Updated on 2024/04/27

写真a

 
NAKAJIMA Mayuka
 
Organization
Academic Assembly Institute of Medicine and Dentistry SHIGAKU KEIRETU Assistant Professor
Faculty of Dentistry Department of Dentistry Assistant Professor
Graduate School of Medical and Dental Sciences Oral Life Science Oral Biological Science Assistant Professor
Title
Assistant Professor
External link

Degree

  • 博士(歯学) ( 2016.3   新潟大学 )

Research Interests

  • ドラッグデリバリー

  • 細菌学

  • 免疫学

  • 分子生物学

  • 歯周病学

Research Areas

  • Nanotechnology/Materials / Nanomaterials

  • Life Science / Conservative dentistry

Research History (researchmap)

  • 令和4年度 新潟大学 若手教員スイングバイ・プログラム 助教

    2022.4

      More details

  • 日本学術振興会 海外特別研究員

    2020.4 - 2022.3

      More details

  • 米国ハーバード大学ジョンA.ポールソン工学応用科学部 博士研究員

    2019.4 - 2022.3

      More details

  • Niigata University   Graduate School of Medical and Dental Sciences

    2018.4 - 2020.3

      More details

  • Niigata University   Medical and Dental Hospital

    2017.4 - 2018.3

      More details

Research History

  • Niigata University   School of Dentistry, Faculty of Dentistry   Assistant Professor

    2022.4

  • Niigata University   Oral Biological Science, Oral Life Science, Graduate School of Medical and Dental Sciences   Assistant Professor

    2022.4

  • Niigata University   Institute of Medicine and Dentistry, Academic Assembly   Assistant Professor

    2022.4

Education

  • Niigata University   Graduate School of Medical and Dental Sciences   歯周診断・再建学分野

    2012.4 - 2016.3

      More details

  • Niigata University   Faculty of Dentistry   School of Dentistry

    2005.4 - 2011.3

      More details

Professional Memberships

  • NIIGATA DENTAL SOCIETY

      More details

  • THE JAPANESE SOCIETY OF CONSERVATIVE DENTISTRY

      More details

  • JAPANESE ASSOCIATION FOR DENTAL RESEARCH

      More details

  • JAPANESE SOCIETY OF PERIODONTOLOGY

      More details

  • International Association for Dental Research

      More details

Studying abroad experiences

  • Harvard University SEAS   Postdoctoral Fellow

    2019.4 - 2022.3

 

Papers

  • Engineering of bioactive nanocomplexes on dental floss for targeted gingival therapy Reviewed

    Mayuka Nakajima, Nao Nakajima, Junling Guo, Samir Mitragotri

    Bioengineering & Translational Medicine   8 ( 2 )   2022.12

     More details

    Authorship:Lead author   Publishing type:Research paper (scientific journal)   Publisher:Wiley  

    Abstract

    Periodontitis induced by chronic subgingival infection is a ubiquitous disease that causes systemic inflammatory consequences and poses a negative impact on quality of life. The disease is treated and potentially prevented by patient's self‐care aimed at eliminating the oral pathogens from the region. Currently available products for interdental self‐care, including dental floss and interdental brush, have limited ability to prevent the disease. Here, we report a coated dental floss thread, termed “nanofloss,” which uses polyphenol‐based nanocoating to functionalize the floss thread with therapeutic agents. Multiple therapeutics can be integrated into the nanofloss including antibacterial small molecules and proteins. Flossing with nanofloss‐delivered therapeutic agents to the challenging subgingival region with long‐term retention even against the flushing action of the oral fluid in vivo. Our in vitro and in vivo studies demonstrate that chlorhexidine gluconate‐loaded nanofloss effectively treats the subgingival infection by Porphyromonas gingivalis. Collectively, the nanofloss offers a promising and easily usable tool for targeted self‐care of subgingival infection against periodontitis.

    DOI: 10.1002/btm2.10452

    researchmap

    Other Link: https://onlinelibrary.wiley.com/doi/full-xml/10.1002/btm2.10452

  • Extracellular matrix degrading enzyme with stroma-targeting peptides enhance the penetration of liposomes into tumors Reviewed

    Mayumi Ikeda-Imafuku, Yongsheng Gao, Suyog Shaha, Lily Li-Wen Wang, Kyung Soo Park, Mayuka Nakajima, Omokolade Adebowale, Samir Mitragotri

    Journal of Controlled Release   352   1093 - 1103   2022.12

     More details

    Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    DOI: 10.1016/j.jconrel.2022.11.007

    researchmap

  • Cellular Backpackによる歯周免疫療法の開発研究

    中島 麻由佳, 多部田 康一

    特定非営利活動法人日本歯科保存学会学術大会プログラムおよび講演抄録集   157回   57 - 57   2022.10

     More details

    Language:Japanese   Publisher:(NPO)日本歯科保存学会  

    researchmap

  • Backpack結合M2マクロファージの局所投与による歯周炎抑制効果の検証

    中島 麻由佳, 多部田 康一

    日本歯周病学会会誌   64 ( 秋季特別 )   125 - 125   2022.8

     More details

    Language:Japanese   Publisher:(NPO)日本歯周病学会  

    researchmap

  • イオンジェルを用いた経粘膜局所感染制御法の開発

    中島 麻由佳, 多部田 康一

    日本歯周病学会会誌   64 ( 春季特別 )   115 - 115   2022.5

     More details

    Language:Japanese   Publisher:(NPO)日本歯周病学会  

    researchmap

  • Oral Pathobiont-Induced Changes in Gut Microbiota Aggravate the Pathology of Nonalcoholic Fatty Liver Disease in Mice Reviewed

    Kyoko Yamazaki, Tamotsu Kato, Yuuri Tsuboi, Eiji Miyauchi, Wataru Suda, Keisuke Sato, Mayuka Nakajima, Mai Yokoji-Takeuchi, Miki Yamada-Hara, Takahiro Tsuzuno, Aoi Matsugishi, Naoki Takahashi, Koichi Tabeta, Nobuaki Miura, Shujiro Okuda, Jun Kikuchi, Hiroshi Ohno, Kazuhisa Yamazaki

    Frontiers in Immunology   12   2021.10

     More details

    Publishing type:Research paper (scientific journal)   Publisher:Frontiers Media {SA}  

