/b regulate multiple molecular signals in the developing brain. To explore the functional roles of Shank3a/b, we performed a mass spectrometry-based proteomic search for proteins interacting with GFP-tagged NT-Ank. In this study, NT-Ank was predicted to form a variety of complexes with a total of 348 proteins, in which RNA-binding (n = 102), spliceosome (n = 22), and ribosome-associated molecules (n = 9) were significantly enriched. Among them, an X-linked intellectual disability-associated protein, Nono, was identified as a NT-Ank-binding protein. Coimmunoprecipitation assays validated the interaction of Shank3 with Nono in the mouse brain. In agreement with these data, the thalamus of Shank3a/b-knockout mice aberrantly expressed splicing isoforms of autism-associated genes, Nrxn1 and Eif4G1, before and after seizures with kainic acid treatment. These data indicate that Shank3 interacts with multiple RNA-binding proteins in the postnatal brain, thereby regulating the homeostatic expression of splicing isoforms for autism-associated genes after birth.


Subject(s)
Mice, Knockout , Nerve Tissue Proteins , RNA-Binding Proteins , Animals , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Mice , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Ankyrin Repeat , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , RNA Splicing , Brain/metabolism , Seizures/metabolism , Seizures/genetics , Seizures/chemically induced , Humans , Protein Binding , Mice, Inbred C57BL
3.
J Immunol ; 210(11): 1804-1814, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37074207

ABSTRACT

Somatic hypermutation (SHM) is necessary for Ab diversification and involves error-prone DNA repair of activation-induced cytidine deaminase-induced lesions in germinal center (GC) B cells but can also cause genomic instability. GC B cells express low levels of the DNA repair protein apurinic/apyrimidinic (AP) endonuclease (APE)1 and high levels of its homolog APE2. Reduced SHM in APE2-deficient mice suggests that APE2 promotes SHM, but these GC B cells also exhibit reduced proliferation that could impact mutation frequency. In this study, we test the hypothesis that APE2 promotes and APE1 suppresses SHM. We show how APE1/APE2 expression changes in primary murine spleen B cells during activation, impacting both SHM and class-switch recombination (CSR). High levels of both APE1 and APE2 early after activation promote CSR. However, after 2 d, APE1 levels decrease steadily with each cell division, even with repeated stimulation, whereas APE2 levels increase with each stimulation. When GC-level APE1/APE2 expression was engineered by reducing APE1 genetically (apex1+/-) and overexpressing APE2, bona fide activation-induced cytidine deaminase-dependent VDJH4 intron SHM became detectable in primary B cell cultures. The C terminus of APE2 that interacts with proliferating cell nuclear Ag promotes SHM and CSR, although its ATR-Chk1-interacting Zf-GRF domain is not required. However, APE2 does not increase mutations unless APE1 is reduced. Although APE1 promotes CSR, it suppresses SHM, suggesting that downregulation of APE1 in the GC is required for SHM. Genome-wide expression data compare GC and cultured B cells and new models depict how APE1 and APE2 expression and protein interactions change during B cell activation and affect the balance between accurate and error-prone repair during CSR and SHM.


Subject(s)
B-Lymphocytes , DNA Repair , Animals , Mice , B-Lymphocytes/metabolism , Cell Culture Techniques , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Immunoglobulin Class Switching/genetics , Mutation , Somatic Hypermutation, Immunoglobulin
4.
Nucleic Acids Res ; 49(12): 7154-7163, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34142156

