1
|
Deota S, Pendergast JS, Kolthur-Seetharam U, Esser KA, Gachon F, Asher G, Dibner C, Benitah SA, Escobar C, Muoio DM, Zhang EE, Hotamışlıgil GS, Bass J, Takahashi JS, Rabinowitz JD, Lamia KA, de Cabo R, Kajimura S, Longo VD, Xu Y, Lazar MA, Verdin E, Zierath JR, Auwerx J, Drucker DJ, Panda S. The time is now: accounting for time-of-day effects to improve reproducibility and translation of metabolism research. Nat Metab 2025; 7:454-468. [PMID: 40097742 DOI: 10.1038/s42255-025-01237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
The constant expansion of the field of metabolic research has led to more nuanced and sophisticated understanding of the complex mechanisms that underlie metabolic functions and diseases. Collaborations with scientists of various fields such as neuroscience, immunology and drug discovery have further enhanced the ability to probe the role of metabolism in physiological processes. However, many behaviours, endocrine and biochemical processes, and the expression of genes, proteins and metabolites have daily ~24-h biological rhythms and thus peak only at specific times of the day. This daily variation can lead to incorrect interpretations, lack of reproducibility across laboratories and challenges in translating preclinical studies to humans. In this Review, we discuss the biological, environmental and experimental factors affecting circadian rhythms in rodents, which can in turn alter their metabolic pathways and the outcomes of experiments. We recommend that these variables be duly considered and suggest best practices for designing, analysing and reporting metabolic experiments in a circadian context.
Collapse
Affiliation(s)
- Shaunak Deota
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Charna Dibner
- Department of Surgery and Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute for Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Deborah M Muoio
- Departments of Medicine and Pharmacology & Cancer Biology, Duke Molecular Physiology Institute, Durham, NC, USA
| | | | - Gökhan S Hotamışlıgil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Katja A Lamia
- Department of Molecular and Cellular Biology and Department of Molecular Medicine, the Scripps Research Institute, La Jolla, CA, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Shingo Kajimura
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- AIRC Institute of Molecular Oncology, Italian Foundation for Cancer Research Institute of Molecular Oncology, Milan, Italy
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity and Metabolism and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
2
|
LEI H, XU G, WEI Z, ZHAO L, LIANG F. Global trend of nondrug and nonsedativehypnotic treatment forinsomnia: a bibliometric study. J TRADIT CHIN MED 2024; 44:595-608. [PMID: 38767645 PMCID: PMC11077152 DOI: 10.19852/j.cnki.jtcm.20240408.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/27/2023] [Indexed: 05/22/2024]
Abstract
OBJECTIVE To present a bibliometric analysis of global scientific publications on the nondrug and nonsedative hypnotic treatment of insomnia with regard to influential institutions, publications, countries, research hotspots, trends, and frontiers. METHODS A literature review was conducted by searching the Web of Science Core Collection (WoSCC) and China National Knowledge Infrastructure (CNKI) databases to identify all publications related to the nondrug and nonsedative hypnotic treatment of insomnia from 2000 to 2021. Eligible publications were reviewed, including annual publication increments, citation analyses, international collaborations, and keyword analyses. The data were analysed using CiteSpace (vers5.8.R3, 6.1.R2 and 6.1.6, College of Computing and Informatics, Philadelphia, PA, USA) and virtualized by knowledge maps. RESULTS:In total, 9832 publications were included in this analysis. The results from the WoSCC showed that the United States of America (Count = 2268, 40.33%), Stanford University (Count = 141, 2.51%), and the United States Department of Health and Human Services were the leading country, institute, and funding agency regarding the number of publications, respectively. 'Cognitive-behavioural therapy" was the most popular research topic generated from the cocited reference. The most frequently co-occurring keywords were insomnia, cognitive behavioural therapy, disorder, depression, quality of life, Meta-analysis, older adult, sleep, prevalence and efficacy, while keywords including clinical practice guideline, guideline, and Tai Chi remained popular after 2021. Circadian rhythm was the strongest research frontier for 2000-2021. In China, Chengdu University of Traditional Chinese Medicine (Count = 69, 4.79%) was the most productive institute in this field. The most frequently co-occurring keywords from Chinese literature were sleep disorder, sleep quality, acupuncture and moxibustion, Parkinson's disease, transcranial magnetic stimulation, health education, music therapy, chronic insomnia, quality of life, and nonmotor symptoms. Traditional Chinese medicine was the strongest research frontier for 2019-2021. CONCLUSION This bibliometric study provides an exhaustive mapping encompassing pertinent institute, publications, influential articles, researchers and topics of the global trend of nondrug and nonsedative hypnotic treatment for insomnia. The results show that the research trend has shifted from primary studies on the efficacy and safety of nondrug and nonsedative hypnotic treatment for insomnia to comorbidity studies. Clinical practice guidelines will potentially become the research frontier for this field post-2021. The findings are important for researchers, clinicians, journal editors, and policy-makers working in the field of nondrug and nonsedative hypnotic treatment for insomnia to understand the strengths and potentials in the current studies and guide future clinical practice, research, and science policy.
Collapse
Affiliation(s)
- Hanzhou LEI
- 1 Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Guixing XU
- 1 Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Zepeng WEI
- 2 Department of Haematology, Royal North Shore Hospital, Sydney 2065, Australia
| | - Ling ZHAO
- 1 Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Fanrong LIANG
- 1 Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
3
|
Tanaka H, Hasebe R, Murakami K, Sugawara T, Yamasaki T, Murakami M. Gateway reflexes describe novel neuro-immune communications that establish immune cell gateways at specific vessels. Bioelectron Med 2023; 9:24. [PMID: 37936169 PMCID: PMC10631009 DOI: 10.1186/s42234-023-00126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023] Open
Abstract
Neuroinflammation is an important biological process induced by complex interactions between immune cells and neuronal cells in the central nervous system (CNS). Recent research on the bidirectional communication between neuronal and immunological systems has provided evidence for how immune and inflammatory processes are regulated by nerve activation. One example is the gateway reflex, in which immune cells bypass the blood brain barrier and infiltrate the CNS to cause neuroinflammation. We have found several modes of the gateway reflex in mouse models, in which gateways for immune cells are established at specific blood vessels in the spinal cords and brain in experimental autoimmune encephalomyelitis and systemic lupus erythematosus models, at retinal blood vessels in an experimental autoimmune uveitis model, and the ankle joints in an inflammatory arthritis model. Several environmental stimulations, including physical and psychological stresses, activate neurological pathways that alter immunological responses via the gateway reflex, thus contributing to the development/suppression of autoimmune diseases. In the manuscript, we describe the discovery of the gateway reflex and recent insights on how they regulate disease development. We hypothesize that artificial manipulation of specific neural pathways can establish and/or close the gateways to control the development of autoimmune diseases.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan.
| | - Rie Hasebe
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan
| | - Kaoru Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
| | - Toshiki Sugawara
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
| | - Takeshi Yamasaki
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan.
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan.
- Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Anagawa 4-9-1, Inage-Ku, Chiba, 263-8555, Japan.
