内 容

セッション/プレナリーレクチャー

S: Session, PL: Plenary Lecture
S 1-1
Atsushi Miyawaki (RIKEN)
Cruising inside Cells
S 1-2
Samie R. Jeffrey (Cornell University)
Imaging RNA and RNA biology using RNA mimics of green fluorescent protein
S 1-3
Na Ji (Janelia Farm, HHMI)
From star to neuron-adaptive optical microscopy for deep brain imaging
S 2-1
Tomomi Nemoto (Hokkaido University)
Improvement of multi-photon microscopy by utilizing new laser technologies
S 2-2
Valentin Nägerl(Université de Bordeaux)
Nanoscale imaging of neural plasticity by STED microscopy
S 2-3
Masanori Matsuzaki (National Institute for Basic Biology)
Dynamics of cortical ensembles during motor learning
PL 1
Eric Betzig (HHMI)
Imaging Life at High Spatiotemporal Resolution
S 3-1
Takaharu Okada (RIKEN)
Cellular dynamics of adaptive immune responses
S 3-2
Guy Shakhar (Weizman Institute)
Understanding the obstacles to CTL tumor elimination through 2-photon imaging
S 3-3
Kenji Kabashima (Kyoto University)
S 3-4
Tatsuo Kinashi (Kansai Medical University)
Visualization of thymocyte trafficking and selection processes: the importance of Rap1 signaling and integrins
PL 2
Lihong V. Wang (Washington University)
Photoacoustic Tomography: Ultrasonically Beating Optical Diffusion and Diffraction
S 4-1
Takeshi Imamura (Ehime University)
Intravital cancer imaging by multi-photon microscopy
S 4-2
Charles P. Lin (Harvard University)
Live imaging of the mouse bone marrow by intravital microscopy
S 4-3
Michio Tomura (Kyoto University)
Visualization of trafficking and presentation of tumor antigen by dendritic cells
S 5-1
Masaru Ishii (Osaka University)
Intravital multiphoton imaging revealing immune cell dynamics in bone destruction in vivo
S 5-2
João P. Pereira (Yale University)
Oxysterols and EBI2 guide osteoclast precursors to bone surfaces and regulate bone mass
S 5-3
Takashi Nagasawa (Kyoto University)
The microenvironmental niches for hematopoietic stem and progenitor cells in bone marrow
S 6-1
Michiyuki Matsuda (Kyoto University)
Transgenic mouse lines with heritable and functional FRET biosensors
S 6-2
Won Do Heo (KAIST)
Optogenetic Control of Cell Signaling in Mammalian Cells
S 6-3
Michael Z. Lin (Stanford University)
New twists and turns - engineering electrosensory and photoregulatory functions into fluorescent proteins
S 6-4
Takeaki Ozawa (Tokyo University)
Luminescent sensors for single cell analysis
S 7-1
Shigetomo Fukuhara (NCVC)
Unveiling the cellular and molecular mechanism of vascular development by fluorescence-based bio-imaging in zebrafish
S 7-2
Suk-Won Jin (Yale University)
Visualizing Endothelial Cells During Zebrafish Development
S 7-3
Seiji Takashima (Osaka University)
ATP concentration imaging in beating heart
S 7-4
Satoshi Nishimura (Jichi Medical University)
Fluorescent two photon imaging in cardiovascular diseases