ABSTRACT

Mammalian MutY homologue (MUTYH) is an adenine DNA glycosylase that excises adenine inserted opposite 8-oxoguanine (8-oxoG). The inherited variations in human MUTYH gene are known to cause MUTYH-associated polyposis (MAP), which is associated with colorectal cancer. MUTYH is involved in base excision repair (BER) with proliferating cell nuclear antigen (PCNA) in DNA replication, which is unique and critical for effective mutation-avoidance. It is also reported that MUTYH has a Zn-binding motif in a unique interdomain connector (IDC) region, which interacts with Rad9-Rad1-Hus1 complex (9-1-1) in DNA damage response, and with apurinic/apyrimidinic endonuclease 1 (APE1) in BER. However, the structural basis for the BER pathway by MUTYH and its interacting proteins is unclear. Here, we determined the crystal structures of complexes between mouse MUTYH and DNA, and between the C-terminal domain of mouse MUTYH and human PCNA. The structures elucidated the repair mechanism for the A:8-oxoG mispair including DNA replication-coupled repair process involving MUTYH and PCNA. The Zn-binding motif was revealed to comprise one histidine and three cysteine residues. The IDC, including the Zn-binding motif, is exposed on the MUTYH surface, suggesting its interaction modes with 9-1-1 and APE1, respectively. The structure of MUTYH explains how MAP mutations perturb MUTYH function.


Subject(s)
DNA Glycosylases/chemistry , Adenine , Adenomatous Polyposis Coli/genetics , Amino Acid Motifs , Animals , DNA/chemistry , DNA Glycosylases/genetics , DNA Repair , DNA Replication , Guanine/analogs & derivatives , Humans , Mice , Models, Molecular , Mutation , Proliferating Cell Nuclear Antigen/chemistry , Zinc
5.
J Biol Chem ; 295(34): 12247-12261, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32647012

ABSTRACT

PCBP1, a member of the poly(C)-binding protein (PCBP) family, has the capability of binding heavily oxidized RNA and therefore participates in the cellular response to oxidative conditions, helping to induce apoptosis. There are four other members of this family, PCBP2, PCBP3, PCBP4, and hnRNPK, but it is not known whether they play similar roles. To learn more, we first tested their affinity for an RNA strand carrying two 8-oxoguanine (8-oxoG) residues at sites located in close proximity to each other, representative of a heavily oxidized strand or RNA with one 8-oxoG or none. Among them, only PCBP2 exhibited highly selective binding to RNA carrying two 8-oxoG residues similar to that observed with PCBP1. In contrast, PCBP3, PCBP4, and hnRNPK bound RNA with or without 8-oxoG modifications and exhibited slightly increased binding to the former. Mutations in conserved RNA-binding domains of PCBP2 disrupted the specific interaction with heavily oxidized RNA. We next tested PCBP2 activity in cells. Compared with WT HeLa S3 cells, PCBP2-KO cells established by gene editing exhibited increased apoptosis with increased caspase-3 activity and PARP1 cleavage under oxidative conditions, which were suppressed by the expression of WT PCBP2 but not one of the mutants lacking binding activity. In contrast, PCBP1-KO cells exhibited reduced apoptosis with much less caspase-3 activity and PARP cleavage than WT cells. Our results indicate that PCBP2 as well as PCBP1 bind heavily oxidized RNA; however, the former may counteract PCBP1 to suppress apoptosis under oxidative conditions.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Guanine/analogs & derivatives , RNA-Binding Proteins/metabolism , RNA/metabolism , Caspase 3/genetics , Caspase 3/metabolism , DNA-Binding Proteins/genetics , Guanine/metabolism , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , MCF-7 Cells , Oxidation-Reduction , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA/genetics , RNA-Binding Proteins/genetics
6.
FASEB J ; 34(12): 16601-16621, 2020 12.
Article in English | MEDLINE | ID: mdl-33107105

ABSTRACT

Developmental and epileptic encephalopathy (DEE) represents a group of neurodevelopmental disorders characterized by infantile-onset intractable seizures and unfavorable prognosis of psychomotor development. To date, hundreds of genes have been linked to the onset of DEE. GNAO1 is a DEE-associated gene encoding the alpha-O1 subunit of guanine nucleotide-binding protein (GαO ). Despite the increasing number of reported children with GNAO1 encephalopathy, the molecular mechanisms underlying their neurodevelopmental phenotypes remain elusive. We herein present that co-immunoprecipitation and mass spectrometry analyses identified another DEE-associated protein, SPTAN1, as an interacting partner of GαO . Silencing of endogenous Gnao1 attenuated the neurite outgrowth and calcium-dependent signaling. Inactivation of GNAO1 in human-induced pluripotent stem cells gave rise to anomalous brain organoids that only weakly expressed SPTAN1 and Ankyrin-G. Furthermore, GNAO1-deficient organoids failed to conduct synchronized firing to adjacent neurons. These data indicate that GαO and other DEE-associated proteins organize the cytoskeletal remodeling and functional polarity of neurons in the developing brain.