    <jats:sec><jats:title>Background &amp;amp; Aims</jats:title><jats:p>Periodontitis increases the risk of nonalcoholic fatty liver disease (NAFLD); however, the underlying mechanisms are unclear. Here, we show that gut dysbiosis induced by oral administration of <jats:italic>Porphyromonas gingivalis</jats:italic>, a representative periodontopathic bacterium, is involved in the aggravation of NAFLD pathology.</jats:p></jats:sec><jats:sec><jats:title>Methods</jats:title><jats:p>C57BL/6N mice were administered either vehicle, <jats:italic>P. gingivalis</jats:italic>, or <jats:italic>Prevotella intermedia</jats:italic>, another periodontopathic bacterium with weaker periodontal pathogenicity, followed by feeding on a choline-deficient, l-amino acid-defined, high-fat diet with 60 kcal% fat and 0.1% methionine (CDAHFD60). The gut microbial communities were analyzed by pyrosequencing the 16S ribosomal RNA genes. Metagenomic analysis was used to determine the relative abundance of the Kyoto Encyclopedia of Genes and Genomes pathways encoded in the gut microbiota. Serum metabolites were analyzed using nuclear magnetic resonance-based metabolomics coupled with multivariate statistical analyses. Hepatic gene expression profiles were analyzed <jats:italic>via</jats:italic> DNA microarray and quantitative polymerase chain reaction.</jats:p></jats:sec><jats:sec><jats:title>Results</jats:title><jats:p>CDAHFD60 feeding induced hepatic steatosis, and in combination with bacterial administration, it further aggravated NAFLD pathology, thereby increasing fibrosis. Gene expression analysis of liver samples revealed that genes involved in NAFLD pathology were perturbed, and the two bacteria induced distinct expression profiles. This might be due to quantitative and qualitative differences in the influx of bacterial products in the gut because the serum endotoxin levels, compositions of the gut microbiota, and serum metabolite profiles induced by the ingested <jats:italic>P. intermedia</jats:italic> and <jats:italic>P. gingivalis</jats:italic> were different.</jats:p></jats:sec><jats:sec><jats:title>Conclusions</jats:title><jats:p>Swallowed periodontopathic bacteria aggravate NAFLD pathology, likely due to dysregulation of gene expression by inducing gut dysbiosis and subsequent influx of gut bacteria and/or bacterial products.</jats:p></jats:sec>

    DOI: 10.1021/acsomega.1c06891

    DOI: 10.1093/pcp/pcac114

    Scopus

    PubMed

    researchmap

  • Oral Pathobiont-Induced Changes in Gut Microbiota Aggravate the Pathology of Nonalcoholic Fatty Liver Disease in Mice Reviewed International journal

    Kyoko Yamazaki, Tamotsu Kato, Yuuri Tsuboi, Eiji Miyauchi, Wataru Suda, Keisuke Sato, Mayuka Nakajima, Mai Yokoji-Takeuchi, Miki Yamada-Hara, Takahiro Tsuzuno, Aoi Matsugishi, Naoki Takahashi, Koichi Tabeta, Nobuaki Miura, Shujiro Okuda, Jun Kikuchi, Hiroshi Ohno, Kazuhisa Yamazaki

    Frontiers in Immunology   12   766170 - 766170   2021.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Frontiers Media SA  

    Background &amp;amp; Aims

    Periodontitis increases the risk of nonalcoholic fatty liver disease (NAFLD); however, the underlying mechanisms are unclear. Here, we show that gut dysbiosis induced by oral administration of Porphyromonas gingivalis, a representative periodontopathic bacterium, is involved in the aggravation of NAFLD pathology.

    Methods

    C57BL/6N mice were administered either vehicle, P. gingivalis, or Prevotella intermedia, another periodontopathic bacterium with weaker periodontal pathogenicity, followed by feeding on a choline-deficient, l-amino acid-defined, high-fat diet with 60 kcal% fat and 0.1% methionine (CDAHFD60). The gut microbial communities were analyzed by pyrosequencing the 16S ribosomal RNA genes. Metagenomic analysis was used to determine the relative abundance of the Kyoto Encyclopedia of Genes and Genomes pathways encoded in the gut microbiota. Serum metabolites were analyzed using nuclear magnetic resonance-based metabolomics coupled with multivariate statistical analyses. Hepatic gene expression profiles were analyzed via DNA microarray and quantitative polymerase chain reaction.

    Results

    CDAHFD60 feeding induced hepatic steatosis, and in combination with bacterial administration, it further aggravated NAFLD pathology, thereby increasing fibrosis. Gene expression analysis of liver samples revealed that genes involved in NAFLD pathology were perturbed, and the two bacteria induced distinct expression profiles. This might be due to quantitative and qualitative differences in the influx of bacterial products in the gut because the serum endotoxin levels, compositions of the gut microbiota, and serum metabolite profiles induced by the ingested P. intermedia and P. gingivalis were different.

    Conclusions

    Swallowed periodontopathic bacteria aggravate NAFLD pathology, likely due to dysregulation of gene expression by inducing gut dysbiosis and subsequent influx of gut bacteria and/or bacterial products.

    DOI: 10.3389/fimmu.2021.766170

    PubMed

    researchmap

  • Topical treatment of periodontitis using an iongel Reviewed International journal

    Mayuka Nakajima, Eden E.L. Tanner, Nao Nakajima, Kelly N. Ibsen, Samir Mitragotri

    Biomaterials   276   121069 - 121069   2021.9

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    Almost 50 % of the U.S. population suffers from oral infections such as periodontitis. Current treatment options for periodontitis include mechanical cleaning procedures, which are performed often under local anesthesia and are time-consuming. Alternate option includes systemic antibiotics which increase the risk of antimicrobial resistance and are not recommended for prolonged usage. Topical treatments of gingiva-based antimicrobial agents have shown limited efficacy due to poor penetration of the therapeutic into deep gingiva where the infection resides. Herein, we report an Iongel of a Deep Eutectic Antimicrobial (IDEA), which simultaneously exhibits deep tissue penetration and antimicrobial activity against P. gingivalis. In vivo studies confirmed that IDEA successfully penetrated into the gingiva and the gingival sulcus, where the pathogens primarily exist, within a short time. In vitro studies confirmed that the dose delivered was adequate to inactivate P. gingivalis biofilm. In vivo studies in a periodontal rat model confirmed that a topical treatment of IDEA eliminated pathogenic bacteria, and the disease progression was significantly suppressed. Safety studies confirmed excellent tolerance to IDEA. Altogether, IDEA offers a promising topical agent against periodontitis.

