Biological and Pharmaceutical Bulletin
Online ISSN : 1347-5215
Print ISSN : 0918-6158
ISSN-L : 0918-6158
Volume 44, Issue 7
Displaying 1-18 of 18 articles from this issue
Review
  • Tadashi Matsuda, Kenji Oritani
    2021 Volume 44 Issue 7 Pages 895-901
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Signal-transducing adaptor protein (STAP)-2 is an adaptor molecule involved in regulation of several intracellular signaling events in immune cells. STAP-2 contains a pleckstrin homology domain at the N-terminus, an src homology domain in the central portion and a proline-rich region at the C-terminus. STAP-2 also has a YXXQ motif, which is a potential signal transducer and activator of transcription (STAT)3-binding site. STAP-2 influences the STAT3 and STAT5 activity, integrin-mediated T cell adhesion, chemokine-induced T cell migration, Fas-mediated T cell apoptosis, Toll-like receptor-mediated macrophage functions, macrophage colony-stimulating factor-induced macrophage activation, and the high-affinity immunoglobulin E receptor-mediated mast cell activation. This article reviews the current understanding of roles of the STAP-2 during immune and/or inflammatory responses, and discusses possible therapeutic applications of targeting STAP-2 proteins in immune-related disorders.

Regular Articles
  • Xu-dong Ding, Yan-yan Cao, Lu Li, Guang-yi Zhao
    2021 Volume 44 Issue 7 Pages 902-909
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Local anesthetic toxicity is closely related to neuronal death and activation of the inflammatory response. Dexmedetomidine (Dex) is an adrenergic α2 receptor agonist that can reduce the neurotoxicity induced by lidocaine. It also has anti-inflammatory effects. However, the mechanism underlying the neuroprotective effects of Dex against lidocaine-induced toxicity remains to be defined. We hypothesized that Dex exerts its neural protective effect through inhibiting inflammasome activation and through anti-pyroptosis effects against local anesthetic-induced nerve injury. In a rat model of lidocaine-induced spinal cord injury, we studied the protective effect of Dex on lidocaine-induced changes in spinal cord function, inflammasome formation and pyroptosis, pro-inflammatory cytokine expression, and protein kinase C (PKC)-δ phosphorylation. Dex reduced lidocaine-induced neurotoxicity and inhibited PKC-δ phosphorylation in the spinal cord of rats. Furthermore, Dex inhibited pyroptosis and inflammasome formation (caspase-1, NLRP3, and apoptosis-associated speck-like protein (ASC)). Finally, Dex attenuated interleukin (IL)-1β and IL-18 expression, as well as microglia response. In conclusion, Dex can reduce the severity of lidocaine-induced spinal cord injury in rats by inhibiting priming and inflammasome activation and reducing pyroptosis via PKC-δ phosphorylation.

  • Masato Maruyama, Yousuke Nakano, Takuya Nishimura, Ryoichi Iwata, Sato ...
    2021 Volume 44 Issue 7 Pages 910-919
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    Advance online publication: April 23, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML
    Supplementary material

    Glioblastoma multiforme (GBM) is the most prevalent malignant primary brain tumor with a high recurrence rate. Despite multimodal therapy including surgical resection, chemotherapy, and radiotherapy, the median survival time after the initial diagnosis of GBM is approximately 14 months. Since cancer stem cells (CSCs) are considered the leading cause of cancer recurrence, glioblastoma stem cell-targeted therapy is a promising strategy for the treatment of GBM. However, because CSC heterogeneity has been implicated in the difficulties of CSC-target therapy, more in-depth knowledge of CSC biology is still required to develop novel therapies. In this study, we established single cell-derived tumorspheres from human glioblastoma U87MG cells. One of these tumorspheres, P4E8 clone, showed CSC-like phenotypes, such as self-renewal capacity, expression of CSC markers, resistance to anti-cancer agents, and in vivo tumorigenicity. Therefore, we used P4E8 cells as a cell-based model of glioblastoma stem cells (GSCs). Gene expression analysis using microarray indicated that the most highly expressed genes in P4E8 cells compared to the parental U87MG were PC3-secreted microprotein (MSMP). Furthermore, MSMP was expressed in patient-derived GSCs and human glioma tissues at the protein level, implying that MSMP might contribute to glioma development and progression.