Subject(s)
Cytoskeleton/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Animals , Brain/metabolism , Brain Diseases/metabolism , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Neurodevelopmental Disorders/metabolism , Neurons/metabolism , Phenotype
7.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998461

ABSTRACT

An imbalance between the production of reactive oxygen species (ROS) and anti-oxidant capacity results in oxidative injury to cellular components and molecules, which in turn disturbs the homeostasis of cells and organs. Although retinitis pigmentosa (RP) is a hereditary disease, non-genetic biological factors including oxidative stress also modulate or contribute to the disease progression. In animal models of RP, the degenerating retina exhibits marked oxidative damage in the nucleic acids, proteins, and lipids, and anti-oxidant treatments substantially suppress photoreceptor cell death and microgliosis. Although the mechanisms by which oxidative stress mediates retinal degeneration have not been fully elucidated, our group has shown that oxidative DNA damage and its defense system are key regulators of microglial activation and photoreceptor degeneration in RP. In this review, we summarize the current evidence regarding oxidative stress in animal models and patients with RP. The clinical efficacy of anti-oxidant treatments for RP has not been fully established. Nevertheless, elucidating key biological processes that underlie oxidative damage in RP will be pivotal to understanding the pathology and developing a potent anti-oxidant strategy that targets specific cell types or molecules under oxidative stress.


Subject(s)
Antioxidants/therapeutic use , Macular Degeneration/metabolism , Microglia/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Retina/metabolism , Retinitis Pigmentosa/metabolism , 8-Hydroxy-2'-Deoxyguanosine/antagonists & inhibitors , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Animals , Catalase/genetics , Catalase/metabolism , DNA Damage , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Disease Models, Animal , Gene Expression Regulation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Macular Degeneration/pathology , Microglia/drug effects , Microglia/pathology , Oxidative Stress/drug effects , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Reactive Oxygen Species/metabolism , Retina/drug effects , Retina/pathology , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
8.
Ann Neurol ; 84(2): 208-224, 2018 08.
Article in English | MEDLINE | ID: mdl-30014510

ABSTRACT

OBJECTIVE: To identify novel autoantibodies for neuropathic pain (NeP). METHODS: We screened autoantibodies that selectively bind to mouse unmyelinated C-fiber type dorsal root ganglion (DRG) neurons using tissue-based indirect immunofluorescence assays (IFA) with sera from 110 NeP patients with various inflammatory and allergic neurologic diseases or other neuropathies, and 50 controls without NeP including 20 healthy subjects and 30 patients with neurodegenerative diseases or systemic inflammatory diseases. IgG purified from IFA-positive patients' sera was subjected to Western blotting (WB) and immunoprecipitation (IP) using mouse DRG lysates. Immunoprecipitates were analyzed by liquid chromatography tandem mass spectrometry (LC-MS/MS) to identify target autoantigens. RESULTS: Antiunmyelinated C-fiber type DRG neuron antibodies were more frequent in patients with NeP than non-NeP subjects (10% vs 0%; p < 0.05). These autoantibodies were all from the IgG2 subclass and colocalized mostly with isolectin B4- and P2X3-positive pain-conducting small neurons but not with S100ß-positive myelinated neurons. WB revealed a common immunoreactive band (approximately 220kDa). IP and LC-MS/MS studies identified plexin D1 as a target autoantigen. Immunoadsorption tests with recombinant human plexin D1 in IFA revealed that all 11 anti-small DRG neuron antibody-positive patients had anti-plexin D1 antibodies. Application of anti-plexin D1 antibody-positive patient sera to cultured DRG neurons increased membrane permeability, leading to cellular swelling. NeP patients with anti-plexin D1 antibodies commonly developed burning pain and current perception threshold abnormalities for C-fibers. Main comorbidities were atopy and collagen-vascular disease. Immunotherapies ameliorated NeP in 7 treated cases. INTERPRETATION: Anti-plexin D1 antibodies are a novel biomarker for immunotherapy-responsive NeP. Ann Neurol 2018;84:208-224.


