ACTA HISTOCHEMICA ET CYTOCHEMICA
Online ISSN : 1347-5800
Print ISSN : 0044-5991
ISSN-L : 0044-5991
Current issue
Displaying 1-4 of 4 articles from this issue
REVIEW
  • Seiya Yamada, Tomoya Mizukoshi, Ayaka Sato, Shin-ichi Sakakibara
    Article type: Review
    2024 Volume 57 Issue 3 Pages 89-100
    Published: June 28, 2024
    Released on J-STAGE: June 28, 2024
    Advance online publication: June 22, 2024
    JOURNAL OPEN ACCESS FULL-TEXT HTML

    Neural stem/progenitor cells (NSPCs) in specific brain regions require precisely regulated metabolite production during critical development periods. Purines—vital components of DNA, RNA, and energy carriers like ATP and GTP—are crucial metabolites in brain development. Purine levels are tightly controlled through two pathways: de novo synthesis and salvage synthesis. Enzymes driving de novo pathway are assembled into a large multienzyme complex termed the “purinosome.” Here, we review purine metabolism and purinosomes as spatiotemporal regulators of neural development. Notably, around postnatal day 0 (P0) during mouse cortical development, purine synthesis transitions from the de novo pathway to the salvage pathway. Inhibiting the de novo pathway affects mTORC1 pathway and leads to specific forebrain malformations. In this review, we also explore the importance of protein-protein interactions of a newly identified NSPC protein—NACHT and WD repeat domain-containing 1 (Nwd1)—in purinosome formation. Reduced Nwd1 expression disrupts purinosome formation, impacting NSPC proliferation and neuronal migration, resulting in periventricular heterotopia. Nwd1 interacts directly with phosphoribosylaminoimidazole–succinocarboxamide synthetase (PAICS), an enzyme involved in de novo purine synthesis. We anticipate this review will be valuable for researchers investigating neural development, purine metabolism, and protein-protein interactions.

    Editor's pick

REGULAR ARTICLE
  • Shogo Takahata, Tomoko Kato, Daijiro Yanagisawa, Haruka Tsubaki, Zulzi ...
    Article type: Regular Article
    2024 Volume 57 Issue 3 Pages 101-108
    Published: June 28, 2024
    Released on J-STAGE: June 28, 2024
    Advance online publication: May 10, 2024
    JOURNAL OPEN ACCESS FULL-TEXT HTML

    Mitochondrial ferritin (FtMt) is a novel ferritin that sequesters iron and plays a protective role against oxidative stress. FtMt shares a high homology with H-ferritin but is expressed only in the brain, heart, and testis. In the midbrain, FtMt expression is observed in the substantia nigra. FtMt plays a neuroprotective role in the pathology of neurodegenerative diseases such as Parkinson’s disease, where excessive iron induces oxidative stress, causing cell death. Herein, we investigated FtMt immunoreactivity in the brains of patients with subarachnoid hemorrhage (SAH). Double immunofluorescence labeling of tyrosine hydroxylase (TH) and FtMt showed high colocalization in the substantia nigra pars compacta (SNc) in control and SAH cases. However, in SAH cases, FtMt immunoreactivity was observed in some TH-negative neurons. Double immunofluorescence labeling of glial cell markers and FtMt showed no apparent colocalization. The number and ratio of FtMt-positive but TH-negative neurons significantly differed between the control and SAH groups. Prussian blue staining in SAH cases showed positive iron staining over a wide surface range and the substantia nigra. Thus, FtMt may be related to iron dynamics in the substantia nigra following subarachnoid hemorrhage.

    Editor's pick

  • Yanan Wei, Si Cheng, Takehiro Tsukada, Kotaro Horiguchi, Yoko Fujiwara ...
    Article type: Regular Article
    2024 Volume 57 Issue 3 Pages 109-118
    Published: June 28, 2024
    Released on J-STAGE: June 28, 2024
    Advance online publication: June 15, 2024
    JOURNAL OPEN ACCESS FULL-TEXT HTML

    Retinoic acid (RA) plays a critical role in cell growth and tissue development. RA is synthesized from retinoids through oxidation processes by the retinaldehyde dehydrogenase (Raldh) family. However, the expression of Raldhs during pituitary development and the identification of Raldh-expressing cells in the adult pituitary have not been fully elucidated. Here, we performed in situ hybridization to localize the three Raldh isoforms (Raldh1-3) in fetal and adult mouse pituitary glands. The results showed that Raldh2 expression was observed in Rathke’s pouch from embryonic day 13.5 (E13.5), and this expression was sustained in the anterior lobe of the pituitary primordium from E15.5 to E17.5. In contrast, Raldh1 and Raldh3 were rarely detectable. Real-time PCR analysis revealed that Raldh2 was the predominant isoform expressed in the adult pituitary, although Raldh1 was also expressed to a lesser extent. In the adult pituitary, Raldh1-expressing cells were primarily observed in the posterior lobe. Raldh2-expressing cells were found in the marginal cell layer and parenchyma of the anterior lobe and were immunopositive for aldolase C (folliculostellate cells), but not for anterior pituitary hormones. These results suggest that RA is an important regulatory factor in the functions of the pituitary throughout its development in mice.

    Editor's pick

  • Yumiko Toyama, Takashi Nirasawa, Maho Morishima, Yuko Saito, Kazuhiro ...
    Article type: Regular Article
    2024 Volume 57 Issue 3 Pages 119-130
    Published: June 28, 2024
    Released on J-STAGE: June 28, 2024
    JOURNAL OPEN ACCESS FULL-TEXT HTML
    Supplementary material

    Pathological hallmark of Alzheimer’s disease (AD) is characterized by the accumulation and aggregation of amyloid β (Aβ) peptides into extracellular plaques of the brain. Clarification of the process of how soluble Aβ starts to assemble into amyloid fibrils is an essential step in elucidating the pathogenesis of AD. In our previous study, Aβ proteoforms including full-length Aβ40 and Aβ42/43 with N- and C-terminal truncated forms were visualized in postmortem brains from AD patients with matrix-assisted laser desorption/ionization-based mass spectrometry imaging (MALDI-MSI). In this study, Aβ proteoforms were consistently visualized by an updated protocol, and uncharacterized peptides such as Aβ1-29 and Aβ10-40 in AD brains were also visualized. To decipher neurotoxic effects of Aβ in patients’ brains, here we integrate liquid chromatography tandem mass spectrometry (LC-MS/MS) based shotgun proteomics with laser microdissection (LMD) excised tissue samples as well as direct tissue imaging with MALDI-MSI. With this approach, we have highlighted dynamic alterations of microtubule associating proteins (MAPs) including MAP1A, MAP1B and MAP2 as well as AD dominant proteins including APP, UCHL1, SNCA, and APOE. Of note, as lipid dysregulation has been implicated with AD pathology, we have challenged to integrate proteomics and lipid imaging for AD and control brain tissue. Spatial multi-omics is also valid to uncover molecular pathology of white matter as well as grey matter and leptomeningeal area, for example, by visualizing heme in patients’ postmortem brains.

    Editor's pick

feedback
Top