    DOI: 10.1016/j.biomaterials.2021.121069

    PubMed

    researchmap

  • β2-Microglobulin and Neutrophil Gelatinase-Associated Lipocalin, Potential Novel Urine Biomarkers in Periodontitis: A Cross-Sectional Study in Japanese. Reviewed International journal

    Mayuka Nakajima, Michihiro Hosojima, Koichi Tabeta, Sayuri Miyauchi, Miki Yamada-Hara, Naoki Takahashi, Haruna Miyazawa, Yumi Matsuda-Matsukawa, Keisuke Sato, Noriko Sugita, Yasutaka Komatsu, Tomomi Ishikawa, Kazuhiro Akiishi, Kazuhisa Yamazaki, Kiminori Kato, Akihiko Saito, Hiromasa Yoshie

    International journal of dentistry   2019   1394678 - 1394678   2019

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)  

    Objectives: Several serum biomarkers have been reported to increase in periodontitis patients as possible mediators linking periodontal inflammation to systemic diseases. However, the relationship between periodontitis and urine biomarkers is still unclear. The aim of this cross-sectional study was to investigate potential urine biomarkers of periodontitis in a Japanese population. Materials and Methods: This study included 108 male subjects, and microbiological and clinical parameters were evaluated as a periodontitis marker. The correlation between nine urine biomarkers (typically used to diagnose kidney disease) and periodontal parameters was analyzed. Based on the findings, β2-microglobulin (β2-MG) and neutrophil gelatinase-associated lipocalin (NGAL) were selected for comparison and multivariate regression analysis, and the Kruskal-Wallis test followed by Bonferroni correction was used to identify differences in their concentrations between the three periodontitis groups (severe, moderate, and no/mild periodontitis). Results: β2-MG and NGAL exhibited a significant correlation with clinical parameters of periodontitis. The prevalence of clinical parameters such as bleeding on probing and number of sites with probing depth (PD) ≥ 6 mm were greater in the β2-MG high group (≥300 μg/g creatinine) than in the normal group (P=0.017 and 0.019, respectively). Multivariate regression analysis indicated that the number of sites with PD ≥ 6 mm was independently associated with urine β2-MG. Moreover, the number of sites with the clinical attachment level (CAL) ≥ 6 mm was greater in the NGAL high group (highest quartile) (P=0.041). Multivariate regression analysis showed that the number of sites with CAL ≥ 6 mm was associated independently with urine NGAL. Finally, β2-MG was significantly higher in the severe periodontitis subjects compared to the no/mild periodontitis subjects. Conclusion: The significant association between urine β2-MG or NGAL and periodontitis was revealed. These biomarkers can potentially be used to screen for or diagnose periodontitis. This trial is registered with the UMIN Clinical Trials Registry UMIN000013485.

    DOI: 10.1155/2019/1394678

    PubMed

    researchmap

  • Oral Administration of Porphyromonas gingivalis Alters the Gut Microbiome and Serum Metabolome. Reviewed International journal

    Tamotsu Kato, Kyoko Yamazaki, Mayuka Nakajima, Yasuhiro Date, Jun Kikuchi, Koji Hase, Hiroshi Ohno, Kazuhisa Yamazaki

    mSphere   3 ( 5 )   2018.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Periodontal disease induced by periodontopathic bacteria like Porphyromonas gingivalis is demonstrated to increase the risk of metabolic, inflammatory, and autoimmune disorders. Although precise mechanisms for this connection have not been elucidated, we have proposed mechanisms by which orally administered periodontopathic bacteria might induce changes in gut microbiota composition, barrier function, and immune system, resulting in an increased risk of diseases characterized by low-grade systemic inflammation. Accumulating evidence suggests a profound effect of altered gut metabolite profiles on overall host health. Therefore, it is possible that P. gingivalis can affect these metabolites. To test this, C57BL/6 mice were administered with P. gingivalis W83 orally twice a week for 5 weeks and compared with sham-inoculated mice. The gut microbial communities were analyzed by pyrosequencing the 16S rRNA genes. Inferred metagenomic analysis was used to determine the relative abundance of KEGG pathways encoded in the gut microbiota. Serum metabolites were analyzed using nuclear magnetic resonance (NMR)-based metabolomics coupled with multivariate statistical analyses. Oral administration of P. gingivalis induced a change in gut microbiota composition. The distributions of metabolic pathways differed between the two groups, including those related to amino acid metabolism and, in particular, the genes for phenylalanine, tyrosine, and tryptophan biosynthesis. Also, alanine, glutamine, histidine, tyrosine, and phenylalanine were significantly increased in the serum of P. gingivalis-administered mice. In addition to altering immune modulation and gut barrier function, oral administration of P. gingivalis affects the host's metabolic profile. This supports our hypothesis regarding a gut-mediated systemic pathology resulting from periodontal disease.IMPORTANCE Increasing evidence suggest that alterations of the gut microbiome underlie metabolic disease pathology by modulating gut metabolite profiles. We have shown that orally administered Porphyromonas gingivalis, a representative periodontopathic bacterium, alters the gut microbiome; that may be a novel mechanism by which periodontitis increases the risk of various diseases. Given the association between periodontal disease and metabolic diseases, it is possible that P. gingivalis can affect the metabolites. Metabolite profiling analysis demonstrated that several amino acids related to a risk of developing diabetes and obesity were elevated in P. gingivalis-administered mice. Our results revealed that the increased risk of various diseases by P. gingivalis might be mediated at least in part by alteration of metabolic profiles. The findings should add new insights into potential links between periodontal disease and systemic disease for investigators in periodontal disease and also for investigators in the field of other diseases, such as metabolic diseases.

    DOI: 10.1128/mSphere.00460-18

    PubMed

    researchmap

  • Increased serum PCSK9, a potential biomarker to screen for periodontitis, and decreased total bilirubin associated with probing depth in a Japanese community survey Reviewed

    K. Tabeta, M. Hosojima, M. Nakajima, S. Miyauchi, H. Miyazawa, N. Takahashi, Y. Matsuda, N. Sugita, Y. Komatsu, K. Sato, T. Ishikawa, K. Akiishi, K. Yamazaki, K. Kato, A. Saito, H. Yoshie

    Journal of Periodontal Research   53 ( 3 )   446 - 456   2018.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Blackwell Munksgaard  