Subject(s)
Autoantibodies/blood , Cell Adhesion Molecules, Neuronal/blood , Neuralgia/blood , Neuralgia/diagnosis , Adult , Aged , Animals , Biomarkers/blood , Cells, Cultured , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HeLa Cells , Humans , Immunotherapy/methods , Intracellular Signaling Peptides and Proteins , Male , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Middle Aged , Neuralgia/therapy , Retrospective Studies , Young Adult
9.
Adv Exp Med Biol ; 1128: 1-11, 2019.
Article in English | MEDLINE | ID: mdl-31062322

ABSTRACT

The brain or central nervous system (CNS) utilizes a vast amount of energy to sustain its basic functions, and most of the energy in the brain is derived from glucose. Whole-body energy and glucose homeostasis in the periphery of the human body are regulated by insulin, while the brain had been considered as an "insulin-insensitive" organ, because bulk brain glucose uptake is not affected by insulin in either rodents and humans. However, recently it has become clear that the actions of insulin are more widespread in the CNS and are a critical part of normal development, food intake, and energy balance, as well as plasticity throughout adulthood. Moreover, there are substantial evidence demonstrating that brain insulin is derived from pancreas, neurons, and astrocytes. In this chapter, I reviewed recent progress in roles of insulin in the brain, expression of insulin genes, and multiple origins of the brain insulin.


Subject(s)
Brain Chemistry , Brain/physiology , Insulin/physiology , Eating , Energy Metabolism , Humans
10.
Adv Exp Med Biol ; 1128: 27-44, 2019.
Article in English | MEDLINE | ID: mdl-31062324

ABSTRACT

Accumulating clinical data indicates that insulin resistance and diabetes mellitus (DM) are major risk factors for Alzheimer's disease (AD); however, the exact mechanisms on how insulin resistance and DM act as risk factors for AD remain unclear. Recent progress in gene expression profiling of AD brains revealed that brain insulin production and insulin signaling are significantly impaired, indicating that AD brain exhibits a feature of brain diabetes with depletion of brain insulin, which causes mitochondrial dysfunction with increased oxidative stress, thereby increasing sensitivity to peripheral diabetes. Such diabetic condition in early stage of AD brain can be exacerbated by peripheral diabetes, namely, through hyperglycemia, hyperinsulinemia, or impaired insulin response. In this chapter, I reviewed mitochondrial dysfunction and oxidative stress in AD brain and discussed how those events are involved in AD pathogenesis.


Subject(s)
Alzheimer Disease/physiopathology , Brain/physiopathology , Insulin/physiology , Mitochondria/pathology , Oxidative Stress , Diabetes Mellitus/physiopathology , Humans
11.
J Biol Chem ; 292(7): 2785-2794, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28035004

ABSTRACT

The human MutT homolog 1 (hMTH1, human NUDT1) hydrolyzes oxidatively damaged nucleoside triphosphates and is the main enzyme responsible for nucleotide sanitization. hMTH1 recently has received attention as an anticancer target because hMTH1 blockade leads to accumulation of oxidized nucleotides in the cell, resulting in mutations and death of cancer cells. Unlike Escherichia coli MutT, which shows high substrate specificity for 8-oxoguanine nucleotides, hMTH1 has broad substrate specificity for oxidized nucleotides, including 8-oxo-dGTP and 2-oxo-dATP. However, the reason for this broad substrate specificity remains unclear. Here, we determined crystal structures of hMTH1 in complex with 8-oxo-dGTP or 2-oxo-dATP at neutral pH. These structures based on high quality data showed that the base moieties of two substrates are located on the similar but not the same position in the substrate binding pocket and adopt a different hydrogen-bonding pattern, and both triphosphate moieties bind to the hMTH1 Nudix motif (i.e. the hydrolase motif) similarly and align for the hydrolysis reaction. We also performed kinetic assays on the substrate-binding Asp-120 mutants (D120N and D120A), and determined their crystal structures in complex with the substrates. Analyses of bond lengths with high-resolution X-ray data and the relationship between the structure and enzymatic activity revealed that hMTH1 recognizes the different oxidized nucleotides via an exchange of the protonation state at two neighboring aspartate residues (Asp-119 and Asp-120) in its substrate binding pocket. To our knowledge, this mechanism of broad substrate recognition by enzymes has not been reported previously and may have relevance for anticancer drug development strategies targeting hMTH1.