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Nishi-11, Kita-21, Kuta-Ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
4
|
Oishi K, Yajima Y, Yoshida Y, Hagihara H, Miyakawa T, Higo-Yamamoto S, Toyoda A. Metabolic profiles of saliva in male mouse models of chronic sleep disorders induced by psychophysiological stress. Sci Rep 2023; 13:11156. [PMID: 37429932 PMCID: PMC10333369 DOI: 10.1038/s41598-023-38289-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
Disordered sleep is a global social problem and an established significant risk factor for psychological and metabolic diseases. We profiled non-targeted metabolites in saliva from mouse models of chronic sleep disorder (CSD). We identified 288 and 55 metabolites using CE-FTMS and LC-TOFMS, respectively, among which concentrations of 58 (CE-FTMS) and three (LC-TOFMS) were significantly changed by CSD. Pathway analysis revealed that CSD significantly suppressed glycine, serine and threonine metabolism. Arginine and proline metabolic pathways were among those that were both upregulated and downregulated. Pathways of alanine, aspartate and glutamate metabolism, genetic information processing, and the TCA cycle tended to be downregulated, whereas histidine metabolism tended to be upregulated in mice with CSD. Pyruvate, lactate, malate, succinate and the glycemic amino acids alanine, glycine, methionine, proline, and threonine were significantly decreased, whereas 3-hydroxybutyric and 2-hydroxybutyric acids associated with ketosis were significantly increased, suggesting abnormal glucose metabolism in mice with CSD. Increases in the metabolites histamine and kynurenic acid that are associated with the central nervous system- and decreased glycine, might be associated with sleep dysregulation and impaired cognitive dysfunction in mice with CSD. Our findings suggested that profiling salivary metabolites could be a useful strategy for diagnosing CSD.
Collapse
Affiliation(s)
- Katsutaka Oishi
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
- Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan.
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan.
- School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Yuhei Yajima
- College of Agriculture, Ibaraki University, Ami, Ibaraki, Japan
- Ibaraki Prefecture Livestock Research Center, Ishioka, Ibaraki, Japan
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Yuta Yoshida
- College of Agriculture, Ibaraki University, Ami, Ibaraki, Japan
| | - Hideo Hagihara
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Sayaka Higo-Yamamoto
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Atsushi Toyoda
- College of Agriculture, Ibaraki University, Ami, Ibaraki, Japan
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| |
Collapse
|
5
|
Yoshida Y, Yajima Y, Fujikura Y, Zhuang H, Higo-Yamamoto S, Toyoda A, Oishi K. Identification of salivary microRNA profiles in male mouse model of chronic sleep disorder. Stress 2023; 26:21-28. [PMID: 36522611 DOI: 10.1080/10253890.2022.2156783] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chronic sleep disorders (CSD) comprise a potential risk factor for metabolic and cardiovascular diseases, obesity and stroke. Thus, the identification of biomarkers for CSD is an important step in the early prevention of metabolic dysfunctions induced by sleep dysfunction. Diagnostic saliva samples can be easily and noninvasively collected. Thus, we aimed to identify whole microRNA (miRNA) profiles of saliva in control and psychophysiologically stressed CSD mouse models and compare them at Zeitgeber time (ZT) 0 (lights on) and ZT12 (lights off). The findings of two-way ANOVA revealed that the expression of 342 and 109 salivary miRNAs was affected by CSD and the time of day, respectively. Interactions were found in 122 miRNAs among which, we identified 197 (ZT0) and 62 (ZT12) upregulated, and 40 (ZT0) and seven (ZT12) downregulated miRNAs in CSD mice. We showed that miR-30c-5p, which is elevated in the plasma of patients with hypersomnia, was upregulated in the saliva of CSD mice collected at ZT0. The miRNAs, miR-10a-5p, miR-146b-5p, miR-150-5p, and miR-25-3p are upregulated in the serum of humans with poor sleep quality, and these were also upregulated in the saliva of CSD mice collected at ZT0. The miRNAs miR-30c, miR146b-5p, miR150, and miR-25-5p are associated with cardiovascular diseases, and we found that plasma concentrations of brain natriuretic peptides were significantly increased in CSD mice. The present findings showed that salivary miRNA profiles could serve as useful biomarkers for predicting CSD.
Collapse
Affiliation(s)
- Yuta Yoshida
- Department of Food and Life Sciences, College of Agriculture, Ibaraki University, Ami, Japan
| | - Yuhei Yajima
- Department of Food and Life Sciences, College of Agriculture, Ibaraki University, Ami, Japan
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu-City, Japan
| | - Yuri Fujikura
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Haotong Zhuang
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Sayaka Higo-Yamamoto
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Atsushi Toyoda
- Department of Food and Life Sciences, College of Agriculture, Ibaraki University, Ami, Japan
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu-City, Japan
| | - Katsutaka Oishi
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
- Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Japan
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
6
|
Han B, Kikuta S, Kamogashira T, Kondo K, Yamasoba T. Sleep deprivation induces delayed regeneration of olfactory sensory neurons following injury. Front Neurosci 2022; 16:1029279. [DOI: 10.3389/fnins.2022.1029279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
The circadian system, which is essential for the alignment of sleep/wake cycles, modulates adult neurogenesis. The olfactory epithelium (OE) has the ability to generate new neurons throughout life. Loss of olfactory sensory neurons (OSNs) as a result of injury to the OE triggers the generation of new OSNs, which are incorporated into olfactory circuits to restore olfactory sensory perception. This regenerative potential means that it is likely that the OE is substantially affected by sleep deprivation (SD), although how this may occur remains unclear. The aim of this study is to address how SD affects the process of OSN regeneration following OE injury. Mice were subjected to SD for 2 weeks, which induced changes in circadian activity. This condition resulted in decreased activity during the night-time and increased activity during the daytime, and induced no histological changes in the OE. However, when subjected to SD during the regeneration process after OE injury, a significant decrease in the number of mature OSNs in the dorsomedial area of the OE, which is the only area containing neurons expressing NQO1 (quinone dehydrogenase 1), was observed compared to the NQO1-negative OE. Furthermore, a significant decrease in proliferating basal cells was observed in the NQO1-positive OE compared to the NQO1-negative OE, but no increase in apoptotic OSNs was observed. These results indicate that SD accompanied by disturbed circadian activity could induce structurally negative effects on OSN regeneration, preferentially in the dorsomedial area of the OE, and that this area-specific regeneration delay might involve the biological activity of NQO1.
Collapse
|
7
|
Miyazaki K, Itoh N, Saiki P, Kuroki Y. Supplementation with Eurycoma longifolia Extract Modulates Diurnal Body Temperature Fluctuation and Sleep Rhythm in Mice. J Nutr Sci Vitaminol (Tokyo) 2022; 68:342-347. [PMID: 36047106 DOI: 10.3177/jnsv.68.342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Eurycoma longifolia (Tongkat Ali; TA) is a traditional medicinal herb, commonly known as Malaysian ginseng. The root tea has been traditionally applied to treat fevers, aches, sexual dysfunction and other ailments. We evaluated the effects of TA extract supplementation on diurnal core body temperature (BT) and sleep architecture in model mice. Dietary supplementation with TA extract for 4 wk resulted in significantly and moderately reduced BT during the rest and active phases, respectively. A high dose delayed the onset of BT elevation at the start of the active phase, indicating that the effect was dose-dependent. Electroencephalography findings revealed that dietary supplementation with TA extract changed sleep rhythms and delta power during the inactive phase of NREM sleep, indicating improved sleep quality. Our findings suggested that dietary TA extract could be a promising natural aid that alleviates sleep problems via thermoregulation.
