KU-UW Alliance Project ― International Joint Symposioum 抄録
(Talk 1) 9:45Robert Balderasベクトン・ディッキンソン株式会社![]() Defining Deep Biology with High Dimensional Platform Technologies. "Spatial protein localization and detection at the single cell level"Flow cytometry has long been a cornerstone technology in cell biology, allowing for the quantitative analysis of cell populations based on specific biological markers. In addition, flow cytometry also provides robust data regarding cell size, granularity, and fluorescence intensity with high throughput. However, it often lacks the spatial context of cellular morphology and complex structural features. Recent advancements in 2-dimensional electronic imaging technology have enabled the integration of imaging capabilities with traditional flow cytometry, creating a powerful hybrid approach. This new technology platform allows for the simultaneous acquisition of quantitative and qualitative data, enhancing the interpretation of cellular characteristics based on a series of imaging-based parameters. Thus, we can now visualize individual cells in a flow system, capturing detailed morphologic information alongside traditional readouts. Imaging flow cytometry also facilitates the study of cellular interactions, subcellular localization of proteins, detection of cell morphology and alterations in cellular architecture in response to various stimuli. This presentation will demonstrate the use of a series of defined imaging parameters in a variety of primary and cell line model systems. |
(Talk 2) 10:35宮西 正憲神戸大学大学院医学研究科細胞医科学分野![]() Toward Visualizing the Cellular Heterogeneity of Bone MarrowThe hematopoietic system comprises the largest cellular population in the human body, with bone marrow as the primary site of lifelong blood-cell production. Unlike solid organs with defined architecture, bone marrow lacks an organized scaffold, making its cellular organization difficult to interpret through classical histology. This structural fluidity has required distinct analytical strategies. Flow cytometry has therefore been central, as its population-level resolution suits the dynamic and heterogeneous nature of hematopoiesis. However, the limited parameter capacity of conventional multicolor cytometers has hindered comprehensive characterization of the hematopoietic hierarchy, particularly the rare, undifferentiated hematopoietic stem cell (HSC) compartment. Long-term HSCs (LT-HSCs) reside at the top of this hierarchy, generating all mature blood lineages. Although panoramic single-cell technologies have broadened our understanding, high-resolution visualization of rare undifferentiated human populations remains difficult. Mouse systems have enabled functional dissection of HSC heterogeneity, but equivalent progress in humans has been limited. Here, we introduce an optimized analytical approach that enables high-resolution visualization of previously inaccessible undifferentiated regions within human hematopoiesis. By integrating multiple data modalities, this strategy also opens the door to in silico identification of molecular features that may define human LT-HSCs. In the presentation, I will share how this framework reveals aspects of the hematopoietic system—long hidden due to its structural invisibility—in a way that provides new conceptual and experimental possibilities. |
(Talk 3) 10:50Claudia Morenoワシントン大学![]() Heart rate adaptability: from ion channels to heart dysfunctionMy research centers on the cardiac pacemaker, a specialized tissue near the right atrium responsible for generating the rhythmic electrical signals that initiate each heartbeat. Despite its discovery over a century ago, major gaps remain in our understanding of how this oscillator functions, adapts across species, and becomes dysregulated with age. My team uses an integrative approach to investigate pacemaker function from the ion channels that generate the electrical activity all the way to in-vivo physiology. We explore pacemaker adaptability across distinct timescales: from short-term adjustments that occur within seconds—such as the rapid rise in heart rate during stress—to long-term evolutionary adaptations observed in species with extreme heart rates. |
(Talk 4) 11:10Yasemin Sancakワシントン大学Novel Biochemical Methods to Investigate Mitochondria, Metabolism and SignalingComing Soon |
(Talk 5) 13:00佐藤 康彦カールツアイス株式会社![]() Introducing light-field 4D microscopy technology that realizes one volume per one snapZEISS has introduced the LSM 910 and LSM 990 confocal microscopes, integrating refined spectral multiplexing technology with Airyscan-based super-resolution imaging. The newly implemented Lightfield 4D technology enables single-snap volumetric acquisition by reconstructing three-dimensional images from spatial fluorescence signal distributions. This innovation addresses a major limitation of conventional confocal microscopy by allowing truly simultaneous volumetric imaging. In this presentation, we introduce the principles and performance of Lightfield 4D technology and discuss its potential to advance high-speed, high-resolution three-dimensional imaging in life science research. |