Subject(s)
DNA Repair Enzymes/metabolism , Phosphoric Monoester Hydrolases/metabolism , Crystallography, X-Ray , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , Humans , Kinetics , Mutation , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/genetics , Protein Conformation , Substrate Specificity
12.
Neuropathology ; 38(3): 247-259, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29603402

ABSTRACT

The protein µ-crystallin (CRYM) is a novel component of the marsupial lens that has two functions: it is a key regulator of thyroid hormone transportation and a reductase of sulfur-containing cyclic ketimines. In this study, we examined changes of the expression pattern of CRYM in different rat organs during development using immunohistochemistry and immunoblotting. As CRYM is reportedly expressed in the corticospinal tract, we also investigated CRYM expression in human cases of amyotrophic lateral sclerosis (ALS) using immunohistochemistry. In the rat brain, CRYM was expressed in the cerebral cortex, basal ganglia, hippocampus and corticospinal tract in the early postnatal period. As postnatal development progressed, CRYM expression was restricted to large pyramidal neurons in layers V and VI of the cerebral cortex and pyramidal cells in the deep layer of CA1 in the hippocampus. Even within the same regions, CRYM-positive and negative neurons were distributed in a mosaic pattern. In the kidney, CRYM was expressed in epithelial cells of the proximal tubule and mesenchymal cells of the medulla in the early postnatal period; however, CRYM expression in the medulla was lost as mesenchymal cell numbers decreased with the rapid growth of the medulla. In human ALS brains, we observed marked loss of CRYM in the corticospinal tract, especially distally. Our results suggest that CRYM may play roles in development of cortical and hippocampal pyramidal cells in the early postnatal period, and in the later period, performs cell-specific functions in selected neuronal populations. In the kidney, CRYM may play roles in maturation of renal function. The expression patterns of CRYM may reflect significance of its interactions with T3 or ketimines in these cells and organs. The results also indicate that CRYM may be used as a marker of axonal degeneration in the corticospinal tract.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Crystallins/metabolism , Kidney/metabolism , Pyramidal Tracts/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/growth & development , Brain/pathology , Female , Humans , Kidney/growth & development , Male , Neurons/metabolism , Pyramidal Tracts/pathology , Rats, Sprague-Dawley , Young Adult , mu-Crystallins
14.
Cancer Sci ; 108(1): 108-115, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27761963

ABSTRACT

We previously reported that celecoxib, a selective COX-2 inhibitor, strongly inhibited human colon cancer cell proliferation by suppressing the Wnt/ß-catenin signaling pathway. 2,5-Dimethylcelecoxib (DM-celecoxib), a celecoxib analog that does not inhibit COX-2, has also been reported to have an antitumor effect. In the present study, we elucidated whether DM-celecoxib inhibits intestinal cancer growth, and its underlying mechanism of action. First, we compared the effect of DM-celecoxib with that of celecoxib on the human colon cancer cell lines HCT-116 and DLD-1. 2,5-Dimethylcelecoxib suppressed cell proliferation and inhibited T-cell factor 7-like 2 expression with almost the same strength as celecoxib. 2,5-Dimethylcelecoxib also inhibited the T-cell factor-dependent transcription activity and suppressed the expression of Wnt/ß-catenin target gene products cyclin D1 and survivin. Subsequently, we compared the in vivo effects of celecoxib and DM-celecoxib using the Mutyh-/- mouse model, in which oxidative stress induces multiple intestinal carcinomas. Serum concentrations of orally administered celecoxib and DM-celecoxib elevated to the levels enough to suppress cancer cell proliferation. Repeated treatment with celecoxib and DM-celecoxib markedly reduced the number and size of the carcinomas without showing toxicity. These results suggest that the central mechanism for the anticancer effect of celecoxib derivatives is the suppression of the Wnt/ß-catenin signaling pathway but not the inhibition of COX-2, and that DM-celecoxib might be a better lead compound candidate than celecoxib for the development of novel anticancer drugs.