Collapse
Affiliation(s)
- Koyomi Miyazaki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology
| | - Nanako Itoh
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology
| | - Papawee Saiki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology
| | | |
Collapse
|
8
|
Tomita S, Kusada H, Kojima N, Ishihara S, Miyazaki K, Tamaki H, Kurita R. Polymer-based chemical-nose systems for optical-pattern recognition of gut microbiota. Chem Sci 2022; 13:5830-5837. [PMID: 35685788 PMCID: PMC9132137 DOI: 10.1039/d2sc00510g] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Gut-microbiota analysis has been recognized as crucial in health management and disease treatment. Metagenomics, a current standard examination method for the gut microbiome, is effective but requires both expertise and significant amounts of general resources. Here, we show highly accessible sensing systems based on the so-called chemical-nose strategy to transduce the characteristics of microbiota into fluorescence patterns. The fluorescence patterns, generated by twelve block copolymers with aggregation-induced emission (AIE) units, were analyzed using pattern-recognition algorithms, which identified 16 intestinal bacterial strains in a way that correlates with their genome-based taxonomic classification. Importantly, the chemical noses classified artificial models of obesity-associated gut microbiota, and further succeeded in detecting sleep disorder in mice through comparative analysis of normal and abnormal mouse gut microbiota. Our techniques thus allow analyzing complex bacterial samples far more quickly, simply, and inexpensively than common metagenome-based methods, which offers a powerful and complementary tool for the practical analysis of the gut microbiome.
Collapse
Affiliation(s)
- Shunsuke Tomita
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
- DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), DBT-AIST International Center for Translational & Environmental Research (DAICENTER) Japan
| | - Hiroyuki Kusada
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology Japan
| | - Naoshi Kojima
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Sayaka Ishihara
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Koyomi Miyazaki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology Japan
| | - Hideyuki Tamaki
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology Japan
- JST ERATO Nomura Microbial Community Control Project, University of Tsukuba Japan
| | - Ryoji Kurita
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
- DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), DBT-AIST International Center for Translational & Environmental Research (DAICENTER) Japan
- Faculty of Pure and Applied Sciences, University of Tsukuba Japan
| |
Collapse
|
9
|
Lee JH, Moon E, Park J, Oh CE, Hong YR, Yoon M. Optimization of Analysis of Circadian Rest-Activity Rhythm Using Cosinor Analysis in Mice. Psychiatry Investig 2022; 19:380-385. [PMID: 35620823 PMCID: PMC9136527 DOI: 10.30773/pi.2021.0395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/05/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Data processing in analysis of circadian rhythm was performed in various ways. However, there was a lack of evidence for the optimal analysis of circadian rest-activity rhythm. Therefore, we aimed to perform mathematical simulations of data processing to investigate proper evidence for the optimal analysis of circadian rest-activity rhythm. METHODS Locomotor activities of 20 ICR male mice were measured by infrared motion detectors. The data of locomotor activities was processed using data summation, data average, and data moving average methods for cosinor analysis. Circadian indices were estimated according to time block, respectively. Also, statistical F and p-values were calculated by zero-amplitude test. RESULTS The data moving average result showed well-fitted cosine curves independent of data processing time. Meanwhile, the amplitude, MESOR, and acrophase were properly estimated within 800 seconds in data summation and data average methods. CONCLUSION These findings suggest that data moving average would be an optimal method for data processing in a cosinor analysis and data average within 800-second data processing time might be adaptable. The results of this study can be helpful to analyze circadian restactivity rhythms and integrate the results of the studies using different data processing methods.
Collapse
Affiliation(s)
- Jung Hyun Lee
- Department of Pediatrics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Eunsoo Moon
- Department of Psychiatry and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.,Department of Psychiatry, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jeonghyun Park
- Department of Psychiatry and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Chi Eun Oh
- Department of Pediatrics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Yoo Rha Hong
- Department of Pediatrics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Min Yoon
- Department of Applied Mathematics, Pukyung National University, Busan, Republic of Korea
| |
Collapse
|
10
|
Lange T, Luebber F, Grasshoff H, Besedovsky L. The contribution of sleep to the neuroendocrine regulation of rhythms in human leukocyte traffic. Semin Immunopathol 2022; 44:239-254. [PMID: 35041075 PMCID: PMC8901522 DOI: 10.1007/s00281-021-00904-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022]
Abstract
Twenty-four-hour rhythms in immune parameters and functions are robustly observed phenomena in biomedicine. Here, we summarize the important role of sleep and associated parameters on the neuroendocrine regulation of rhythmic immune cell traffic to different compartments, with a focus on human leukocyte subsets. Blood counts of "stress leukocytes" such as neutrophils, natural killer cells, and highly differentiated cytotoxic T cells present a rhythm with a daytime peak. It is mediated by morning increases in epinephrine, leading to a mobilization of these cells out of the marginal pool into the circulation following a fast, beta2-adrenoceptor-dependent inhibition of adhesive integrin signaling. In contrast, other subsets such as eosinophils and less differentiated T cells are redirected out of the circulation during daytime. This is mediated by stimulation of the glucocorticoid receptor following morning increases in cortisol, which promotes CXCR4-driven leukocyte traffic, presumably to the bone marrow. Hence, these cells show highest numbers in blood at night when cortisol levels are lowest. Sleep adds to these rhythms by actively suppressing epinephrine and cortisol levels. In addition, sleep increases levels of immunosupportive mediators, such as aldosterone and growth hormone, which are assumed to promote T-cell homing to lymph nodes, thus facilitating the initiation of adaptive immune responses during sleep. Taken together, sleep-wake behavior with its unique neuroendocrine changes regulates human leukocyte traffic with overall immunosupportive effects during nocturnal sleep. In contrast, integrin de-activation and redistribution of certain leukocytes to the bone marrow during daytime activity presumably serves immune regulation and homeostasis.
Collapse
Affiliation(s)
- Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany. .,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.