(Talk 6) 13:20菊田 順一神戸大学大学院医学研究科免疫学分野![]() Dissecting inflammatory dynamics and their regulation through intravital imagingInflammation is a fundamental biological response essential for host defense against pathogens and the facilitation of tissue repair. However, dysregulated inflammatory responses underlie the pathogenesis of a wide range of diseases, including autoimmune disorders, chronic inflammatory conditions, and malignancies. Recent advances in immunology have led to the development of therapeutic strategies that modulate immune function, such as immune checkpoint inhibitors and molecularly targeted agents. Nevertheless, many inflammatory diseases still lack curative treatments, and their underlying mechanisms remain incompletely understood. To address these challenges, we have established intravital imaging techniques that enable the high-resolution, spatiotemporal visualization of immune cell dynamics in living animals. Through this approach, we have identified previously unrecognized regulatory mechanisms of immune responses, including the role of sympathetic nerve fibers innervating the bone marrow in orchestrating immune cell mobilization during inflammation. Furthermore, we have demonstrated that receptor activator of nuclear factor κB ligand (RANKL), a key osteoclastogenic factor, regulates vascular permeability within the bone marrow microenvironment. More recently, we have extended our imaging systems to multiple organs and developed in vivo models of fibrotic diseases, such as pulmonary fibrosis and systemic sclerosis, thereby enabling the real-time tracking of pathogenic macrophage subsets involved in tissue fibrosis. In this symposium, I will highlight the utility of intravital imaging in advancing our understanding of inflammatory processes and discuss its promise in elucidating disease mechanisms and guiding the development of innovative therapeutic strategies. |
(Talk 7) 13:35Smita Yadavワシントン大学![]() Unraveling Mechanisms Underlying Neurodevelopmental Disorders using Cortical OrganoidsNeurodevelopmental disorders currently affect more than 4% of children worldwide. Genetic studies of brain developmental disorders have brought to light important candidates including protein kinases that remain largely understudied despite being highly druggable therapeutic targets. To illuminate how the human kinome contributes to brain development and neurological disorders, the Yadav laboratory integrates a wide variety of techniques in molecular neurobiology, stem cell technology, chemical-genetics, proteomics, and microscopy. Our recent work has defined the function of one such protein kinase, TAOK1, mutations in which cause neurodevelopmental delay, autism, speech deficits, and macrocephaly. I will present our findings that established the function of TAOK1 as a membrane remodeling kinase, and our ongoing work towards understanding its role in brain disorders using proteomic and scRNA seq analyses of human cortical brain organoids that recapitulate the TAOK1 neuropathology. |
(Talk 8) 13:55Marta Sodenワシントン大学![]() Approaches for investigating neural circuits that govern motivated behaviorUnderstanding how neural circuits control specific behaviors necessitates a multi-layered approach, as we investigate how cell-type specific gene expression, neuronal excitability, anatomical connectivity and responsivity to stimuli intersect to drive circuit output. Recent advances in gene mutagenesis, transcriptomic, and in vivo imaging technologies have enabled the dissection of neural circuits with unprecedented precision, allowing us to isolate the effects of co-released neuropeptides and neurotransmitters and identify specific postsynaptic target neurons. We recently identified a novel peptidergic neural circuit from the periaqueductal gray to the ventral tegmental area that simultaneously evokes threat-response and reward-related behaviors in mice. Using this example, I will discuss how we have developed and implemented an approach to investigating neural circuits that incorporates CRISPR mutagenesis, activity-dependent transcriptomics, optogenetics, whole-cell patch clamp, and in vivo sensors in order to uncover circuit structure and function. I will highlight the advantages of this multifaceted approach, discuss current challenges, and outline technologies in development that will further advance our ability to understand specific circuit control of behavior. |