Subject(s)
Celecoxib/pharmacology , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/pathology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Blood Cell Count , Body Weight/drug effects , Celecoxib/blood , Celecoxib/therapeutic use , Cell Line, Tumor , DNA Glycosylases/deficiency , DNA Glycosylases/genetics , Female , Humans , Intestinal Neoplasms/metabolism , Male , Mice , Oxidative Stress/drug effects , Proteolysis/drug effects , Pyrazoles/blood , Pyrazoles/therapeutic use , Sulfonamides/blood , Sulfonamides/therapeutic use , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Transcription, Genetic/drug effects , beta Catenin/antagonists & inhibitors
15.
Genes Cells ; 21(10): 1030-1048, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27545963

ABSTRACT

Genomewide association studies have shown that a nonsynonymous single nucleotide polymorphism in PRKCH is associated with cerebral infarction and atherosclerosis-related complications. We examined the role of PKCη in lipid metabolism and atherosclerosis using apolipoprotein E-deficient (Apoe-/- ) mice. PKCη expression was augmented in the aortas of mice with atherosclerosis and exclusively detected in MOMA2-positive macrophages within atherosclerotic lesions. Prkch+/+ Apoe-/- and Prkch-/- Apoe-/- mice were fed a high-fat diet (HFD), and the dyslipidemia observed in Prkch+/+ Apoe-/- mice was improved in Prkch-/- Apoe-/- mice, with a particular reduction in serum LDL cholesterol and phospholipids. Liver steatosis, which developed in Prkch+/+ Apoe-/- mice, was improved in Prkch-/- Apoe-/- mice, but glucose tolerance, adipose tissue and body weight, and blood pressure were unchanged. Consistent with improvements in LDL cholesterol, atherosclerotic lesions were decreased in HFD-fed Prkch-/- Apoe-/- mice. Immunoreactivity against 3-nitrotyrosine in atherosclerotic lesions was dramatically decreased in Prkch-/- Apoe-/- mice, accompanied by decreased necrosis and apoptosis in the lesions. ARG2 mRNA and protein levels were significantly increased in Prkch-/- Apoe-/- macrophages. These data show that PKCη deficiency improves dyslipidemia and reduces susceptibility to atherosclerosis in Apoe-/- mice, showing that PKCη plays a role in atherosclerosis development.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Lipid Metabolism , Protein Kinase C/deficiency , Animals , Aorta/metabolism , Apoptosis , Atherosclerosis/pathology , Diet, High-Fat , Disease Susceptibility , Dyslipidemias/metabolism , Fatty Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Oxidative Stress
16.
Neurochem Res ; 42(9): 2658-2665, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28462451

ABSTRACT

Molecular hydrogen (H2), as a new medical gas, has protective effects in neurological disorders including Parkinson's disease (PD). In our previous report, the neuroprotective effect of drinking water with saturated H2 (H2 water) in PD mice might be due to stomach-brain interaction via release of gastric hormone, ghrelin. In the present study, we assessed the effect of H2-induced ghrelin more precisely. To confirm the contribution of ghrelin in H2 water-drinking PD model mice, ghrelin-knock out (KO) mice were used. Despite the speculation, the effect of H2 water was still observed in ghrelin-KO PD model mice. To further check the involvement of ghrelin, possible contribution of ghrelin-induced vagal afferent effect was tested by performing subdiaphragmatic vagotomy before treating with H2 water and administration of MPTP (1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine). The protective effect of H2 water was still observed in the vagotomized mice in substantia nigra, suggesting that stimulation of vagal afferent nerves is not involved in H2-induced neuroprotection. Other neuroprotective substitutes in ghrelin-KO mice were speculated because H2-induced neuroprotection was not cancelled by ghrelin receptor antagonist, D-Lys3 GHRP-6, in ghrelin-KO PD model mice, unlike in wild-type PD model mice. Our results indicate that ghrelin may not be the only factor for H2-induced neuroprotection and other factors can substitute the role of ghrelin when ghrelin is absent, raising intriguing options of research for H2-responsive factors.