    Background and Objectives: Previous reports suggest that several serum biomarkers play roles in the pathogenesis, inflammatory response, and oxidative stress in periodontitis caused by bacterial infections, linking chronic periodontitis to atherosclerotic vascular disease (ASVD). The aim of this preliminary study was to investigate, in a Japanese cross-sectional community survey, potential serum biomarkers of periodontitis that are associated with ASVD and chronic periodontitis. Material and Methods: The study cohort included a total of 108 male subjects who underwent annual health examinations. Serum biomarkers (high-sensitivity C-reactive protein [hs-CRP], proprotein convertase subtilisin/kexin type 9 [PCSK9], interleukin-6, tumor necrosis factor-α, soluble CD14, myeloperoxidase, matrix metalloproteinase-3, adiponectin, total bilirubin [TBIL], and serum lipids) were analyzed to determine their association (if any) with periodontal parameters. Aortic stiffness was evaluated using the brachial-ankle aortic pulse wave velocity (PWV) index and the cardio-ankle vascular index (CAVI). Results: The concentrations of PCSK9 and hs-CRP were increased (P =.001 and.042, respectively), and the concentration of TBIL was decreased (P =.046), in subjects with periodontal disease (determined as a probing depth of ≥4 mm in at least one site) compared with periodontally healthy subjects. The ratio of low-density lipoprotein cholesterol (LDL-C) to high-density lipoprotein cholesterol and the concentrations of triglycerides, remnant-like particles-cholesterol, and oxidized LDL were elevated in subjects with periodontal disease compared with periodontally healthy subjects (P =.038,.007,.002, and.049, respectively). Multivariate regression analyses indicated that the number of sites with a pocket depth of ≥4 mm was associated with the concentration of PCSK9 and inversely associated with the concentration of TBIL independently (standardized β =.243, P =.040
    standardized β = −.443, P =.0002
    respectively). Analysis of receiver operating characteristic curves of PCSK9 indicated moderate accuracy for predicting the presence of disease sites (probing depth ≥ 4 mm) (area under the curve = 0.740). No significance in the values of PWV and CAVI was observed between subjects with periodontal disease and periodontally healthy subjects. Conclusion: In Japanese male subjects, the concentrations of serum PCSK9 and TBIL were correlated with periodontal parameters. Moreover, PCSK9 could be a candidate biomarker for diagnosing chronic periodontitis, and may also have potential to evaluate the risk for periodontitis to cause ASVD. Longitudinal studies of larger populations are necessary to confirm the exact association of periodontitis with increased serum PCSK9 and decreased TBIL.

    DOI: 10.1111/jre.12533

    Scopus

    PubMed

    researchmap

  • Ligature-induced periodontitis in mice induces elevated levels of circulating interleukin-6 but shows only weak effects on adipose and liver tissues Reviewed

    Y. Matsuda, T. Kato, N. Takahashi, M. Nakajima, K. Arimatsu, T. Minagawa, K. Sato, H. Ohno, K. Yamazaki

    JOURNAL OF PERIODONTAL RESEARCH   51 ( 5 )   639 - 646   2016.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Background and Objectives: Our previous study demonstrated using an oral gavage model that Porphyromonas gingivalis could induce various inflammatory changes linked to periodontitis-associated systemic diseases by altering gut microbiota. A ligature-induced periodontitis model is similar to human periodontitis in various aspects: in both cases, alveolar bone resorption depends on oral bacterial load, and gingival tissue becomes infiltrated with inflammatory cells. Therefore, this model may be suitable for the analysis of bacterial burden and gingival tissue inflammation with changes related to systemic diseases.
    Material and Methods: Periodontal tissue destruction was induced by a 2 wk ligature placement around the bilateral maxillary second molar. We analyzed the expression profile of various genes in several tissues, levels of systemic inflammatory markers and induction of insulin resistance. In addition, we studied changes in gut microbiota composition and bacterial load in the oral cavity.
    Results: Two weeks after ligature placement gingival inflammation was significantly induced with a disrupted gingival epithelial barrier and alveolar bone resorption accompanied by increased bacterial burden in the oral cavity. Gene expression analysis of the gingival tissue of ligated mice demonstrated that interleukin (Il) 1b was significantly elevated and Il6 and Il17a tended to be higher in ligated mice than in untreated mice. Although serum IL-6 was significantly elevated and serum amyloid A tended to be higher in ligated compared to untreated mice, endotoxin levels did not differ between the two groups. Among the genes whose expressions are closely related to glucose and lipid metabolisms, only phosphoenolpyruvate carboxykinase 1 (Pck1) and acetyl-coenzyme A carboxylase alpha (Acaca) showed significant changes following ligature placement in the liver, with the former upregulated and the latter downregulated. However, insulin sensitivity did not change following ligature placement. Furthermore, ligature placement weakly affected the composition of gut microbiota and gene expression in the intestines.
    Conclusion: The results suggest that increased oral commensals and gingival inflammation have limited roles in the pathological changes to adipose and liver tissues, which are important organs whose dysfunctions contribute to the development of periodontitis-related systemic diseases.

    DOI: 10.1111/jre.12344

    Web of Science

    researchmap

  • Brazilian propolis mitigates impaired glucose and lipid metabolism in experimental periodontitis in mice Reviewed

    Mayuka Nakajima, Kei Arimatsu, Takayoshi Minagawa, Yumi Matsuda, Keisuke Sato, Naoki Takahashi, Takako Nakajima, Kazuhisa Yamazaki

    BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE   16   2016.8

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:BIOMED CENTRAL LTD  

    Background: Periodontitis has been implicated as a risk factor for metabolic disorders associated with insulin resistance. Recently, we have demonstrated that orally administered Porphyromonas gingivalis, a representative periodontopathic bacterium, induces endotoxemia via reduced gut barrier function coupled with changes in gut microbiota composition, resulting in systemic inflammation and insulin resistance. Propolis, a resinous substance collected by honeybees from leaf buds and cracks in the bark of various plants, can positively affect metabolic disorders in various experimental models. In this study, we thus aimed to clarify the effect of propolis on impaired glucose and lipid metabolism induced by P. gingivalis administration.
    Methods: Eight-week-old male C57BL/6 mice were orally administered P. gingivalis strain W83, propolis ethanol extract powder with P. gingivalis, or vehicle. We then analyzed the expression profile of glucose and lipid metabolism-related genes in the liver and adipose tissues. Serum endotoxin levels were also evaluated by a limulus amebocyte lysate test. In addition, we performed histological analysis of the liver and quantified alveolar bone loss by measuring the root surface area on the lower first molar.
    Results: Oral administration of P. gingivalis induced downregulation of genes that improve insulin sensitivity in adipose tissue (C1qtnf9, Irs1, and Sirt1), but upregulation of genes associated with lipid droplet formation and gluconeogenesis (Plin2, Acox, and G6pc). However, concomitant administration of propolis abrogated these adverse effects of P. gingivalis. Consistent with gene expression, histological analysis showed that administered propolis suppressed hepatic steatosis induced by P. gingivalis. Furthermore, propolis inhibited the elevation of serum endotoxin levels induced by P. gingivalis administration. Contrary to the systemic effects, propolis had no beneficial effect on alveolar bone loss.
    Conclusion: These results suggest that administration of propolis may be effective in suppressing periodontopathic bacteria-induced metabolic changes that increase the risk of various systemic diseases.