    Editor's pick

    The authors established single cell-derived tumorsphere clone, named P4E8, from human glioblastoma U87MG cells and indicated that P4E8 cells had the CSC-like phenotype such as self-renewal capacity, expression of CSC markers, resistance to several anti-cancer agents and high tumorigenicity in vivo.  Furthermore, DNA microarray analysis identified that PC3-secreted microprotein (MSMP) was the highest expressed gene in P4E8 cells compared to U87MG cells.  In addition, authors first indicated the expression of MSMP protein in patient-derived glioma stem cells (GSCs) and human glioma tissues.  These findings raised the possibility that MSMP may contribute to glioma development and/or progression.

  • Yasuhiro Nakamura, Momoko Yoshida, Kazunari Tanigawa, Ayako Harada, Fu ...
    2021 Volume 44 Issue 7 Pages 920-925
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML
    Supplementary material

    Type I platelet-activating factor-acetylhydrolase (PAF-AH) forms a complex consisting of two catalytic subunits (α1 and/or α2) with a regulatory subunit (β). Although this protein was discovered as an enzyme that degrades an acetyl ester linked at the sn-2 position of platelet-activating factor (PAF), its physiological function remains unknown. In this study, to examine whether knockout mice lacking the catalytic subunits of this enzyme showed a different phenotype from that of wild-type mice, we measured and compared the body weights of knockout mice and control mice. The body weights of knockout mice were significantly increased compared to those of the control mice during 6 to 20 weeks from birth. Food intake was also significantly increased in knockout mice compared with control mice during these periods. Since a decrease in testis weight was reported in the knockout mice, we expected a decrease in testosterone levels. We measured and compared the amounts of testosterone in the serum and testis of knockout and control mice using liquid chromatography-tandem mass spectrometry, and found that testosterone levels in both the serum and testis were significantly decreased in the knockout mice compared with the control mice. These results suggest that a deficiency of type I PAF-AH catalytic subunits causes an increase in body weight, in part, due to reduced testosterone levels in male mice.

  • Ya li Yang, Zhan wen Lin, Pei ting He, Hua Nie, Qian yin Yao, Sheng yu ...
    2021 Volume 44 Issue 7 Pages 926-931
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    Advance online publication: April 29, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Background Astragalus polysaccharide (APS) had shown great promise in anti-tumour activities in our previous studies. The present study was designed to investigate whether APS has synergistic effect with cisplatin on the growth-inhibitory of human nasopharyngeal carcinoma cell lines and the possible mechanism. Methods Here, nasopharyngeal carcinoma cell lines (CNE-1) were divided into CNE-1 group, Cisplatin treatment group (2 µg/mL Cisplatin), APS treatment group (200 µg/mL APS) and combination group (2 µg/mL Cisplatin and 200 µg/mL APS). The proliferation inhibition rate of CNE-1 cells was determined by Cell Counting Kit-8 (CCK-8) method after treatment with different concentrations of APS for 24, 48, and 72 h. Apoptosis rates and cell cycle retardation of cells were detected by flow cytometry. Cell migration and invasion was evaluated by transwell assay. Western blotting and quantitative (q)RT-PCR were performed to detect the expression of Bcl-2, Bax, caspase-3, matrix metalloproteinase-2 (MMP-2), p53 and matrix metalloproteinase-9 (MMP-9) proteins in CNE-1 cells. Results APS have an inhibition on the proliferation of CNE-1 cells with time and dose dependence manner. Both the APS and combination therapy could promote apoptosis of CNE-1 cells, with the count of cells increased in G0/G1 and S phase while decreased in G2/M phase, and inhibited the migration and invasion of CNE-1 cells. Moreover, co-administration of Cisplatin and APS was more efficacious for the antitumor effect than either agent alone, as evidenced by the significant decrease in MMP-9 level and increase in p53. Conclusion APS, in combination with cisplatin, had significantly synergistic growth-inhibitory effect on nasopharyngeal carcinoma cell lines, which may be related to cell cycle and migration induction.

  • Akio Negishi, Shinji Oshima, Norimitsu Horii, Mizue Mutoh, Naoko Inoue ...
    2021 Volume 44 Issue 7 Pages 932-936
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    Advance online publication: May 07, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    The “INTERACTIONS” section of package inserts aims to provide alert-type warnings in clinical practice; however, these also include many drug–drug interactions that occur rarely. Moreover, considering that drug–drug interaction alert systems were created based on package inserts, repeated alerts can lead to alert fatigue. Although investigations have been conducted to determine prescriptions that induce drug–drug interactions, no studies have focused explicitly on the adverse events induced by drug–drug interactions. We, therefore, sought to investigate the true occurrence of adverse events caused by drug pair contraindications for coadministration in routine clinical practice. Toward this, we created a list of drug combinations that were designated as “contraindications for coadministration” and extracted the cases of adverse drug events from the Japanese Adverse Drug Event Report database that occurred due to combined drug usage. We then calculated the reporters’ recognition rate of the drug–drug interactions. Out of the 2121 investigated drug pairs, drug–drug interactions were reported in 43 pairs, 23 of which included an injected drug and many included catecholamines. Warfarin potassium and miconazole (19 reports), azathioprine and febuxostat (11 reports), and warfarin potassium and iguratimod (six reports) were among the 20 most-commonly reported oral medication pairs that were contraindicated for coadministration, for which recognition rates of drug–drug interactions were high. Although these results indicate that only a few drug pair contraindications for coadministration were associated with adverse drug events (43 pairs out of 2121 pairs), it remains necessary to translate these findings into clinical practice.

  • Kodai Ishida, Tomohiro Yako, Miruto Tanaka, Wataru Otsu, Shinsuke Naka ...
    2021 Volume 44 Issue 7 Pages 937-946
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML
    Supplementary material

    The corneal epithelium is continuously exposed to oxygen, light, and environmental substances. Excessive exposure to those stresses is thought to be a risk factor for eye diseases. Photokeratitis is damage to the corneal epithelium resulting in a painful eye condition caused by unprotected exposure to UV rays, usually from sunlight, and is often found in people who spend a long time outdoors. In modern life, human eyes are exposed to artificial light from light-emitting diode (LED) displays of computers and smartphones, and it has been shown that short-wavelength (blue) LED light can damage eyes, especially photoreceptors. However, the effect of blue LED light on the cornea is less understood. In addition, it is important to develop new treatments for preserving human eyesight and eye health from light stress. Here, we used human corneal epithelial cells-transformed (HCE-T) cells as an in-vitro model to investigate the protective effect of NSP-116, an imidazolyl aniline derivative, against the oxidative stress induced by light in the corneal epithelium. Treatment with 10 µM NSP-116 significantly increased the cell viability and reduced the death ratio following UV or blue LED light exposure. Furthermore, NSP-116 treatment decreased light-induced reactive oxygen species production and preserved the mitochondrial membrane potential. Immunoblotting data showed that NSP-116 suppressed the stress response pathway. Finally, NSP-116 treatment prevented corneal epithelial apoptosis induced by blue LED light in an in-vivo mouse model. In conclusion, NSP-116 has a protective effect against oxidative stress and corneal cell death from both UV and blue LED light exposure.

  • Aliaa Fouad, Kenjiro Matsumoto, Kikuko Amagase, Hiroyuki Yasuda, Makot ...
    2021 Volume 44 Issue 7 Pages 947-957
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Transient receptor potential melastatin 8 (TRPM8) is a non-selective cation channel activated by mild cooling and chemical agents including menthol. Nonsteroidal anti-inflammatory drugs have antipyretic, analgesic effects, and they can cause stomach and small intestinal injury. The current study investigated the role of TRPM8 in the pathogenesis of indomethacin-induced small intestinal injury. In male TRPM8-deficient (TRPM8KO) and wild-type (WT) mice, intestinal injury was induced via the subcutaneous administration of indomethacin. In addition, the effect of WS-12, a specific TRPM8 agonist, was examined in TRPM8KO and WT mice with indomethacin-induced intestinal injury. TRPM8KO mice had a significantly higher intestinal ulcerogenic response to indomethacin than WT mice. The repeated administration of WS-12 significantly attenuated the severity of intestinal injury in WT mice. However, this response was abrogated in TRPM8KO mice. Furthermore, in TRPM8-enhanced green fluorescent protein (EGFP) transgenic mice, which express EGFP under the direction of TRPM8 promoter, the EGFP signals in the indomethacin-treated intestinal mucosa were upregulated. Further, the EGFP signals were commonly found in calcitonin gene-related peptide (CGRP)-positive sensory afferent neurons and partly colocalized with substance P (SP)-positive neurons in the small intestine. The intestinal CGRP-positive neurons were significantly upregulated after the administration of indomethacin in WT mice. Nevertheless, this response was abrogated in TRPM8KO mice. In contrast, indomethacin increased the expression of intestinal SP-positive neurons in not only WT mice but also TRPM8KO mice. Thus, TRPM8 has a protective effect against indomethacin-induced small intestinal injury. This response may be mediated by the upregulation of CGRP, rather than SP.

    Editor's pick

    Transient receptor potential melastatin 8 (TRPM8) is a non-selective cation channel activated by mild cooling temperature and chemical cooling agents, including menthol. The authors demonstrate the mucosal protective and anti-inflammatory effects of TRPM8 expressed in sensory afferent neurons via in-vivo studies using TRPM8-deficient mice and specific TRPM8 agonist, and also immunohistochemical studies using TRPM8-enhanced green fluorescent protein (EGFP) transgenic mice. The TRPM8-mediated protective and anti-inflammatory effects are accounted for by a protective neuropeptide calcitonin gene-related peptide released from sensory afferent neurons. These findings propose that TRPM8 is a potential target for the treatment of NSAID-induced enteropathy and inflammatory bowel diseases.

  • Hua Chai, Wenyong Peng, Zhongquan Zhu, Duojia Xu, Yuanliang Chen, Zhij ...
    2021 Volume 44 Issue 7 Pages 958-966
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML
    Supplementary material

    Propofol is a commonly used anesthetic drug in clinic. In recent years, a series of non-anesthetic effects of propofol have been discovered. Studies have shown that propofol has many effects on the intestine. Epidermal growth factor (EGF) is one of the most important growth factors that could regulate intestinal growth and development. In the current study, we studied the effect of protocol on the biological activity of EGF on intestinal tissue and cell models. Through flow cytometry, indirect immunofluorescence and Western-blot and other technologies, it was found that propofol reduced the activity of EGF on intestinal cells, which inhibited EGF-induced intestinal cell proliferation and changed the cell behavior of EGF. To further explore the potential mechanism by which propofol down-regulated epidermal growth factor receptor (EGFR)-induced signaling, we carried out a series of related experiments, and found that propofol may inhibit the proliferation of intestinal cells by inhibiting the EGFR-mediated intracellular signaling pathway. The current research will lay the theoretical and experimental basis for further study of the effect of propofol on the intestine.

  • Tomoki Nakayoshi, Koichi Kato, Eiji Kurimoto, Akifumi Oda
    2021 Volume 44 Issue 7 Pages 967-975
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Isomerized aspartic acid (Asp) residues have previously been identified in various aging tissues, and are suspected to contribute to age-related diseases. Asp-residue isomerization occurs nonenzymatically under physiological conditions, resulting in the formation of three types of isomerized Asp (i.e., L-isoAsp, D-Asp, and D-isoAsp) residues. Asp-residue isomerization often accelerates protein aggregation and insolubilization, making structural biology analyses difficult. Recently, Sakaue et al. reported the synthesis of a ribonuclease A (RNase A) in which Asp121 was artificially replaced with different isomerized Asp residues, and experimentally demonstrated that the enzymatic activities of these artificial mutants were completely lost. However, their structural features have not yet been elucidated. In the present study, the three-dimensional (3D) structures of these artificial-mutant RNases A were predicted using molecular dynamics (MD) simulations. The 3D structures of wild-type and artificial-mutant RNases A were converged by 3000-ns MD simulations. Our computational data show that the structures of the active site and the formation frequencies of the appropriate catalytic dyad structures in the artificial-mutant RNases A were quite different from wild-type RNase A. These computational findings may provide an explanation for the experimental data which show that artificial-mutant RNases A lack enzymatic activity. Herein, MD simulations have been used to evaluate the influences of isomerized Asp residues on the 3D structures of proteins.

  • Daping Fan, Shihuan Yu, Yue Yang, Siying Qu
    2021 Volume 44 Issue 7 Pages 976-983
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Pyoluteorin is a natural occurring antibiotic and its anti-tumor activity has rarely been reported. This study aims to investigate the anti-tumor effects of pyoluteorin on human non-small cell lung cancer (NSCLC) cells. The cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis was determined through caspase3 activity assay and immunoblotting. Autophagy was measured by transmission electron microscope (TEM) and immunostaining. The autophagy-related proteins were detected through immunoblotting. We found that pyoluteorin showed significant anti-tumor effects on human NSCLC cell lines H1299 (IC50 = 1.57 µM) and H2030 (IC50 = 1.94 µM). Moreover, pyoluteorin could induce apoptosis and autophagy as evidence by the upregulation of caspase3 activity, the accumulation of LC3 and expression of apoptosis or autophagy related proteins. In addition, pyoluteorin induced autophagy through c-Jun N-terminal kinase/B-cell lymphoma-2 (JNK/Bcl-2) signal pathway. Blocking JNK/Bcl-2 pathway significantly attenuated pyoluteorin-induced autophagy. Moreover, inhibition of autophagy by 3-methyladenine (3-MA) or Beclin1 knockout greatly promoted pyoluteorin-induced apoptosis and cell death. Our results showed that pyoluteorin could induce both apoptosis and autophagy in human NSCLC cells. Combination of pyoluteorin with autophagy inhibitior significantly promoted pyoluteorin-induced apoptosis and may be a potential anticancer strategy in the NSCLC therapy.

    Editor's pick

    Authors demonstrate that pyoluteorin can inhibit cell proliferation by inducing autophagy and apoptosis in NSCLC cell lines through the JNK/Bcl2 pathway. Inhibition of autophagy via 3-MA or Beclin1 knockout enhance pyoluteorin-induced apoptosis. Authors believe that pyoluteorin combined with autophagy inhibitor may be a potential anticancer drug for human NSCLC.

  • Keita Kitamura, Kenta Umehara, Ryo Ito, Yoshiyuki Yamaura, Takafumi Ko ...
    2021 Volume 44 Issue 7 Pages 984-991
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    Advance online publication: April 24, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML
    Supplementary material

    In vitro blood–brain barrier (BBB) models are essential research tools for use in developing brain-targeted drugs and understanding the physiological and pathophysiological functions of the BBB. To develop BBB models with better functionalities, three-dimensional (3D) culture methods have gained significant attention as a promising approach. In this study, we report on the development of a human conditionally immortalized cell-based multicellular spheroidal BBB (hiMCS-BBB) model. After being seeded into non-attachment culture wells, HASTR/ci35 (astrocytes) and HBPC/ci37 cells (brain pericytes) self-assemble to form a spheroid core that is then covered with an outer monolayer of HBMEC/ci18 cells (brain microvascular endothelial cells). The results of immunocytochemistry showed the protein expression of several cellular junction and BBB-enriched transporter genes in HBMEC/ci18 cells of the spheroid model. The permeability assays showed that the hiMCS-BBB model exhibited barrier functions against the penetration of dextran (5 and 70 kDa) and rhodamine123 (a P-glycoprotein substrate) into the core. On the other hand, facilitation of 2-(N-[7-nitrobenz-2-oxa-1,3-diazol-4-yl]amino)-2-deoxyglucose (2-NBDG; a fluorescent glucose analog) uptake was observed in the hiMCS-BBB model. Furthermore, tumor necrosis factor-alpha treatment elicited an inflammatory response in HBMEC/ci18 cells, thereby suggesting that BBB inflammation can be recapitulated in the hiMCS-BBB model. To summarize, we have developed an hiMCS-BBB model that possesses fundamental BBB properties, which can be expected to provide a useful and highly accessible experimental platform for accelerating various BBB studies.

    Editor's pick

    Kitamura et al. have developed a new human immortalized cell-based multicellular spheroidal blood-brain barrier (BBB) model, in which astrocytes and pericytes form a spheroid core that is covered with an outer monolayer of brain microvascular endothelial cells. This layered structure is likely to play a critical role in bringing out high levels of BBB characteristics in the model and allows researchers to examine various BBB functions, including drug permeability assays. Therefore, the work opens up new avenues for accelerating in vitro BBB modeling, which in turn significantly contributes to CNS drug development as well as elucidation of molecular bases of CNS diseases.