Subject(s)
Brain/metabolism , Deuterium/administration & dosage , Gastric Mucosa/metabolism , Ghrelin/deficiency , Parkinsonian Disorders/metabolism , Animals , Brain/drug effects , Brain/pathology , Ghrelin/antagonists & inhibitors , Ghrelin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Stomach/drug effects , Vagotomy/methods , Vagus Nerve/metabolism , Vagus Nerve/surgery
17.
Pathol Int ; 67(11): 564-574, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29027306

ABSTRACT

Oxidative stress including iron excess has been associated with carcinogenesis. The level of 8-oxoguanine, a major oxidatively modified base in DNA, is maintained very low by three distinct enzymes, encoded by OGG1, MUTYH and MTH1. Germline biallelic inactivation of MUTYH represents a familial cancer syndrome called MUTYH-associated polyposis. Here, we used Mutyh-deficient mice to evaluate renal carcinogenesis induced by ferric nitrilotriacetate (Fe-NTA). Although the C57BL/6 background is cancer-resistant, a repeated intraperitoneal administration of Fe-NTA induced a high incidence of renal cell carcinoma (RCC; 26.7%) in Mutyh-deficient mice in comparison to wild-type mice (7.1%). Fe-NTA treatment also induced renal malignant lymphoma, which did not occur without the Fe-NTA treatment in both the genotypes. Renal tumor-free survival after Fe-NTA treatment was marginally different (P = 0.157) between the two genotypes. Array-based comparative genome hybridization analyses revealed, in RCC, the loss of heterozygosity in chromosomes 4 and 12 without p16INKA inactivation; these results were confirmed by a methylation analysis and showed no significant difference between the genotypes. Lymphomas showed a preference for genomic amplifications. Dlk1 inactivation by promoter methylation may be involved in carcinogenesis in both tumors. Fe-NTA-induced murine RCCs revealed significantly less genomic aberrations than those in rats, demonstrating a marked species difference.


Subject(s)
Chromosome Aberrations/chemically induced , DNA Glycosylases/deficiency , Ferric Compounds/toxicity , Kidney Neoplasms/chemically induced , Kidney Neoplasms/genetics , Nitrilotriacetic Acid/analogs & derivatives , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrilotriacetic Acid/toxicity , Oxidative Stress/physiology , Rats , Species Specificity
18.
Proc Natl Acad Sci U S A ; 111(25): 9217-22, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24927551

ABSTRACT

Somatic hypermutation (SHM) of antibody variable region genes is initiated in germinal center B cells during an immune response by activation-induced cytidine deaminase (AID), which converts cytosines to uracils. During accurate repair in nonmutating cells, uracil is excised by uracil DNA glycosylase (UNG), leaving abasic sites that are incised by AP endonuclease (APE) to create single-strand breaks, and the correct nucleotide is reinserted by DNA polymerase ß. During SHM, for unknown reasons, repair is error prone. There are two APE homologs in mammals and, surprisingly, APE1, in contrast to its high expression in both resting and in vitro-activated splenic B cells, is expressed at very low levels in mouse germinal center B cells where SHM occurs, and APE1 haploinsufficiency has very little effect on SHM. In contrast, the less efficient homolog, APE2, is highly expressed and contributes not only to the frequency of mutations, but also to the generation of mutations at A:T base pair (bp), insertions, and deletions. In the absence of both UNG and APE2, mutations at A:T bp are dramatically reduced. Single-strand breaks generated by APE2 could provide entry points for exonuclease recruited by the mismatch repair proteins Msh2-Msh6, and the known association of APE2 with proliferating cell nuclear antigen could recruit translesion polymerases to create mutations at AID-induced lesions and also at A:T bp. Our data provide new insight into error-prone repair of AID-induced lesions, which we propose is facilitated by down-regulation of APE1 and up-regulation of APE2 expression in germinal center B cells.