    DOI: 10.1186/s12906-016-1305-8

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-2875-2111

  • Microbiological and Clinical Effects of Sitafloxacin and Azithromycin in Periodontitis Patients Receiving Supportive Periodontal Therapy Reviewed

    Takako Nakajima, Takafumi Okui, Harue Ito, Mayuka Nakajima, Tomoyuki Honda, Yasuko Shimada, Koichi Tabeta, Kohei Akazawa, Kazuhisa Yamazaki

    ANTIMICROBIAL AGENTS AND CHEMOTHERAPY   60 ( 3 )   1779 - 1787   2016.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC MICROBIOLOGY  

    Sitafloxacin (STFX) is a newly developed quinolone that has robust antimicrobial activity against periodontopathic bacteria. We previously reported that oral administration of STFX during supportive periodontal therapy was as effective as conventional mechanical debridement under local anesthesia microbiologically and clinically for 3 months. The aim of the present study was to examine the short-term and long-term microbiological and clinical effects of systemic STFX and azithromycin (AZM) on active periodontal pockets during supportive periodontal therapy. Fifty-one patients receiving supportive periodontal therapy were randomly allocated to the STFX group (200 mg/day of STFX for 5 days) or the AZM group (500 mg/day of AZM for 3 days). The microbiological and clinical parameters were examined until 12 months after the systemic administration of each drug. The concentration of each drug in periodontal pockets and the antimicrobial susceptibility of clinical isolates were also analyzed. The proportions of red complex bacteria, i.e., Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia, which are the representative periodontopathic bacteria, were significantly reduced at 1 month and remained lower at 12 months than those at baseline in both the STFX and AZM groups. Clinical parameters were significantly improved over the 12-month period in both groups. An increase in the MIC of AZM against clinical isolates was observed in the AZM group. These results indicate that monotherapy with systemic STFX and AZM might be an alternative treatment during supportive periodontal therapy in patients for whom invasive mechanical treatment is inappropriate.

    DOI: 10.1128/AAC.02575-15

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-2875-2111

  • Oral Administration of P. gingivalis Induces Dysbiosis of Gut Microbiota and Impaired Barrier Function Leading to Dissemination of Enterobacteria to the Liver Reviewed

    Mayuka Nakajima, Kei Arimatsu, Tamotsu Kato, Yumi Matsuda, Takayoshi Minagawa, Naoki Takahashi, Hiroshi Ohno, Kazuhisa Yamazaki

    PLOS ONE   10 ( 7 )   2015.7

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:PUBLIC LIBRARY SCIENCE  

    Although periodontitis has been implicated as a risk factor for various systemic diseases, the precise mechanisms by which periodontitis induces systemic disease remain to be elucidated. We have previously revealed that repeated oral administration of Porphyromonas gingivalis elicits endotoxemia via changes in the gut microbiota of the ileum, and thereby induces systemic inflammation and insulin resistance. However, it is not clear to what extent a single administration of P. gingivalis could affect gut microbiota composition, gut barrier function, and subsequent influx of gut microbiota into the liver. Therefore, in the present study, C57BL/6 mice were orally administered P. gingivalis (strain W83) once and compared to sham-inoculated mice. The phylogenetic structure and diversity of microbial communities in the gut and liver were analyzed by pyrosequencing the 16S ribosomal RNA genes. Serum endotoxin activity was determined by a Limulus amebocyte lysate test. Gene expression in the intestine and expression of 16S rRNA genes in the blood and liver were examined by quantitative polymerase chain reaction. Administration of P. gingivalis significantly altered gut microbiota, with an increased proportion of phylum Bacteroidetes, a decreased proportion of phylum Firmicutes, and increased serum endotoxin levels. In the intestinal tissues, gene expression of tjp-1 and occludin, which are involved in intestinal permeability, were downregulated. Higher amounts of bacterial DNA were detected in the liver of infected mice. Importantly, changes in gut microbiota preceded systemic inflammatory changes. These results further support the idea that disturbance of the gut microbiota composition by orally derived periodontopathic bacteria may be a causal mechanism linking periodontitis and systemic disease.

    DOI: 10.1371/journal.pone.0134234

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-2875-2111

  • Oral pathobiont induces systemic inflammation and metabolic changes associated with alteration of gut microbiota Reviewed

    Kei Arimatsu, Hitomi Yamada, Haruna Miyazawa, Takayoshi Minagawa, Mayuka Nakajima, Mark I. Ryder, Kazuyoshi Gotoh, Daisuke Motooka, Shota Nakamura, Tetsuya Iida, Kazuhisa Yamazaki

    SCIENTIFIC REPORTS   4   2014.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:NATURE PUBLISHING GROUP  

    Periodontitis has been implicated as a risk factor for metabolic disorders such as type 2 diabetes, atherosclerotic vascular diseases, and non-alcoholic fatty liver disease. Although bacteremias from dental plaque and/or elevated circulating inflammatory cytokines emanating from the inflamed gingiva are suspected mechanisms linking periodontitis and these diseases, direct evidence is lacking. We hypothesize that disturbances of the gut microbiota by swallowed bacteria induce a metabolic endotoxemia leading metabolic disorders. To investigate this hypothesis, changes in the gut microbiota, insulin and glucose intolerance, and levels of tissue inflammation were analysed in mice after oral administration of Porphyromonas gingivalis, a representative periodontopathogens. Pyrosequencing revealed that the population belonging to Bacteroidales was significantly elevated in P. gingivalis-administered mice which coincided with increases in insulin resistance and systemic inflammation. In P. gingivalis-administered mice blood endotoxin levels tended to be higher, whereas gene expression of tight junction proteins in the ileum was significantly decreased. These results provide a new paradigm for the interrelationship between periodontitis and systemic diseases.

    DOI: 10.1038/srep04828

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-2875-2111

  • Th2 cytokines efficiently stimulate periostin production in gingival fibroblasts but periostin does not induce an inflammatory response in gingival epithelial cells Reviewed

    Mayuka Nakajima, Tomoyuki Honda, Sayuri Miyauchi, Kazuhisa Yamazaki

    ARCHIVES OF ORAL BIOLOGY   59 ( 2 )   93 - 101   2014.2

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:PERGAMON-ELSEVIER SCIENCE LTD  

    Objectives: This study aims to clarify whether gingival fibroblasts produce periostin in response to Th2 cytokines which are elevated in periodontitis lesion and, if so, whether periostin affects the inflammatory response and matrix-protein metabolism.
    Design: Human gingival fibroblasts, periodontal ligament cells and the gingival epithelial cell line epi4 were stimulated with interleukin-4 (IL-4), IL-13, tumour necrosis factor-alpha (TNF-alpha) and Porphyromonas gingivalis lipopolysaccharide (LPS). Periostin expression was analysed by real-time polymerase chain-reaction (PCR) and Western blotting. The expression of the IL-4 receptor alpha-chain was evaluated by immunocytochemistry. The effect of periostin on the production of inflammatory cytokines and the expression of matrix protein-related genes was analysed by real-time PCR and enzyme-linked immunosorbent assay (ELISA).
    Results: While IL-4 and IL-13 significantly induced periostin production in gingival fibroblasts and periodontal ligament cells, no effect was observed in epi4 cells. No stimulatory effect of TNF-alpha or P. gingivalis LPS on the production of periostin was observed. The effect of periostin on the production of inflammatory cytokines was weak in gingival fibroblasts; however, little or no effect was observed on periodontal ligament cells or epi4 cells. No significant effect of periostin on the expression of matrix protein-related genes was found.
    Conclusion: The results suggest that gingival fibroblasts may be a source of periostin in periodontitis lesions but periostin has only a limited role either in the inflammatory response or in matrix-protein metabolism. Thus, the role of periostin in the cellular interaction between epithelial and mesenchymal cells in gingiva may be distinct from that of skin. (C) 2013 Elsevier Ltd. All rights reserved.

    DOI: 10.1016/j.archoralbio.2013.10.004

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-2875-2111

▶ display all

MISC

  • Topical Treatment of Periodontitis Using an Iongel Reviewed

    Mayuka Nakajima, Samir Mitragotri

    99th General Session of the IADR   2021.7

     More details

  • イオンジェルを用いた局所歯周治療法の開発研究 Reviewed

    中島麻由佳, Samir Mitragotri, 多部田康一

    プログラムおよび講演抄録集   151項   2021.6

     More details

  • Topical application of choline-based ionic liquid (CAGE) to the treatment of oral infectious disease. Reviewed

    Eden E. L. Tanner, Nao Nakajima, Kelly N. Ibsen, Samir Mitragotr

    AIChE Annual Meeting   2020.11

     More details

  • Periodontal disease exacerbates NAFLD via dysbiosis of gut microbiota

    山崎恭子, 山崎恭子, 中島麻由佳, 竹内麻衣, 原実生, 都野隆博, 都野隆博, 松岸葵, 松岸葵, 松川由実, 佐藤圭祐, 高橋直紀, 多部田康一, 坪井裕理, 菊地淳, 加藤完, 大野博司, 山崎和久

    腸内細菌学雑誌   34 ( 2 )   2020

  • Porphyromonas gingivalis経口投与は腸内細菌の変化を介してNAFLD病態に影響を与える

    山崎恭子, 山崎恭子, 中島麻由佳, 竹内麻衣, 原実生, 原実生, 都野隆博, 都野隆博, 松岸葵, 松岸葵, 松川由実, 佐藤圭祐, 高橋直紀, 多部田康一, 山崎和久

    新潟歯学会雑誌   50 ( 2 )   2020

  • Porphyromonas gingivalisがNASH病態形成に与える影響の解析

    山崎恭子, 山崎恭子, 中島麻由佳, 中島麻由佳, 竹内麻衣, 竹内麻衣, 原(山田)実生, 原(山田)実生, 都野隆博, 都野隆博, 松岸葵, 松岸葵, 松川(松田, 由美, 佐藤圭祐, 高橋直紀, 多部田康一, 山崎和久

    日本歯周病学会会誌(Web)   61 ( 春季特別 )   125 - 125   2019.5

     More details

    Language:Japanese   Publisher:(NPO)日本歯周病学会  

    J-GLOBAL

    researchmap

  • Porphyromonas gingivalisがNASH病態を進行させるメカニズムの解析

    山崎恭子, 山崎恭子, 中島麻由佳, 竹内麻衣, 原実生, 原実生, 都野隆博, 都野隆博, 松岸葵, 松岸葵, 松川由実, 佐藤圭祐, 高橋直紀, 多部田康一, 山崎和久

    日本歯科保存学会学術大会プログラムおよび講演抄録集(Web)   151st   2019

  • Porphyromonas gingivalisが腸管上皮バリア機能へ及ぼす影響の解析

    都野隆博, 都野隆博, 高橋直紀, 竹内麻衣, 原実生, 中島麻由佳, 多部田康一, 山崎和久

    日本歯科保存学会学術大会プログラムおよび講演抄録集(Web)   150th   2019

  • Porphyromonas gingivalis口腔投与がDSS誘導性実験的腸炎におよぼす影響の解析

    都野隆博, 都野隆博, 高橋直紀, 竹内麻衣, 原実生, 中島麻由佳, 多部田康一, 山崎和久

    日本歯周病学会会誌(Web)   61   2019

  • Porphyromonas gingivalis口腔投与のコラーゲン誘導性関節炎増悪メカニズムの解析

    佐藤圭祐, 高橋直紀, 中島麻由佳, 松田由実, 山田実生, 横地麻衣, 中島貴子, 山崎和久

    日本歯科保存学会学術大会プログラムおよび講演抄録集(Web)   147th   56 (WEB ONLY) - 56   2017.10

     More details

    Language:Japanese   Publisher:(NPO)日本歯科保存学会  

    J-GLOBAL

    researchmap

  • Porphyromonas gingivalis口腔投与のコラーゲン誘導性関節炎増悪メカニズムの解析

    佐藤圭祐, 佐藤圭祐, 高橋直紀, 高橋直紀, 中島麻由佳, 中島麻由佳, 松田由実, 松田由実, 山田実生, 山田実生, 横地麻衣, 横地麻衣, 多部田康一, 中島貴子, 山崎和久

    新潟歯学会雑誌   46 ( 2 )   116 - 116   2016.12

     More details

    Language:Japanese   Publisher:新潟歯学会  

    J-GLOBAL

    researchmap

  • Periodontopathic bacteria increases the risk of rheumatoid arthritis by affecting gut immune system