| | - Finn Luebber
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.,Social Neuroscience Lab, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | | |
Collapse
|
11
|
Duncan M, Guerriero L, Kohler K, Beechem L, Gillis B, Salisbury F, Wessel C, Wang J, Sunderam S, Bachstetter A, O’Hara B, Murphy M. Chronic Fragmentation of the Daily Sleep-Wake Rhythm Increases Amyloid-beta Levels and Neuroinflammation in the 3xTg-AD Mouse Model of Alzheimer's Disease. Neuroscience 2022; 481:111-122. [PMID: 34856352 PMCID: PMC8941625 DOI: 10.1016/j.neuroscience.2021.11.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 01/17/2023]
Abstract
Fragmentation of the daily sleep-wake rhythm with increased nighttime awakenings and more daytime naps is correlated with the risk of development of Alzheimer's disease (AD). To explore whether a causal relationship underlies this correlation, the present study tested the hypothesis that chronic fragmentation of the daily sleep-wake rhythm stimulates brain amyloid-beta (Aβ) levels and neuroinflammation in the 3xTg-AD mouse model of AD. Female 3xTg-AD mice were allowed to sleep undisturbed or were subjected to chronic sleep fragmentation consisting of four daily sessions of enforced wakefulness (one hour each) evenly distributed during the light phase, five days a week for four weeks. Piezoelectric sleep recording revealed that sleep fragmentation altered the daily sleep-wake rhythm to resemble the pattern observed in AD. Levels of amyloid-beta (Aβ40 and Aβ42) determined by ELISA were higher in hippocampal tissue collected from sleep-fragmented mice than from undisturbed controls. In contrast, hippocampal levels of tau and phospho-tau differed minimally between sleep fragmented and undisturbed control mice. Sleep fragmentation also stimulated neuroinflammation as shown by increased expression of markers of microglial activation and proinflammatory cytokines measured by q-RT-PCR analysis of hippocampal samples. No significant effects of sleep fragmentation on Aβ, tau, or neuroinflammation were observed in the cerebral cortex. These studies support the concept that improving sleep consolidation in individuals at risk for AD may be beneficial for slowing the onset or progression of this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- M.J. Duncan
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| | - L.E. Guerriero
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - K. Kohler
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - L.E. Beechem
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536
| | - B.D. Gillis
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536
| | - F. Salisbury
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - C. Wessel
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - J. Wang
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - S. Sunderam
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - A.D. Bachstetter
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536
| | - B.F. O’Hara
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - M.P. Murphy
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| |
Collapse
|
12
|
Murakami K, Tanaka Y, Murakami M. The gateway reflex: breaking through the blood barriers. Int Immunol 2021; 33:743-748. [PMID: 34505147 DOI: 10.1093/intimm/dxab064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
We have been studying inflammatory diseases, with a special focus on IL-6, and discovered two concepts related to inflammation development. One is the gateway reflex, which is induced by the activation of specific neural circuits followed by establishing gateways for autoreactive CD4 + T cells to pass through blood barriers toward the central nervous system (CNS) and retina during tissue-specific inflammatory diseases. We found that the formation of these gateways is dependent on the IL-6 amplifier, which is machinery for enhanced NF-κB activation in endothelial cells at specific sites. We have found five gateway reflexes in total. Here, we introduce the gateway reflex and the IL-6 amplifier.
Collapse
Affiliation(s)
- Kaoru Murakami
- Division of Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Nishi 7, Kita 15 jo, Kita-ku, Sapporo 060-0808, Hokkaido, Japan
| | - Yuki Tanaka
- Division of Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Nishi 7, Kita 15 jo, Kita-ku, Sapporo 060-0808, Hokkaido, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| | - Masaaki Murakami
- Division of Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Nishi 7, Kita 15 jo, Kita-ku, Sapporo 060-0808, Hokkaido, Japan.,Group of Quantum Immunology, Institute for Quantum Life Science, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| |
Collapse
|
13
|
Zhao Q, Koyama S, Yoshihara N, Takagi A, Komiyama E, Wada A, Oka A, Ikeda S. The Alopecia Areata Phenotype Is Induced by the Water Avoidance Stress Test In cchcr1-Deficient Mice. Biomedicines 2021; 9:biomedicines9070840. [PMID: 34356904 PMCID: PMC8301465 DOI: 10.3390/biomedicines9070840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/11/2021] [Accepted: 07/15/2021] [Indexed: 11/16/2022] Open
Abstract
We recently discovered a nonsynonymous variant in the coiled-coil alpha-helical rod protein 1 (CCHCR1) gene within the alopecia areata (AA) risk haplotype. We also reported that the engineered mice with this risk allele exhibited. To investigate more about the involvement of the CCHCR1 gene in AA pathogenesis, we developed an AA model using C57BL/6N cchcr1 gene knockout mice. In this study, mice (6–8 weeks) were divided into two groups: cchcr1−/− mice and wild-type (WT) littermates. Both groups were subjected to a water avoidance stress (WAS) test. Eight weeks after the WAS test, 25% of cchcr1−/− mice exhibited non-inflammatory foci of alopecia on the dorsal skin. On the other hand, none of wild-type littermates cause hair loss. The foci resembled human AA in terms of gross morphology, trichoscopic findings and histological findings. Additionally, gene expression microarray analysis of cchcr1−/− mice revealed abnormalities of hair related genes compared to the control. Our results strongly suggest that CCHCR1 is associated with AA pathogenesis and that cchcr1−/− mice are a good model for investigating AA.
Collapse
Affiliation(s)
- Qiaofeng Zhao
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (Q.Z.); (S.K.); (N.Y.); (A.T.); (E.K.); (A.W.)
| | - Satoshi Koyama
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (Q.Z.); (S.K.); (N.Y.); (A.T.); (E.K.); (A.W.)
| | - Nagisa Yoshihara
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (Q.Z.); (S.K.); (N.Y.); (A.T.); (E.K.); (A.W.)
| | - Atsushi Takagi
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (Q.Z.); (S.K.); (N.Y.); (A.T.); (E.K.); (A.W.)
| | - Etsuko Komiyama
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (Q.Z.); (S.K.); (N.Y.); (A.T.); (E.K.); (A.W.)
| | - Akino Wada
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (Q.Z.); (S.K.); (N.Y.); (A.T.); (E.K.); (A.W.)
| | - Akira Oka
- The Institute of Medical Sciences, Tokai University, Kanagawa 259-1193, Japan;
| | - Shigaku Ikeda
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (Q.Z.); (S.K.); (N.Y.); (A.T.); (E.K.); (A.W.)
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Correspondence: ; Tel.: +81-3-5802-1089; Fax: +81-3-3813-2205
| |
Collapse
|
14
|
Memory dysfunction and anxiety-like behavior in a mouse model of chronic sleep disorders. Biochem Biophys Res Commun 2020; 529:175-179. [DOI: 10.1016/j.bbrc.2020.05.218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/31/2020] [Indexed: 01/12/2023]
|
15
|
Dietary natural cocoa ameliorates disrupted circadian rhythms in locomotor activity and sleep-wake cycles in mice with chronic sleep disorders caused by psychophysiological stress. Nutrition 2020; 75-76:110751. [DOI: 10.1016/j.nut.2020.110751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/16/2019] [Accepted: 12/31/2019] [Indexed: 12/25/2022]
|
16
|
Higo-Yamamoto S, Yamamoto S, Miyazaki K, Nakakita Y, Kaneda H, Takata Y, Nakamura T, Oishi K. Dietary Heat-Killed Lactobacillus brevis SBC8803 Attenuates Chronic Sleep Disorders Induced by Psychophysiological Stress in Mice. J Nutr Sci Vitaminol (Tokyo) 2019; 65:164-170. [PMID: 31061285 DOI: 10.3177/jnsv.65.164] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We previously reported that dietary heat-killed Lactobacillus brevis SBC8803 affects sleep in mice and humans. The present study examined whether SBC8803 improves psychophysiological stress-induced chronic sleep disorders (CSD) using a mouse model characterized by disrupted circadian rhythms of wheel-running activity and sleep-wake cycles. Mice were fed with a diet supplemented with 0.5% heat-killed SBC8803 for 6 wk and imposed stress-induced CSD for last 2 wk. Dietary SBC8803 suppressed the reduction in wheel-running activity induced by CSD. Electroencephalography (EEG) revealed that SBC8803 significantly restored wakefulness and increased non-rapid eye movement (NREM) sleep during the second half of the active phase during CSD. The CSD-induced reduction in EEG slow wave activity, a marker of NREM sleep intensity, during the beginning of the inactive phase was significantly improved by SBC8803 supplementation. These findings suggest that dietary heat-killed SBC8803 confers beneficial effects on insomnia and circadian sleep disorders induced by psychophysiological stress.