Subject(s)
B-Lymphocytes/metabolism , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Endonucleases/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Germinal Center/metabolism , Mutation , Somatic Hypermutation, Immunoglobulin/physiology , Animals , B-Lymphocytes/cytology , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Germinal Center/cytology , Mice , Mice, Knockout , Multifunctional Enzymes , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism
19.
J Immunol ; 193(2): 931-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24935922

ABSTRACT

Activation-induced cytidine deaminase (AID) initiates a process generating DNA mutations and breaks in germinal center (GC) B cells that are necessary for somatic hypermutation and class-switch recombination. GC B cells can "tolerate" DNA damage while rapidly proliferating because of partial suppression of the DNA damage response by BCL6. In this study, we develop a model to study the response of mouse GC B cells to endogenous DNA damage. We show that the base excision repair protein apurinic/apyrimidinic endonuclease (APE) 2 protects activated B cells from oxidative damage in vitro. APE2-deficient mice have smaller GCs and reduced Ab responses compared with wild-type mice. DNA double-strand breaks are increased in the rapidly dividing GC centroblasts of APE2-deficient mice, which activate a p53-independent cell cycle checkpoint and a p53-dependent apoptotic response. Proliferative and/or oxidative damage and AID-dependent damage are additive stresses that correlate inversely with GC size in wild-type, AID-, and APE2-deficient mice. Excessive double-strand breaks lead to decreased expression of BCL6, which would enable DNA repair pathways but limit GC cell numbers. These results describe a nonredundant role for APE2 in the protection of GC cells from AID-independent damage, and although GC cells uniquely tolerate DNA damage, we find that the DNA damage response can still regulate GC size through pathways that involve p53 and BCL6.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/immunology , DNA Damage , Endonucleases/immunology , Germinal Center/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/metabolism , Cell Cycle/genetics , Cell Cycle/immunology , Cell Proliferation , Cells, Cultured , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , DNA Breaks, Double-Stranded , DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Endonucleases/deficiency , Endonucleases/genetics , Flow Cytometry , Germinal Center/metabolism , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Multifunctional Enzymes , Oxidative Stress/immunology , Proto-Oncogene Proteins c-bcl-6 , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
20.
Neuropathol Appl Neurobiol ; 41(2): 227-44, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24707896

ABSTRACT

AIMS: Galectin-1, a member of the ß-galactoside-binding lectin family, accumulates in neurofilamentous lesions in the spinal cords of both sporadic and familial amyotrophic lateral sclerosis (ALS) patients with a superoxide dismutase 1 gene (SOD1) mutation (A4V). The aim of this study was to evaluate the roles of endogenous galectin-1 in the pathogenesis of ALS. METHODS: Expression of galectin-1 in the spinal cord of mutant SOD1 transgenic (SOD1(G93A) ) mice was examined by pathological analysis, real-time RT-PCR and Western blotting. The effects of galectin-1 deficiency were evaluated by cross-breeding SOD1(G93A) mice with galectin-1 null (Lgals1(-/-) ) mice. RESULTS: Before ALS-like symptoms developed in SOD1(G93A) /Lgals1(+/+) mice, strong galectin-1 immunoreactivity was observed in swollen motor axons and colocalized with aggregated neurofilaments. Electron microscopic observations revealed that the diameters of swollen motor axons in the spinal cord were significantly smaller in SOD1(G93A) /Lgals1(-/-) mice, and there was less accumulation of vacuoles compared with SOD1(G93A) /Lgals1(+/+) mice. In symptomatic SOD1(G93A) /Lgals1(+/+) mice, astrocytes surrounding motor axons expressed a high level of galectin-1. CONCLUSIONS: Galectin-1 accumulates in neurofilamentous lesions in SOD1(G93A) mice, as previously reported in humans with ALS. Galectin-1 accumulation in motor axons occurs before the development of ALS-like symptoms and is associated with early processes of axonal degeneration in SOD1(G93A) mice. In contrast, galectin-1 expressed in astrocytes may be involved in axonal degeneration during symptom presentation.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Axons/pathology , Galectin 1/deficiency , Nerve Degeneration/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Axons/metabolism , Blotting, Western , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Immunoelectron , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration/pathology , Real-Time Polymerase Chain Reaction , Superoxide Dismutase/genetics , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL