23 research outputs found

    DNA methylation dynamics in mouse preimplantation embryos revealed by mass spectrometry

    Get PDF
    Following fertilization in mammals, paternal genomic 5-methyl-2′-deoxycytidine (5 mC) content is thought to decrease via oxidation to 5-hydroxymethyl-2′-deoxycytidine (5 hmC). This reciprocal model of demethylation and hydroxymethylation is inferred from indirect, non-quantitative methods. We here report direct quantification of genomic 5 mC and 5 hmC in mouse embryos by small scale liquid chromatographic tandem mass spectrometry (SMM). Profiles of absolute 5 mC levels in embryos produced by in vitro fertilization (IVF) and intracytoplasmic sperm injection (ICSI) were almost identical. By 10 h after fertilization, 5 mC levels had declined by ∼40%, consistent with active genomic DNA demethylation. Levels of 5 mC in androgenotes (containing only a paternal genome) and parthenogenotes (containing only a maternal genome) underwent active 5 mC loss in the first 6 h, showing that both parental genomes can undergo demethylation independently. We found no evidence for net loss of 5 mC 10-48 h after fertilization, implying that any passive â€'demethylation' following DNA replication was balanced by active 5 mC maintenance methylation. However, levels of 5 mC declined during development after 48 h, to 1% (measured as a fraction of G-residues) in blastocysts (∼96 h). 5 hmC levels were consistently low (<0. 2% of G-residues) throughout development in normal diploid embryos. This work directly quantifies the dynamics of global genomic DNA modification in mouse preimplantation embryos, suggesting that SMM will be applicable to other biomedical situations with limiting sample sizes

    Diabetes Mellitus Accelerates Aβ Pathology in Brain Accompanied by Enhanced GAβ Generation in Nonhuman Primates

    No full text
    <div><p>Growing evidence suggests that diabetes mellitus (DM) is one of the strongest risk factors for developing Alzheimer’s disease (AD). However, it remains unclear why DM accelerates AD pathology. In cynomolgus monkeys older than 25 years, senile plaques (SPs) are spontaneously and consistently observed in their brains, and neurofibrillary tangles are present at 32 years of age and older. In laboratory-housed monkeys, obesity is occasionally observed and frequently leads to development of type 2 DM. In the present study, we performed histopathological and biochemical analyses of brain tissue in cynomolgus monkeys with type 2 DM to clarify the relationship between DM and AD pathology. Here, we provide the evidence that DM accelerates Aβ pathology <i>in vivo</i> in nonhuman primates who had not undergone any genetic manipulation. In DM-affected monkey brains, SPs were observed in frontal and temporal lobe cortices, even in monkeys younger than 20 years. Biochemical analyses of brain revealed that the amount of GM1-ganglioside-bound Aβ (GAβ)—the endogenous seed for Aβ fibril formation in the brain—was clearly elevated in DM-affected monkeys. Furthermore, the level of Rab GTPases was also significantly increased in the brains of adult monkeys with DM, almost to the same levels as in aged monkeys. Intraneuronal accumulation of enlarged endosomes was also observed in DM-affected monkeys, suggesting that exacerbated endocytic disturbance may underlie the acceleration of Aβ pathology due to DM.</p></div

    KUS121, an ATP regulator, mitigates chorioretinal pathologies in animal models of age-related macular degeneration

    Get PDF
    Age-related macular degeneration (AMD) is a leading cause of blindness among elderly people. The appearance of drusen is a clinical manifestation and a harbinger of both exudative and atrophic AMD. Recently, antibody-based medicines have been used to treat the exudative type. However, they do not restore good vision in patients. Moreover, no effective treatment is available for atrophic AMD. We have created small chemicals (Kyoto University Substances; KUSs) that act as ATP regulators inside cells. In the present study, we examined the in vivo efficacy of KUS121 in C-C chemokine receptor type 2-deficient mice, a mouse model of AMD. Systemic administration of KUS121 prevented or reduced drusen-like lesions and endoplasmic reticulum stress, and then substantially mitigated chorioretinal pathologies with significant preservation of visual function. Additionally, we confirmed that long-term oral administration of KUS121 caused no systemic complications in drusen-affected monkeys. ATP regulation by KUSs may represent a novel strategy in the treatment of drusen and prevention of disease progression in AMD

    Cerebral amyloid angiopathy in the brains of aged monkeys with DM.

    No full text
    <p>Images of temporal lobe (TL) sections from normal cynomolgus monkeys (A, B) and cynomolgus monkeys with DM (C, D). Sections were immunostained with anti-Aβ antibody and counterstained with hematoxylin. In the brains of DM-affected aged monkeys, we observed very severe CAA lesions (arrowheads) as compared to normal aged monkeys. CT, normal aged monkeys. DM, DM-affected aged monkeys. Scale bars, 100 μm.</p

    Histopathology of adult monkeys with type 2 diabetes mellitus.