Collapse
Affiliation(s)
- Sayaka Higo-Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)
| | - Saori Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)
| | - Koyomi Miyazaki
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)
| | | | | | | | | | - Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST).,Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo.,School of Integrative and Global Majors (SIGMA), University of Tsukuba
| |
Collapse
|
17
|
Abstract
The systemic regulation of immune reactions by the nervous system is well studied and depends on the release of hormones. Some regional regulations of immune reactions, on the other hand, depend on specific neural pathways. Better understanding of these regulations will expand therapeutic applications for neuroimmune and organ-to-organ functional interactions. Here, we discuss one regional neuroimmune interaction, the gateway reflex, which converts specific neural inputs into local inflammatory outputs in the CNS. Neurotransmitters released by the inputs stimulate specific blood vessels to express chemokines, which serve as a gateway for immune cells to extravasate into the target organ such as the brain or spinal cord. Several types of gateway reflexes have been reported, and each controls distinct CNS blood vessels to form gateways that elicit local inflammation, particularly in the presence of autoreactive immune cells. For example, neural stimulation by gravity creates the initial entry point to the CNS by CNS-reactive pathogenic CD4+ T cells at the dorsal vessels of fifth lumbar spinal cord, while pain opens the gateway at the ventral side of blood vessels in the spinal cord. In addition, it was recently found that local inflammation by the gateway reflex in the brain triggers the activation of otherwise resting neural circuits to dysregulate organ functions in the periphery including the upper gastrointestinal tract and heart. Therefore, the gateway reflex represents a novel bidirectional neuroimmune interaction that regulates organ functions and could be a promising target for bioelectric medicine.
Collapse
Affiliation(s)
- D Kamimura
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - M Murakami
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
18
|
Sleep Disturbance as a Potential Modifiable Risk Factor for Alzheimer's Disease. Int J Mol Sci 2019; 20:ijms20040803. [PMID: 30781802 PMCID: PMC6412395 DOI: 10.3390/ijms20040803] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/30/2019] [Accepted: 02/03/2019] [Indexed: 12/13/2022] Open
Abstract
Sleep disturbance is a common symptom in patients with various neurodegenerative diseases, including Alzheimer’s disease (AD), and it can manifest in the early stages of the disease. Impaired sleep in patients with AD has been attributed to AD pathology that affects brain regions regulating the sleep–wake or circadian rhythm. However, recent epidemiological and experimental studies have demonstrated an association between impaired sleep and an increased risk of AD. These studies have led to the idea of a bidirectional relationship between AD and impaired sleep; in addition to the conventional concept that impaired sleep is a consequence of AD pathology, various evidence strongly suggests that impaired sleep is a risk factor for the initiation and progression of AD. Despite this recent progress, much remains to be elucidated in order to establish the benefit of therapeutic interventions against impaired sleep to prevent or alleviate the disease course of AD. In this review, we provide an overview of previous studies that have linked AD and sleep. We then highlight the studies that have tested the causal relationship between impaired sleep and AD and will discuss the molecular and cellular mechanisms underlying this link. We also propose future works that will aid the development of a novel disease-modifying therapy and prevention of AD via targeting impaired sleep through non-pharmacological and pharmacological interventions.
Collapse
|
19
|
Shigiyama F, Kumashiro N, Tsuneoka Y, Igarashi H, Yoshikawa F, Kakehi S, Funato H, Hirose T. Mechanisms of sleep deprivation-induced hepatic steatosis and insulin resistance in mice. Am J Physiol Endocrinol Metab 2018; 315:E848-E858. [PMID: 29989853 DOI: 10.1152/ajpendo.00072.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sleep deprivation is associated with increased risk for type 2 diabetes mellitus. However, the underlying mechanisms of sleep deprivation-induced glucose intolerance remain elusive. The aim of this study was to investigate the mechanisms of sleep deprivation-induced glucose intolerance in mice with a special focus on the liver. We established a mouse model of sleep deprivation-induced glucose intolerance using C57BL/6J male mice. A single 6-h sleep deprivation by the gentle handling method under fasting condition induced glucose intolerance. Hepatic glucose production assessed by a pyruvate challenge test was significantly increased, as was hepatic triglyceride content (by 67.9%) in the sleep deprivation group, compared with freely sleeping control mice. Metabolome and microarray analyses were used to evaluate hepatic metabolites and gene expression levels and to determine the molecular mechanisms of sleep deprivation-induced hepatic steatosis. Hepatic metabolites, such as acetyl coenzyme A, 3β-hydroxybutyric acid, and certain acylcarnitines, were significantly increased in the sleep deprivation group, suggesting increased lipid oxidation in the liver. In contrast, fasted sleep-deprived mice showed that hepatic gene expression levels of elongation of very long chain fatty acids-like 3, lipin 1, perilipin 4, perilipin 5, and acyl-CoA thioesterase 1, which are known to play lipogenic roles, were 2.7, 4.5, 3.7, 2.9, and 2.8 times, respectively, those of the fasted sleeping control group, as assessed by quantitative RT-PCR. Sleep deprivation-induced hepatic steatosis and hepatic insulin resistance seem to be mediated through upregulation of hepatic lipogenic enzymes.
Collapse
Affiliation(s)
- Fumika Shigiyama
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Naoki Kumashiro
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Hiroyuki Igarashi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Fukumi Yoshikawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Saori Kakehi
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine , Tokyo , Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiromasa Funato
- Department of Anatomy, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Takahisa Hirose
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| |
Collapse
|
20
|
Saiki P, Nakajima Y, Van Griensven LJLD, Miyazaki K. Real-time monitoring of IL-6 and IL-10 reporter expression for anti-inflammation activity in live RAW 264.7 cells. Biochem Biophys Res Commun 2018; 505:885-890. [PMID: 30301531 DOI: 10.1016/j.bbrc.2018.09.173] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 11/29/2022]
Abstract
In previous study, we suggested that the interleukin (IL)-6 and IL-10 could serve as a good biomarker for anti-inflammation that related to chronic inflammatory disease. Recently, we are finding new anti-inflammation compounds from natural products by screening of IL-6 and IL-10 levels. Although, we could measure IL-6 and IL-10 levels by several methods. However, all methods could not measure continuous kinetic of IL-6 and IL-10 levels. Most methods have multiple steps and take a long time. Therefore, there is no a suitable method for screening. To this end, we established IL-6 and IL-10 promoter assay which can monitor with reference gene as Glyceraldehyde 3-phosphate dehydrogenase (gapdh) promoter in living single cell. It could determine IL-6 and IL-10 levels continuously in real-time within two steps. We evaluated IL-6 and IL-10 reporter expression in LPS-induced RAW 264.7 cells with well-known anti-inflammatory compounds such as quercetin, xanthones, β-D-glucan and dexamethasone. As the results, the expression of IL-6 and IL-10 reporters were strongly induced by LPS. The expression of IL-6 reporter was inhibited by all anti-inflammation compounds in LPS-induced RAW 264.7 cells. The expression of IL-10 reporter was inhibited by quercetin, xanthones and dexamethasone in LPS-induced RAW 264.7 cells. While, expression of IL-10 reporter was induced by β-D-glucan. These results indicated that this assay could use for determination of IL-6 and IL-10 reporter expression in LPS-induced RAW 264.7 cells for anti-inflammation activity. Moreover, the results showed that natural compounds have an effect on the time course of IL-6 and IL-10 expressions. Therefore, real-time monitoring has a merit for natural compounds screening. We suggested that this assay could serve as a compound screening assay for anti-inflammation activity.