    K. Sato, N. Takahashi, M. Nakajima, Y. Matsuda, M. Yamada, M. Yokoji, T. Kato, H. Ohno, K. Yamazaki

    EUROPEAN JOURNAL OF IMMUNOLOGY   46   640 - 640   2016.8

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:WILEY-BLACKWELL  

    Web of Science

    researchmap

  • 結紮誘導歯周炎モデルマウスにおける全身への影響とそのメカニズムの解析

    松田由実, 松田由実, 高橋直紀, 高橋直紀, 中島麻由佳, 中島麻由佳, 佐藤圭祐, 佐藤圭祐, 多部田康一, 中島貴子, 山崎和久

    新潟歯学会雑誌   46 ( 1 )   48 - 48   2016.7

     More details

    Language:Japanese   Publisher:新潟歯学会  

    J-GLOBAL

    researchmap

  • Porphyromonas gingivalis口腔投与のコラーゲン誘導性関節炎増悪メカニズムの解析

    佐藤圭祐, 佐藤圭祐, 高橋直紀, 高橋直紀, 中島麻由佳, 中島麻由佳, 松田由実, 松田由実, 山田実生, 山田実生, 横地麻衣, 横地麻衣, 多部田康一, 中島貴子, 山崎和久

    日本歯周病学会会誌(Web)   58 ( 春季特別 )   116 - 116   2016.4

     More details

    Language:Japanese   Publisher:(NPO)日本歯周病学会  

    J-GLOBAL

    researchmap

  • 歯周病安定期治療中の活動性歯周ポケットに対する経口抗菌療法の細菌学的,臨床的効果(第3報)

    伊藤晴江, 中島貴子, 奥井隆文, 奥井隆文, 中島麻由佳, 中島麻由佳, 本田朋之, 本田朋之, 島田靖子, 島田靖子, 多部田康一, 山崎和久

    日本歯周病学会会誌(Web)   58   2016

  • 歯周炎と血液・尿中バイオロジカルマーカーとの関連解析

    中島麻由佳, 中島麻由佳, 多部田康一, 宮内小百合, 杉田典子, 小松康高, 本田朋之, 高橋直紀, 宮澤春菜, 有松圭, 皆川高嘉, 松田由実, 松田由実, 佐藤圭祐, 佐藤圭祐, 山崎和久, 吉江弘正

    日本歯科保存学会学術大会プログラムおよび講演抄録集(Web)   143rd   P126 (WEB ONLY) - 197   2015.11

     More details

    Language:Japanese   Publisher:(NPO)日本歯科保存学会  

    J-GLOBAL

    researchmap

  • 歯周組織局所の炎症およびP.gingivalisによる腸内細菌叢の変動が全身に及ぼす影響とその分子機構の比較

    松田由実, 松田由実, 高橋直紀, 高橋直紀, 有松圭, 有松圭, 中島麻由佳, 中島麻由佳, 佐藤圭祐, 佐藤圭祐, 多部田康一, 中島貴子, 山崎和久

    日本歯周病学会会誌(Web)   57 ( 秋季特別 )   129 - 129   2015.8

     More details

    Language:Japanese   Publisher:(NPO)日本歯周病学会  

    J-GLOBAL

    researchmap

  • Porphyromonas gingivalis経口単回投与によるマウス腸内細菌叢の変動

    中島麻由佳, 中島麻由佳, 有松圭, 有松圭, 高橋直紀, 高橋直紀, 皆川高嘉, 皆川高嘉, 松田由実, 松田由実, 佐藤圭祐, 佐藤圭祐, 中島貴子, 多部田康一, 山崎和久

    新潟歯学会雑誌   45 ( 1 )   26 - 27   2015.6

     More details

    Language:Japanese   Publisher:新潟歯学会  

    J-GLOBAL

    researchmap

  • 歯周炎モデルマウスにおける腸内細菌叢の変動と免疫応答への影響

    高橋直紀, 有松圭, 中島麻由佳, 松田由実, 佐藤圭祐, 多部田康一, 中島貴子, 加藤完, 大野博司, 山崎和久

    腸内細菌学雑誌   29 ( 2 )   91 - 91   2015.4

     More details

    Language:Japanese   Publisher:(公財)日本ビフィズス菌センター  

    J-GLOBAL

    researchmap

  • Porphyromonas gingivalis経口単回投与によるマウス腸内細菌叢の変動

    中島麻由佳, 有松圭, 高橋直紀, 皆川高嘉, 山田ひとみ, 松田由実, 佐藤圭祐, 多部田康一, 中島貴子, 山崎和久

    日本歯科保存学会学術大会プログラムおよび講演抄録集(Web)   141st   B24 (WEB ONLY) - 62   2014.10

     More details

    Language:Japanese   Publisher:(NPO)日本歯科保存学会  

    J-GLOBAL

    researchmap

  • 結紮誘導歯周炎モデルマウスにおける全身への影響とその分子機構の解析

    松田由実, 松田由実, 山田ひとみ, 山田ひとみ, 高橋直紀, 高橋直紀, 有松圭, 有松圭, 皆川高嘉, 皆川高嘉, 中島麻由佳, 中島麻由佳, 多部田康一, 中島貴子, 山崎和久

    日本歯周病学会学術大会プログラムおよび講演抄録集   57th   2014

  • 歯周病原細菌誘導性糖・脂質代謝変動に及ぼすPropolisの効果

    中島麻由佳, 中島麻由佳, 有松圭, 有松圭, 高橋直紀, 高橋直紀, 山田ひとみ, 山田ひとみ, 皆川高嘉, 皆川高嘉, 松田由実, 松田由実, 多部田康一, 中島貴子, 山崎和久

    日本歯周病学会学術大会プログラムおよび講演抄録集   57th   2014

  • Porphyromonas gingivalis経口投与はマウス腸内細菌叢を変動させインスリン抵抗性を誘導する

    有松圭, 有松圭, 山田ひとみ, 山田ひとみ, 宮内小百合, 宮沢春菜, 中島麻由佳, 中島麻由佳, 多部田康一, 中島貴子, 山崎和久

    新潟歯学会雑誌   44 ( 2 )   2014

  • Porphyromonas gingivalis経口投与によるマウス腸内細菌叢の変動と内毒素血症の関連

    有松圭, 有松圭, 山田ひとみ, 山田ひとみ, 宮内小百合, 宮澤春菜, 宮澤春菜, 中島麻由佳, 中島麻由佳, 多部田康一, 中島貴子, 山崎和久

    日本歯周病学会学術大会プログラムおよび講演抄録集   57th   2014

  • 歯周病安定期治療中の活動性歯周ポケットに対する経口抗菌療法の細菌学的効果(第2報)