ポスター発表

P:Poster
P 1
Takahiro Adachi (Tokyo Medical and Dental University)
In vivo imaging of calcium signaling in B cells of mice expressing the genetically encoded YC3.60 calcium indicator
P 2
Shinji Ihara (National Institute of Genetics)
In vivo imaging of basement membranes reveals non-canonical pathway of GPI anchor modifier enzyme in C. elegans
P 3
Yoichi Ueta (University of Occupational and Environmental Health)
Simultaneous visualization of neurohypophysial hormones and neuronal activity in the hypothalamic neurosecretory neurons by multi-dimensional fluorescent imaging and optogenetics
P 4
Mikiko Ohno (Kyoto University)
Nardilysin controls circulatory dynamics through regulating sympathetic nervous system
P 5
Yasushi Okada (Quantitative Biology Center, RIKEN)
Direct measurement of the binding rate constant of kinesin to microtubules in living cells
P 6
Mako Kamiya (The University of Tokyo)
A spontaneously blinking fluorophore based on intramolecular spirocyclization for live-cell super-resolution imaging
P 7
Hisato Kondoh (Kyoto Sangyo University)
Nuclear dynamics of Sox2 that depends on interactions with partner transcription factors
P 8
Naoaki Saito (Kobe University)
Fluorescence imaging analysis of cerebellar granule neurons in Rac knockout mice manifesting balance impairment
P 9
Moritoshi Sato (The University of Tokyo)
Exploring molecular processes at the plasma membrane
P 10
Yuhkoh Satouh (Osaka University)
Live imaging of mammalian fertilization
P 11
Takeo Saneyoshi (Brain Sci. Inst., RIKEN)
Conversion of a transient Ca2+ signaling into a persistent structural modification of dendritic spines by CaMKII/TIAM complex formation during synaptic plasticity
P 12
Kyoko Shirakabe (Keio University)
Real-time imaging of ectodomain shedding using fluorescence signal
P 13
Takamasa Mizoguchi (Chiba University)
The actin dynamics live imaging reveals that Mib1 controls appropriate actin rearrangements and coordinates corrective cell migration
P 14
Yoshihiro Miwa (University of Tsukuba)
NIR non-invasive fluorescence imaging applied for mouse models
P 15
Katsuyuki Yui (Nagasaki University)
Imaging of CD8+ T cells protecting against liver-stage infection with malaria parasites
P 16
Tadashi Yokosuka (RIKEN, Center for Integrative Medical Sciences)
Subcellular imaging of the E3 ubiquitin ligases, c-Cbl and Cbl-b, to control T cell signals by clustering of ubiquitin at TCR microclusters
P 17
Kazuo Kitamura (The University of Tokyo)
Experience-Dependent Clustering of Sensory Synaptic Inputs in the Mouse Barrel Cortex
P 18
Ryosuke Enoki (Hokkaido University)
Multi-functional analysis of circadian rhythms in the mammalian master clock
P 19
Kazuki Horikawa (University of Tokushima)
Fluorescent biosensors for inflammatory response
P 20
Yoshihiro Ueda (Kansai Medical University)
Visualization of Rap1 activation during thymocyte development within the thymic tissues by 2-photon microscopy
P 21
Nobuhiko Miyasaka (RIKEN, Brain Science Institute)
Genetic Visualization of Neurons at Single-Cell Resolution Reveals a Comprehensive Axon Projection Map of the Secondary Olfactory Pathway in Zebrafish
P 22
Ryu John Iwatate (The University of Tokyo)
Asymmetric rhodamine-based fluorescence probes for multi-color in vivo imaging
P 23
Yusuke Nasu (The University of Tokyo)
Nanoscale Characterization of Protein Clusters Responsible for Cell Death Using Super-resolution Microscopy
P 24
Kouichirou Iijima (Hokkaido University)
Improved methods of open-skull surgery for in vivo imaging using biocompatible materials
P 25
Rika Suzuki (Keio University)
Cytoskeletal Dynamics Changes Intracellular ATP level
P 26
Ayumi Nagasawa (Institute of Molecular and Cellular Biosciences)
ERK signaling activity is required for tip cells to form second branch vessel in zebrafish angiogenesis
P 27
Takashi Saitou (Ehime University Hospital)
Assimilation of Live Imaging and Mathematical Modeling toward Understanding the Regulatory Mode of G1/S Transition Wave in Zebrafish Developing Notochord
P 28
Kiichi Kaminaga (Ibaraki University)
Visualization of cell cycle arrest by X-irradiation in single HeLa cells using Fluorescent Ubiquitination-based Cell Cycle Indicator
P 29
Yusuke Ohba (Hokkaido University)
Development of FRET-based biosensors for detection of CML cells resistant to molecular target drugs
P 30
Yoichiro Fujioka (Hokkaido University)
Ca2+ signaling mediated influenza virus internalization into host cells
P 31
Naoyoshi Koike (Keio University)
Cell cycle imaging in irradiated murine glioma stem cells
P 32
Takuto Kawahata (Kagoshima University)
Spatio-temporal assessment of nucleolar stress response by a novel visualized reporter system and its application of antitumor drugs discovery
P 33
Tadahiro Iimura (Ehime University)
Morphometrical fluorescence imaging of skeletal homeostasis
P 34
Yusuke Oshima (Ehime University)
Development of wide-field of view 2p-excited light-sheet microscopy
P 35
Haruna Takeda (Kanazawa Medical University)
In vivo imaging using a two-photon microscopy in the mouse intestine
P 36
Kenji Kamimoto (Tokyo University)
Multi-dimensional cell fate tracing reveals the existence of a potential progenitor cell population in the biliary tree of the regenerating mouse liver
P 37
Yasuhiro Tanaka (National Institute for Basic Biology)
Two distinct layer-specific dynamics of cortical ensembles during learning of a motor task
P 38
Toru Hiratsuka (Kyoto University)
Waves of localized extracellular signal-regulated kinase activation regulate cell proliferation in mouse epidermis
P 39
Wakako Sadaie (Kyoto University)
Quantitative measurements of protein-protein interaction in living cells with fluorescence cross-correlation spectroscopy
P 40
Atsuro Sakurai (Kyoto University)
Visualization of Wnt/β-catenin signaling activity in intestinal organoids
P 41
Noriyuki Kawabata (Kyoto University)
Constitutively active mutation of Hras can be inhibited by cell contact interface