Collapse
Affiliation(s)
- Papawee Saiki
- Biomedical Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba, Ibaraki, Japan.
| | - Yoshihiro Nakajima
- Health Research Institute, National Institute of Advance Industrial Science and Technology, Takamatsu, Kagawa, Japan
| | - Leo J L D Van Griensven
- Plant Research International, Wageningen University and Research Centre, Wageningen, the Netherlands
| | - Koyomi Miyazaki
- Biomedical Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| |
Collapse
|
21
|
Gateway reflex: Local neuroimmune interactions that regulate blood vessels. Neurochem Int 2018; 130:104303. [PMID: 30273641 DOI: 10.1016/j.neuint.2018.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/28/2018] [Indexed: 02/06/2023]
Abstract
Neuroimmunology is a research field that intersects neuroscience and immunology, with the larger aim of gaining significant insights into the pathophysiology of chronic inflammatory diseases such as multiple sclerosis. Conventional studies in this field have so far mainly dealt with immune responses in the nervous system (i.e. neuroinflammation) or systemic immune regulation by the release of glucocorticoids. On the other hand, recently accumulating evidence has indicated bidirectional interactions between specific neural activations and local immune responses. Here we discuss one such local neuroimmune interaction, the gateway reflex. The gateway reflex represents a mechanism that translates specific neural stimulations into local inflammatory outcomes by changing the state of specific blood vessels to allow immune cells to extravasate, thus forming the gateway. Several types of gateway reflex have been identified, and each regulates distinct blood vessels to create gateways for immune cells that induce local inflammation. The gateway reflex represents a novel therapeutic strategy for neuroinflammation and is potentially applicable to other inflammatory diseases in peripheral organs.
Collapse
|
22
|
Takeishi K, Kawaguchi H, Akioka K, Noguchi M, Arimura E, Abe M, Ushikai M, Okita S, Tanimoto A, Horiuchi M. Effects of Dietary and Lighting Conditions on Diurnal Locomotor Activity and Body Temperature in Microminipigs. ACTA ACUST UNITED AC 2018; 32:55-62. [PMID: 29275299 DOI: 10.21873/invivo.11204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/25/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022]
Abstract
The effects of dietary and lighting conditions on diurnal rhythm of locomotor activity (LA) and body temperature (BT) using four adult male microminipigs were investigated. Different feeding times, diet and lighting conditions were applied sequentially for 3 weeks in each phase as follows: Phase I: Morning mealtime, normal diet, 12-h lights on; phase II: mealtime changed to afternoon; phase III: diet changed to high-fat diet; phase IV: lighting changed to 20-h on; and phase V: phase I repeated. LA was measured by an actigraph which was worn on the body of each pig. A BT recording module (Thermochron Type-SL) was implanted in the neck subcutaneously. Phase II increased BT compared with phase I. Phase III increased LA and BT compared with phase II. Phase IV increased LA compared with phase III. LA in phase V was higher compared with phase I. These results can be extrapolated to other diurnal animals such as humans. This study provides an example of the effects of diet and lighting on biological activities in microminipigs under low-invasive procedures measuring LA and BT, leading to low variations in these measures.
Collapse
Affiliation(s)
- Kaichiro Takeishi
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Hiroaki Kawaguchi
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Kohei Akioka
- Laboratory of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Michiko Noguchi
- Laboratory of Theriogenology, Azabu University, Kanagawa, Japan
| | - Emi Arimura
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.,Department of Life and Environmental Science, Kagoshima Prefectural College, Kagoshima, Japan
| | - Masaharu Abe
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Miharu Ushikai
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Shinobu Okita
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Akihide Tanimoto
- Department of Pathology, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Masahisa Horiuchi
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
23
|
Saiki P, Kawano Y, Van Griensven LJLD, Miyazaki K. The anti-inflammatory effect of Agaricus brasiliensis is partly due to its linoleic acid content. Food Funct 2018; 8:4150-4158. [PMID: 29022634 DOI: 10.1039/c7fo01172e] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
For hundreds of years mushrooms have been used as functional food for health. The basidiomycete Agaricus brasiliensis (A. brasiliensis) is famous for the medicinal properties of its beta glucans and of its antioxidants. Most researchers have studied polysaccharides from A. brasiliensis for their anti-inflammatory activity. However, active compounds from this mushroom have not yet been studied for the inactivation of NO inhibitory activity. The present study aimed to find the active compounds from A. brasiliensis for their NO inhibitory activity related inflammatory activity. This study found that linoleic acid isolated from A. brasiliensis inhibited NO production and suppressed the expression of pro-inflammatory cytokines including TNF-α, IL-6, IL-1β, and NOS2 in RAW 264.7 cells. Linoleic acid also suppressed the expression of NF-κB subunit p50 and restored PPARα. This leads to the conclusion that linoleic acid from A. brasiliensis could reduce NO production and inflammatory activity in RAW 264.7 cells by the inhibition of p50 and via the activation of PPARα. This study suggests that linoleic acid present in A. brasiliensis could play a role in the prevention of inflammatory diseases for which this edible mushroom is already known.
Collapse
Affiliation(s)
- Papawee Saiki
- Biomedical Research Institute, National institute of Advance Industrial Science and Technology, Tsukuba, Ibaraki, Japan.
| | | | | | | |
Collapse
|
24
|
Kamimura D, Ohki T, Arima Y, Murakami M. Gateway reflex: neural activation-mediated immune cell gateways in the central nervous system. Int Immunol 2018; 30:281-289. [DOI: 10.1093/intimm/dxy034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/12/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Daisuke Kamimura
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Hokkaido, Japan
| | - Takuto Ohki
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Hokkaido, Japan
| | - Yasunobu Arima
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Hokkaido, Japan
| | - Masaaki Murakami
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Hokkaido, Japan
| |
Collapse
|
25
|
Oishi K, Ohyama S, Higo-Yamamoto S. Chronic sleep disorder induced by psychophysiological stress induces glucose intolerance without adipose inflammation in mice. Biochem Biophys Res Commun 2018; 495:2616-2621. [PMID: 29288667 DOI: 10.1016/j.bbrc.2017.12.158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 12/26/2017] [Indexed: 01/11/2023]
Abstract
Sleep disturbances are associated with various metabolic diseases such as hypertension and diabetes. We had previously established a mouse model of a psychophysiological stress-induced chronic sleep disorder (CSD) characterized by disrupted circadian rhythms of wheel-running activity, core body temperature, and sleep-wake cycles. To evaluate the underlying mechanisms of metabolic disorders induced by CSD, we created mice with CSD for six weeks and fed them with a high-fat diet. Glucose intolerance with hyperglycemia resulted, although plasma insulin levels and body weight increases were identical between control and CSD mice. Gluconeogenesis and glycolysis were enhanced and suppressed, respectively, in the livers of CSD mice, because the mRNA expression of Pck1 was significantly increased, whereas that of Gck and Pklr were significantly decreased in the CSD mice. Adipose inflammation induced by the high-fat diet seemed suppressed by the CSD, because the mRNA expression levels of Adgre1, Ccl2, and Tnf were significantly downregulated in the adipose tissues of CSD mice. These findings suggest that CSD impair glucose tolerance by inducing gluconeogenesis and suppressing glycolysis. Hyperphasia with hypoleptinemia, hypercorticosteronemia, and increased plasma free fatty acids might be involved in the impaired glucose metabolism under a CSD. Further studies are needed to elucidate the endocrine and molecular mechanisms underlying the associations between sleep disorders and impaired glucose homeostasis that consequently causes diabetes.