    中島貴子, 伊藤晴江, 奥井隆文, 中島麻由佳, 中島麻由佳, 宮下博考, 宮下博考, 多部田康一, 山崎和久

    日本歯周病学会学術大会プログラムおよび講演抄録集   56th   2013

  • 歯周ポケットからの臨床分離株に対する抗菌薬の有効性,耐性の検討

    中島貴子, 奥井隆文, 伊藤晴江, 中島麻由佳, 中島麻由佳, 多部田康一, 山崎和久

    日本歯科保存学会学術大会プログラムおよび講演抄録集(Web)   139th   2013

  • Porphyromonas gingivalis口腔感染マウスモデルで誘導されるインスリン抵抗性は脂肪組織及び肝臓における炎症反応と関連する

    有松圭, 有松圭, 土門久哲, 土門久哲, 山田ひとみ, 山田ひとみ, 宮内小百合, 宮内小百合, 宮沢春菜, 宮沢春菜, 皆川高嘉, 皆川高嘉, 中島麻由佳, 中島麻由佳, 中島貴子, 多部田康一, 山崎和久

    日本歯周病学会学術大会プログラムおよび講演抄録集   56th   2013

  • Porphyromonas gingivalis口腔感染はマウス腸内細菌叢を変動サせインスリン抵抗性を誘導する

    山崎和久, 有松圭, 有松圭, 土門久哲, 土門久哲, 山田ひとみ, 山田ひとみ, 宮内小百合, 宮内小百合, 宮澤春菜, 宮澤春菜, 皆川高嘉, 皆川高嘉, 中島麻由佳, 中島麻由佳, 中島貴子, 多部田康一

    日本歯周病学会学術大会プログラムおよび講演抄録集   56th   2013

▶ display all

Presentations

  • イオンジェルを⽤いた経粘膜局所感染制御法の開発

    中島 麻由佳, 多部田康一

    第65回春季⽇本⻭周病学会学術⼤会  2022.6 

     More details

    Presentation type:Oral presentation (general)  

    researchmap

Awards

  • UJA論文賞 奨励賞

    2024.5   海外日本人研究者ネットワーク(UJA)   Engineering of bioactive nanocomplexes on dental floss for targeted gingival therapy

    中島麻由佳

     More details

  • 奨励賞

    2023.10   日本歯科保存学会   Topical treatment of periodontitis using an iongel(Biomaterials. 276巻 121069頁 (2021年発行))

    中島麻由佳

     More details

  • 優秀発表賞(優秀ポスター賞)

    2022.11   日本歯科保存学会  

    中島麻由佳

     More details

  • オーラルサイエンス研究会優秀発表賞

    2022.11   オーラルサイエンス研究会  

    中島麻由佳

     More details

  • JACP/JSPポスター General/Basic Research部門(医歯薬出版賞)最優秀賞

    2022.10   第108回アメリカ歯周病学会共催日本臨床歯周病学会・日本歯周病学会  

    中島麻由佳

     More details

  • Flash Talk Audience Award in The 5th Japan-US Science Forum

    2020.12   JSPS Washington Office, Consulate-General of Japan in Boston, UJA  

     More details

  • 新潟歯学会賞

    2016.4   新潟歯学会  

    中島 麻由佳

     More details

  • IADR/Unilever Hatton Divisional Award

    2015.3   93rd International Association for Dental Research  

    中島 麻由佳

     More details

▶ display all

Research Projects

  • MPN 細菌操作による作為的口腔 Symbiotic マイクロバイオーム再構築

    2023.11 - 2025.3

    System name:Ⅲ研究助成金

    Awarding organization:テルモ生命科学振興財団

    中島麻由佳

      More details

    Authorship:Principal investigator 

    researchmap

  • Metal-Phenolic Network(MPN)細菌操作による口腔マイクロバイオーム 創薬研究

    2023.8 - 2028.3

    System name:医学系研究助成(臨床)

    Awarding organization:武田科学振興財団

    中島麻由佳

      More details

    Authorship:Principal investigator 

    researchmap

  • MPN武装細菌による口腔マイクロバイオームの再構築

    Grant number:23K18357

    2023.6 - 2025.3

    System name:科学研究費助成事業

    Research category:挑戦的研究(萌芽)

    Awarding organization:日本学術振興会

    多部田 康一, 江島 広貴, 野中 由香莉, 高橋 直紀, 中島 麻由佳

      More details

    Grant amount:\6500000 ( Direct Cost: \5000000 、 Indirect Cost:\1500000 )

    researchmap

  • Development of new modalities in periodontal treatment using cell binding backpack

    Grant number:23H03079

    2023.4 - 2026.3

    System name:Grants-in-Aid for Scientific Research

    Research category:Grant-in-Aid for Scientific Research (B)

    Awarding organization:Japan Society for the Promotion of Science

      More details

    Grant amount:\18850000 ( Direct Cost: \14500000 、 Indirect Cost:\4350000 )

    researchmap

  • Cellular Backpack応用によるin-situ細胞医薬の創製

    2023.1 - 2025.4

    System name:研究奨励金

    Awarding organization:上原記念生命科学財団

    中島麻由佳

      More details

    Authorship:Principal investigator 

    researchmap

  • The mechanism of specific induction of IgA-producing cells into the salivary glands

    Grant number:19K18993

    2022.4 - 2025.3

    System name:Grants-in-Aid for Scientific Research Grant-in-Aid for Early-Career Scientists

    Research category:Grant-in-Aid for Early-Career Scientists

    Awarding organization:Japan Society for the Promotion of Science

      More details

    Grant amount:\4290000 ( Direct Cost: \3300000 、 Indirect Cost:\990000 )

    researchmap

  • 腸内細菌叢の変動を介した歯周炎とNASHの関連メカニズムの解明

    2018.4 - 2021.3

    System name:特別研究員奨励費

    Awarding organization:新潟大学

    中島 麻由佳

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 腸内細菌叢の変動を介した歯周炎とNASHの関連メカニズムの解明

    2017.8 - 2018.3

    System name:研究活動スタート支援

    Awarding organization:新潟大学

    中島 麻由佳

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

▶ display all

 

Teaching Experience

  • 学問の扉 知と方法の最前線

    2023
    Institution name:新潟大学

  • 総合模型実習

    2023
    Institution name:新潟大学

  • 歯周病学

    2022
    Institution name:新潟大学