  • Daping Fan, Shihuan Yu, Yue Yang, Siying Qu
    2021 Volume 44 Issue 7 Pages 992-998
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    The RAS protein activator like 2 (Rasal2) has been reported to be a tumor suppressor in variety of cancers; while an oncogenic protein in ovarian cancer and triple negative breast cancer (TNBC). However, the exact role of Rasal2 in non-small cell lung cancer (NSCLC) is lacking. This study aimed to investigate the role of Rasal2 in NSCLC and the underlying mechanisms. Rasal2 expression level was measured in NSCLC tissue and cells by using quantitative (q)-PCR and immunoblotting analysis. The clinical implication of Rasal2 in NSCLC patients was also analyzed. The function role of Rasal2 in NSCLC cells were measured by small interfering RNA (si-RNA), immunostaining, transwell assay and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Low Rasal2 expression level was observed in human NSCLC tissue and cell lines and significantly related to tumor thickness, ulceration and TNM staging in NSCLC patients. Rasal2 knockdown significantly increased NSCLC cell invasion and migration. Mechanistically, we showed that Rasal2 knockdown significantly increased the phosphorylation level of extracellular signal-regulated kinase (ERK)/Raf1/mitogen-activated protein extracellular kinase (MEK) thus activated Ras/ERK signal pathway. Thus, our data showed that Rasal2 is downregulated in NSCLC cells and act as an epithelial–mesenchymal transition (EMT) and metastasis suppressor through the Ras/ERK pathway. Rasal2 may be a prognostic biomarker for NSCLC in the future.

  • Yanyu Wang, Yanping Zhao, Chaochun Wei, Nana Tian, Hong Yan
    2021 Volume 44 Issue 7 Pages 999-1006
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Flavonoids are potential strikingly natural compounds with antioxidant activity and acetylcholinesterase (AChE) inhibitory activity for treating Alzheimer’s disease (AD). In present study, in line with our interests in flavonoid derivatives as AChE inhibitors, a four-dimensional quantitative structure–activity relationship (4D-QSAR) molecular model was proposed. The data required to perform 4D-QSAR analysis includes 52 compounds reported in the literature, usually analogs, and their measured biological activities in a common assay. The model was generated by a complete set of 4D-QSAR program which was written by our group. The best model was found after trying multiple experiments. It had a good predictive ability with the cross-validation correlation coefficient Q2 = 0.77, the internal validation correlation coefficient R2 = 0.954, and the external validation correlation coefficient R2pred = 0.715. The molecular docking analysis was also carried out to understand exceedingly the interactions between flavonoids and the AChE targets, which was in good agreement with the 4D-QSAR model. Based on the information provided by the 4D-QSAR model and molecular docking analysis, the idea for optimizing the structures of flavonoids as AChE inhibitors was put forward which maybe provide theoretical guidance for the research and development of new AChE inhibitors.

  • Hirohito Esaki, Shoma Izumi, Akari Fukao, Shiho Ito, Naoya Nishitani, ...
    2021 Volume 44 Issue 7 Pages 1007-1013
    Published: July 01, 2021
    Released on J-STAGE: July 01, 2021
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Nicotine has been known to enhance recognition memory in various species. However, the brain region where nicotine acts and exerts its effect remains unclear. Since the medial prefrontal cortex (mPFC) is associated with memory, we examined the role of the mPFC in nicotine-induced enhancement of recognition memory using the novel object recognition test in male C57BL/6J mice. Systemic nicotine administration 10 min before training session significantly enhanced object recognition memory in test session that was performed 24 h after the training. Intra-mPFC infusion of mecamylamine, a non-selective nicotinic acetylcholine receptor (nAChR) antagonist, 5 min before nicotine administration blocked the effect of nicotine. Additionally, intra-mPFC infusion of dihydro-β-erythroidine, a selective α4β2 nAChR antagonist, or methyllycaconitine, a selective α7 nAChR antagonist, significantly suppressed the nicotine-induced object recognition memory enhancement. Finally, intra-mPFC infusion of nicotine 1 min before the training session augmented object recognition memory in a dose-dependent manner. These findings suggest that mPFC α4β2 and α7 nAChRs mediate the nicotine-induced object recognition memory enhancement.

    Editor's pick

    Nicotine is known to enhance recognition memory in various species. However, the brain region where nicotine acts and exerts its effect remains unclear. In this study, using the novel object recognition test in mice, authors provide evidence that nicotine acts on α4β2 and α7 nicotinic acetylcholine receptors expressed in the medial prefrontal cortex, a brain region associated with memory, and enhances object recognition memory.

Notes
feedback
Top