ランチョンセミナー

1日目:1月27日(火)11:45〜12:45

共催:オリンパス株式会社

ライブ・深部を両立する先進超解像テクノロジー
“Olympus Super Resolution (OSR) ”

超解像ライブイメージングと1分子計測による細胞内物質輸送の制御機構
Super-resolution live imaging and single molecule measurements to dissect the regulatory mechanism of intracellular transport

岡田 康志(理化学研究所 生命システム研究センター)
Yasushi Okada (Quantitative Biology Center, QBiC)

Truck transport is an essential logistics in our society. Similarly, intracellular transport of organelles, proteins or mRNAs plays essential roles as the logistics for the various cellular functions. Most of the intracellular transport is supported by the microtubule-dependent molecular motors, such as kinesins and dynein. If you enlarge a motor molecule to the size of a truck, the cell would cover a larger area than Kyoto prefecture. We can drive our cars to the destination with the help of a map or GPS. Then, how can molecular motors reach their targets within the cell? We have found that some kinesins specifically move along only a small subset of intracellular microtubules. Since such microtubules are often found enriched toward the correct destinations for those kinesins, we surmise that microtubules are not passive substrates as the roads for the transport but they play important roles in the regulation of transport.

For the analyses of the functional differentiations of microtubules, it is essential to visualize single microtubules in living cells. However, it is often difficult due to the limited resolution of optical microscopes. Therefore, we have been trying various super-resolution microscope techniques for nearly 10 years. Although PALM and other super-resolution techniques were proven to be powerful enough to dissect the functional differentiations of microtubules, we found two limitations: slow frame rate and small view field.

We have collaborated with Olympus to overcome these limitations. In this luncheon seminar, we would like to introduce our solutions to overcome these issues.