    No full text
    <p>(A) Hematoxylin-eosin (HE)-stained section of the pancreas from a 17-year-old cynomolgus monkey with type 2 diabetes mellitus (DM) (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0117362#pone.0117362.s002" target="_blank">S1 Table</a>, Number 1). Most of the islets were replaced by abundant amyloid deposits. (B) HE-stained section of the pancreas from an 18-year-old cynomolgus monkey with DM (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0117362#pone.0117362.s002" target="_blank">S1 Table</a>, Number 3). Most of the islets were replaced with severe amyloid deposits. (C) HE-stained section of the pancreas from a 19-year-old cynomolgus monkey with DM (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0117362#pone.0117362.s002" target="_blank">S1 Table</a>, Number 7) showing hyalinized islets. Very few islet cells remain. (D) Direct fast scarlet-stained section of pancreas from an 18-year-old cynomolgus monkey with DM (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0117362#pone.0117362.s002" target="_blank">S1 Table</a>, Number 5). Hyalinized islets with severe amyloid deposition were positive for direct fast scarlet staining. (E) HE-stained section of the liver from an 18-year-old cynomolgus monkey with DM (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0117362#pone.0117362.s002" target="_blank">S1 Table</a>, Number 3). Marked fatty degeneration was observed in the liver. (F) HE-stained section of the aorta from an 18-year-old cynomolgus monkey with DM (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0117362#pone.0117362.s002" target="_blank">S1 Table</a>, Number 5). Mild atheromatosis with foam cells and sterol clefts was observed in the aorta. Scale bars for a-f, 100 μm.</p

    Senile plaques in the brains of adult monkeys with DM.

    No full text
    <p>Images of temporal lobe (TL) and frontal lobe (FL) sections from normal cynomolgus monkeys (A-D) and cynomolgus monkeys with DM (E-H). Sections were immunostained with anti-Aβ antibody and counterstained with hematoxylin. In aged monkey brains, we observed SPs immunostained with anti-Aβ antibody, as previously reported (A). In contrast, we did not observe Ab-immunopositive structures in the normal adult monkey brains (B-D). However, we did observe small but obvious Aβ-immunopositive senile plaques (SPs) in the frontal and temporal cortices of DM-affected adult monkeys (E-H). Scale bars, 100 μm. (I) Quantitative image analysis of Ab-immunopositive area in the sections obtained from female normal adult monkey, DM-affected adult monkey, and normal aged monkey brains. Data obtained from normal aged monkey brains were set as standards. Y-axes show the mean values of the quantified data. CT, normal cynomolgus monkeys. DM, DM-affected monkeys.</p

    Western blot analyses of axonal motor proteins, cathepsin D heavy chain, autophagosome marker LC3, and neprilysin in the brains of normal and DM-affected adult monkeys.

    No full text
    <p>Western blots showing the amounts of axonal motor proteins, cathepsin D heavy chain (CatD HC), autophagosome marker LC3, and neprilysin (NEP) in the brains of normal and DM-affected adult monkeys. Western blot analyses showed that the level of axonal motor proteins such as dynein heavy chain (DHC), dynein intermediate chain (DIC), kinesin heavy chain (KHC), and kinesin light chain (KLC) unchanged. The level of CatD HC increased in DM-affected monkey brains, and LC3-II showed significant increase in DM-affected adult monkeys. We did not observed DM-related changes in LC3-I and neprilysin (NEP) level. CT, normal adult monkeys; DM, DM-affected adult monkeys.</p

    Hypothetical schema of DM-induced Aβ pathology leading to AD onset.

    No full text
    <p>From the results of this study, we propose that DM induces GAβ generation by exacerbating age-dependent endocytic disturbance, resulting in enhanced Aβ pathology in the brain. Although additional studies are needed to clarify the whole mechanisms underlying DM-associated pathology, we hypothesize that, at the very least, enhanced Aβ pathology accompanied by endocytic disturbance might be involved in the development of AD.</p

    The analyses of Aβ and GAβ in the brains of normal and DM-affected monkeys.

    No full text
    <p>(A) Aβ level in young monkey, normal adult monkey, DM-affected monkey, and normal aged monkey brains were assessed with sandwich ELISA. Aβ level was significantly increased in normal aged monkey brains. In DM-affected monkey brains, Aβ level seemed unchanged. Data obtained from young monkey brains were set as standards. Y-axes show the mean values of the quantified data. Values are means ± SD. *P < 0.02. (B) Image of temporal lobe sections from a 19-year-old normal adult monkey and an 18-year-old cynomolgus monkey with DM. Sections were immunostained with the anti-GAβ-specific antibody 4396C and counterstained with hematoxylin. In the brain of the normal adult monkey, we observed little, if any, immunoreactivity for anti-GAβ antibody. By contrast, in the brain of the DM-affected adult monkey, we observed several neurons immunopositive for anti-GAβ antibody. Scale bars, 100 μm. (C) Quantitative image analysis of GAb-immunopositive area in the sections obtained from female normal adult monkey and DM-affected adult monkey brains. Data obtained from normal adult monkey brains were set as standards. Y-axes show the mean values of the quantified data. Values are means ± SD. *P < 0.02. (D) Dot blots showing the amount of GAβ generated in brains of cynomolgus monkeys of different ages. The blot samples were adjusted to 1 μg, 2.5 μg, or 5 μg of total protein. Dot blot analyses showed that GAβ generation increased in an age-dependent manner. (E) Dot blots showing the amount of GAβ generated in the brains of normal adult monkeys and DM-affected adult monkeys. The blot samples were adjusted to 1 μg, 2.5 μg, or 5 μg of total protein. The amount of GAβ in brains samples from DM-affected monkeys was significantly increased compared to those from normal adult monkeys. CT, normal adult monkeys; DM, DM-affected adult monkeys.</p
    corecore