Collapse
Affiliation(s)
- Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan.
| | - Sumika Ohyama
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Sayaka Higo-Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| |
Collapse
|
26
|
Arima Y, Ohki T, Nishikawa N, Higuchi K, Ota M, Tanaka Y, Nio-Kobayashi J, Elfeky M, Sakai R, Mori Y, Kawamoto T, Stofkova A, Sakashita Y, Morimoto Y, Kuwatani M, Iwanaga T, Yoshioka Y, Sakamoto N, Yoshimura A, Takiguchi M, Sakoda S, Prinz M, Kamimura D, Murakami M. Brain micro-inflammation at specific vessels dysregulates organ-homeostasis via the activation of a new neural circuit. eLife 2017; 6. [PMID: 28809157 PMCID: PMC5557598 DOI: 10.7554/elife.25517] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 07/10/2017] [Indexed: 12/13/2022] Open
Abstract
Impact of stress on diseases including gastrointestinal failure is well-known, but molecular mechanism is not understood. Here we show underlying molecular mechanism using EAE mice. Under stress conditions, EAE caused severe gastrointestinal failure with high-mortality. Mechanistically, autoreactive-pathogenic CD4+ T cells accumulated at specific vessels of boundary area of third-ventricle, thalamus, and dentate-gyrus to establish brain micro-inflammation via stress-gateway reflex. Importantly, induction of brain micro-inflammation at specific vessels by cytokine injection was sufficient to establish fatal gastrointestinal failure. Resulting micro-inflammation activated new neural pathway including neurons in paraventricular-nucleus, dorsomedial-nucleus-of-hypothalamus, and also vagal neurons to cause fatal gastrointestinal failure. Suppression of the brain micro-inflammation or blockage of these neural pathways inhibited the gastrointestinal failure. These results demonstrate direct link between brain micro-inflammation and fatal gastrointestinal disease via establishment of a new neural pathway under stress. They further suggest that brain micro-inflammation around specific vessels could be switch to activate new neural pathway(s) to regulate organ homeostasis. DOI:http://dx.doi.org/10.7554/eLife.25517.001
Collapse
Affiliation(s)
- Yasunobu Arima
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takuto Ohki
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoki Nishikawa
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Anesthesiology and Critical Care Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kotaro Higuchi
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mitsutoshi Ota
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Tanaka
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mohamed Elfeky
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Behera, Egypt
| | - Ryota Sakai
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Mori
- Laboratory of Biofunctional Imaging, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tadafumi Kawamoto
- Radioisotope Research Institute, Department of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Andrea Stofkova
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yukihiro Sakashita
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuji Morimoto
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaki Kuwatani
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshichika Yoshioka
- Laboratory of Biofunctional Imaging, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Mitsuyoshi Takiguchi
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Saburo Sakoda
- Department of Neurology, National Hospital Organization Toneyama National Hospital, Osaka, Japan
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daisuke Kamimura
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaaki Murakami
- Division of Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
27
|
Minakawa EN, Miyazaki K, Maruo K, Yagihara H, Fujita H, Wada K, Nagai Y. Chronic sleep fragmentation exacerbates amyloid β deposition in Alzheimer's disease model mice. Neurosci Lett 2017; 653:362-369. [PMID: 28554860 DOI: 10.1016/j.neulet.2017.05.054] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/20/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
Abstract
Sleep fragmentation due to intermittent nocturnal arousal resulting in a reduction of total sleep time and sleep efficiency is a common symptom among people with Alzheimer's disease (AD) and elderly people with normal cognitive function. Although epidemiological studies have indicated an association between sleep fragmentation and elevated risk of AD, a relevant disease model to elucidate the underlying mechanisms was lacking owing to technical limitations. Here we successfully induced chronic sleep fragmentation in AD model mice using a recently developed running-wheel-based device and demonstrate that chronic sleep fragmentation increases amyloid β deposition. Notably, the severity of amyloid β deposition exhibited a significant positive correlation with the extent of sleep fragmentation. These findings provide a useful contribution to the development of novel treatments that decelerate the disease course of AD in the patients, or decrease the risk of developing AD in healthy elderly people through the improvement of sleep quality.
Collapse
Affiliation(s)
- Eiko N Minakawa
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Koyomi Miyazaki
- Physiologically Active Substances Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
| | - Kazushi Maruo
- Department of Clinical Epidemiology, Translational Medical Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan.
| | - Hiroko Yagihara
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Hiromi Fujita
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Keiji Wada
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan; Translational Medical Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8551, Japan.
| | - Yoshitaka Nagai
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan; Department of Neurotherapeutics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
28
|
Ikeda M, Hojo Y, Komatsuzaki Y, Okamoto M, Kato A, Takeda T, Kawato S. Hippocampal spine changes across the sleep-wake cycle: corticosterone and kinases. J Endocrinol 2015; 226:M13-27. [PMID: 26034071 DOI: 10.1530/joe-15-0078] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2015] [Indexed: 12/22/2022]
Abstract
The corticosterone (CORT) level changes along the circadian rhythm. Hippocampus is sensitive to CORT, since glucocorticoid receptors are highly expressed. In rat hippocampus fixed in a living state every 3 h, we found that the dendritic spine density of CA1 pyramidal neurons increased upon waking (within 3 h), as compared with the spine density in the sleep state. Particularly, the large-head spines increased. The observed change in the spine density may be due to the change in the hippocampal CORT level, since the CORT level at awake state (∼30 nM) in cerebrospinal fluid was higher than that at sleep state (∼3 nM), as observed from our earlier study. In adrenalectomized (ADX) rats, such a wake-induced increase of the spine density disappeared. S.c. administration of CORT into ADX rats rescued the decreased spine density. By using isolated hippocampal slices, we found that the application of 30 nM CORT increased the spine density within 1 h and that the spine increase was mediated via PKA, PKC, ERK MAPK, and LIMK signaling pathways. These findings suggest that the moderately rapid increase of the spine density on waking might mainly be caused by the CORT-driven kinase networks.
Collapse
Affiliation(s)
- Muneki Ikeda
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Yasushi Hojo
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Yoshimasa Komatsuzaki
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Masahiro Okamoto
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Asami Kato
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Taishi Takeda
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Suguru Kawato
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| |
Collapse
|
29
|
Matsuo H, Iwamoto A, Otsuka T, Hishida Y, Akiduki S, Aoki M, Furuse M, Yasuo S. Effects of time ofl-ornithine administration on the diurnal rhythms of plasma growth hormone, melatonin, and corticosterone levels in mice. Chronobiol Int 2014; 32:225-34. [DOI: 10.3109/07420528.2014.965312] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Disrupted daily light-dark cycles induce physical inactivity and enhance weight gain in mice depending on dietary fat intake. Neuroreport 2014; 25:865-869. [PMID: 24893203 DOI: 10.1097/wnr.0000000000000202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We evaluated associations between obesity induced by a high-fat diet (HFD) and the environmental light-dark (LD) cycle that entrains the master circadian clock located in the suprachiasmatic nucleus of mammals. Mice were fed normal diet or HFD for 6 weeks in individual cages with running wheels under a normal 12 h light-12 h dark cycle (LD 12 : 12) or an ultradian 3 h light-3 h dark cycle (LD 3 : 3) that might perturb the central clock. Circadian behavioral rhythms in mice fed both diets were disrupted by light-induced direct suppression of the behavior (masking effect) under LD 3 : 3. The ultradian LD cycle reduced the total daily activity of wheel running and enhanced body weight gain in the mice fed the HFD. Secondary effects such as obesity are probably not associated with inactivity induced under these circumstances because wheel-running activity decreased markedly within a few days of transfer from LD 12 : 12 to LD 3 : 3. Food consumption was significantly suppressed under LD 3 : 3 in mice fed the HFD. These findings suggest that the aberrant LD cycle induced physical inactivity and enhanced weight gain depending on dietary fat consumption. This might help to explain the higher incidence of obesity among shift workers.