2日目:1月28日(水)11:45〜12:45

共催:オリンパス株式会社

次世代再生医療研究に向けた多次元イメージングシステム
“Olympus Multi-photon Microscope”
─器官・臓器形成のライブ立体観察─

iPS細胞を用いた機能的なヒト肝臓の創出
Generation of a vascularized human liver from an iPSC-derived organ bud transplant

谷口 英樹(横浜市立大学大学院医学研究科 臓器再生医学)
Hideki Taniguchi (Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine)

A critical shortage of donor organs for treating end-stage organ failure highlights the urgent need for generating organs from human induced pluripotent stem cells (iPSCs).
Despite many reports describing func- tional cell differentiation, no studies have succeeded in generating a three-dimensional vascularized organ such as liver.
Here we show the generation of vascularized and functional human liver from human iPSCs by transplantation of liver buds created in vitro (iPSC-LBs).

幹細胞からの自己組織的な立体神経組織形成とその原理
Self-organized formation of complex tissues from stem cells

永樂 元次(理化学研究所、多細胞システム形成研究センター)
Mototsugu Eiraku (RIKEN, Center for the Developmental Biology)

In vitro generation of a functional organ with complex structures is a major challenge of cell biology. In recent decade, there has been an increasing interest in 3D tissue formation from stem cells. For example, stem cell researchers have demonstrated that embryonic stem (ES) cells or induced pluripotent stem (iPS) cells could be steered to differentiate into 3D tissues, such as brain, retina, inner ear, liver, kidney and stomach. In addition, it was also reported that a single-cell culture of intestinal stem cells formed crypto-like organoids that self-renew and produce cells such as Paneth cells and enterocytes. These 3D stem cell culture technologies are expected to contribute to a future regenerative medicine and drug discovery. Especially, one of the most important applications of that is production of in vitro models for drug screening and studying mechanisms that cause diseases using disease-specific iPS cells.

We previously demonstrated that mouse ES cells self-formed apico-basally polarized cortical tissues using an efficient 3D aggregation culture (Eiraku et al., Cell Stem Cell, 2008). We also reported about self-organized formation of optic cup (retinal primordia) and a stratified neural retina from mouse ES cells (Eiraku et al., Nature, 2011). Transplantation experiments indicated that the mouse ES/iPS cell-derived 3D retinal tissue could develop a structured outer nuclear layer with outer segments even in an advanced retinal degeneration model that lacked photoreceptors (Assawachananont J et al., Stem Cell Reports, 2014). Using two-photon live imaging analysis and measurement of mechanical properties in developing retinal tissue, we revealed that the optic-cup morphogenesis can spontaneously occur in a three-dimensional stem-cell culture even in the absence of external forces, and that retinal progenitors have a latent intrinsic order to generate the optic-cup structure. Based on this self-organizing phenomenon, we proposed the “relaxationexpansion” model to interpret the tissue dynamics that enable the spontaneous invagination of the neural retina. This model involves three consecutive local rules (relaxation, apical constriction, and expansion), and its computer simulation recapitulates the optic-cup morphogenesis in silico.

In addition, we have attempted to apply these mouse ES cell culture to human ES cells. As a result, we clearly demonstrated that the ES cell culture method for mouse optic cup formation was applicable to human ES cells and, as seen in the case of mouse ES cells, human ES cell-derived retinal tissue spontaneously formed optic cup structure with the peripheral margin, called the ciliary margin (Nakano et al., Cell Stem Cell, 2012; Kuwahara et al., in press). The neural retina in human ES cell culture is thick and spontaneously curves in an apically convex manner, which is not seen in mouse ES cell culture. We also demonstrated that human ES cell-derived cortical neuroepithleium self-formed a multilayered structure including three neuronal zones and three progenitor zones (Kadoshima et al., PNAS, 2013).

Thus, we have mainly focused on the 3D tissue generations from mouse and human pluripotent stem cells and endeavored to understand the molecular and cellular mechanisms underlying selforganization phenomena in neural development. I will talk about our researches about in vitro tissue generations and a future direction of an in vitro histogenesis.