Collapse
|
31
|
Miyazaki K, Itoh N, Yamamoto S, Higo-Yamamoto S, Nakakita Y, Kaneda H, Shigyo T, Oishi K. Dietary heat-killed Lactobacillus brevis SBC8803 promotes voluntary wheel-running and affects sleep rhythms in mice. Life Sci 2014; 111:47-52. [PMID: 25058921 DOI: 10.1016/j.lfs.2014.07.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/12/2014] [Accepted: 07/11/2014] [Indexed: 01/03/2023]
Abstract
AIMS We previously reported that heat-killed Lactobacillus brevis SBC8803 enhances appetite via changes in autonomic neurotransmission. Here we assessed whether a diet supplemented with heat-killed SBC8803 affects circadian locomotor rhythmicity and sleep architecture. MAIN METHODS AND KEY FINDINGS Daily total activity gradually increased in mice over 4 weeks and supplementation with heat-killed SBC8803 significantly intensified the increase, which reached saturation at 25 days. Electroencephalography revealed that SBC8803 supplementation significantly reduced the total amount of time spent in non-rapid eye movement (NREM) sleep and increased the amount of time spent being awake during the latter half of the nighttime, but tended to increase the total amount of time spent in NREM sleep during the daytime. Dietary supplementation with SBC8803 can extend the duration of activity during the nighttime and of sleep during the daytime. Daily voluntary wheel-running and sleep rhythmicity become intensified when heat-killed SBC8803 is added to the diet. SIGNIFICANCE Dietary heat-killed SBC8803 can modulate circadian locomotion and sleep rhythms, which might benefit individuals with circadian rhythms that have been disrupted by stress or ageing.
Collapse
Affiliation(s)
- Koyomi Miyazaki
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan.
| | - Nanako Itoh
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Saori Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Sayaka Higo-Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Yasukazu Nakakita
- Frontier Laboratories of Value Creation, Sapporo Breweries Ltd., Yaizu 325-0013, Japan
| | - Hirotaka Kaneda
- Corporate Planning Department, Sapporo Holdings Ltd., Tokyo 150-8522, Japan
| | - Tatsuro Shigyo
- Frontier Laboratories of Value Creation, Sapporo Breweries Ltd., Yaizu 325-0013, Japan
| | - Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan; Department of Medical Genome Science, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8561, Japan; Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan
| |
Collapse
|
32
|
Razzoli M, Karsten C, Yoder JM, Bartolomucci A, Engeland WC. Chronic subordination stress phase advances adrenal and anterior pituitary clock gene rhythms. Am J Physiol Regul Integr Comp Physiol 2014; 307:R198-205. [PMID: 24829500 PMCID: PMC4101617 DOI: 10.1152/ajpregu.00101.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/11/2014] [Indexed: 12/24/2022]
Abstract
Circadian rhythms in glucocorticoids are the product of interactions between the hypothalamic-pituitary-adrenal (HPA) axis and the mammalian clock gene system. The adrenal clock can generate the glucocorticoid rhythm that in turn synchronizes other peripheral clocks to maintain homeostasis. Stress acutely activates and chronically upregulates the HPA axis, suggesting that the adrenal clock could be modulated by stress. However, there is no direct evidence that stress affects the adrenal clock rhythm. We tested the hypothesis that a model of chronic subordination stress (CSS) that has a major impact on HPA axis regulation, metabolism, and emotional behavior alters adrenal and pituitary clock gene rhythms. Clock gene rhythms were assessed using mPER2::Luciferase (PER2Luc) knockin mice in which in vitro bioluminescence rhythms reflect the Per2 clock gene expression. PER2Luc mice that experienced CSS for 2 wk showed positive energy balance reflected by increased body weight and food intake. Additionally, CSS phase advanced the adrenal (∼2 h) and the pituitary (∼1 h) PER2Luc rhythm compared with control mice. The activity rhythm was not affected. The adrenal clock phase shift was associated with increased feed conversion efficiency, suggesting that the metabolic phenotype in CSS mice may be related to altered adrenal clock rhythmicity. Interestingly, a single subordination experience followed by 8 h sensory housing also phase advanced the adrenal, but not the pituitary, PER2Luc rhythm. Overall, these data demonstrate a stress-induced phase shift in a peripheral clock gene rhythm and differential stress sensitivity of two peripheral clocks within the HPA axis, suggesting a link between clock desynchrony and individual vulnerability to stress.
Collapse
Affiliation(s)
- Maria Razzoli
- Departments of Integrative Biology and Physiology and
| | - Carley Karsten
- Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - J Marina Yoder
- Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | | | | |
Collapse
|
33
|
Oishi K, Yamamoto S, Itoh N, Miyazaki K, Nemoto T, Nakakita Y, Kaneda H. Disruption of behavioral circadian rhythms induced by psychophysiological stress affects plasma free amino acid profiles without affecting peripheral clock gene expression in mice. Biochem Biophys Res Commun 2014; 450:880-4. [PMID: 24971530 DOI: 10.1016/j.bbrc.2014.06.083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 06/17/2014] [Indexed: 11/18/2022]
Abstract
Disordered circadian rhythms are associated with various psychiatric conditions and metabolic diseases. We recently established a mouse model of a psychophysiological stress-induced chronic sleep disorder (CSD) characterized by reduced amplitude of circadian wheel-running activity and sleep-wake cycles, sleep fragmentation and hyperphagia. Here, we evaluate day-night fluctuations in plasma concentrations of free amino acids (FAA), appetite hormones and prolactin as well as the hepatic expression of circadian clock-related genes in mice with CSD (CSD mice). Nocturnal increases in wheel-running activity and circadian rhythms of plasma prolactin concentrations were significantly disrupted in CSD mice. Hyperphagia with a decreased leptin/ghrelin ratio was found in CSD mice. Day-night fluctuations in plasma FAA contents were severely disrupted without affecting total FAA levels in CSD mice. Nocturnal increases in branched-chain amino acids such as Ile, Leu, and Val were further augmented in CSD mice, while daytime increases in Gly, Ala, Ser, Thr, Lys, Arg, His, Tyr, Met, Cys, Glu, and Asn were significantly attenuated. Importantly, the circadian expression of hepatic clock genes was completely unaffected in CSD mice. These findings suggest that circadian clock gene expression does not always reflect disordered behavior and sleep rhythms and that plasma FFA profiles could serve as a potential biomarker of circadian rhythm disorders.
Collapse
Affiliation(s)
- Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan; Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan.
| | - Saori Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Nanako Itoh
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Koyomi Miyazaki
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Tadashi Nemoto
- Bio-Interface Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Yasukazu Nakakita
- Frontier Laboratories of Value Creation, Sapporo Breweries Ltd., Yaizu, Shizuoka, Japan
| | - Hirotaka Kaneda
- Corporate Planning Department, Sapporo Holdings Ltd., Shibuya-ku, Tokyo, Japan
| |
Collapse
|