(Talk 9) 15:05佐藤 勇次10x Genomics Japan 株式会社![]() Unparalleled single cell spatial discoveryPowering innovation in labs around the world, the Xenium platform is a complete solution that combines ultimate versatility with easy, streamlined workflows. We invite you to see for yourself at the symposium, where you can explore how the Xenium platform gives you:
Join us to learn how our technologies can help you sharpen your research with single cell multiomics, explore tissue and tumor architecture with spatial profiling and see further than ever before with high-throughput in situ transcriptomics at subcellular resolution. With this comprehensive toolkit, you can combine these multidimensional analyses into a more holistic approach to developing better therapeutics and predicting patient outcomes. |
(Talk 10) 15:25堀江 真史神戸大学大学院医学研究科分子病理学分野![]() Spatial Mapping of Small Cell Lung Carcinoma: Intratumoral Diversity and Discovery of a New Macrophage SubsetSmall cell lung cancer (SCLC) is a highly aggressive neuroendocrine carcinoma with a poor prognosis and limited treatment options. Recent advances in molecular profiling have revealed inter-tumoral heterogeneity within SCLC, leading to a refined classification based on key transcriptional regulators such as ASCL1, NEUROD1, POU2F3, and YAP1. Although this framework has opened new avenues for targeted therapy and biomarker development, the extent of intratumoral heterogeneity—as well as the contribution of the tumor immune microenvironment, particularly tumor-associated macrophages (TAMs)—remains incompletely understood. The emergence of novel technologies, including single-cell RNA sequencing and spatial transcriptomics now enables deeper interrogation of tumor biology, lineage plasticity, and the tumor microenvironment. Here, we performed spatial multi-omics profiling of 18 surgically resected lung cancer samples, including 16 SCLC cases. Transcriptomic subtyping revealed marked heterogeneity, with multiple coexisting cellular states present within individual tumors. Integration of trajectory analysis and copy-number estimation highlighted potential developmental pathways underlying this diversity and identified candidate regulatory mechanisms governing cellular plasticity. We further identified a previously unrecognized MMP12-positive TAM population enriched at the tumor interface and spatially associated with necrotic and hypoxic regions. In vitro direct co-culture under hypoxic conditions confirmed that MMP12 induction in TAMs is both hypoxia-dependent and contact-dependent. Together, these findings delineate the complex spatial architecture of SCLC, uncover extensive intratumoral heterogeneity, and reveal a novel TAM subset that may contribute to treatment resistance and represent a potential therapeutic target. |
(Talk 11) 15:40Nobuhiko Hamazakiワシントン大学![]() Reconstruction of human post-gastrulation development in vitroUnderstanding the mechanistic underpinnings of human post-gastrulation development is crucial, as disruptions during this period often result in severe developmental diseases such as neural tube defects, anencephaly, and cardiac malformations. Despite its importance, research into this stage is significantly restricted due to ethical and technical barriers associated with accessing and studying human in vivo embryos. Consequently, researchers have sought alternative approaches, with human pluripotent stem cell (hPSC)-derived embryo models emerging as a promising solution. As part of this effort, we have established two distinct in vitro models, the RA-gastruloid and the AP-gastruloid, which successfully recapitulate broad aspects of human embryogenesis. This presentation will highlight recent progress using these models to elucidate the complex regulatory mechanisms governing early human development and discuss the potential of these systems to provide foundational insights into the etiology of developmental disorders. |
(Talk 12) 16:00David Schechnerワシントン大学TBDComing Soon |
(Talk 13) 16:50千原 典夫神戸大学大学院医学研究科脳神経内科学分野![]() Engineering Immune Cells for Neuroprotection: Lessons from Multiple Sclerosis and Neuromyelitis OpticaIn line with the session theme “Designing the Future: Innovations in Cell Engineering,” this presentation will examine how rethinking immune cell function inside the central nervous system (CNS) can guide next-generation therapies for neuroinflammatory disease. Treatments for multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) have largely targeted peripheral lymphoid tissues, using B-cell–directed, anti-cytokine, and complement-inhibiting agents. These approaches have reduced relapse rates but only partially address progression and neurodegeneration. On the other hand, the roles of microglia, border-associated macrophages, and infiltrating lymphocytes within the brain and spinal cord remain less well defined. Emerging single-cell and spatial technologies, together with advanced imaging, are now revealing immune cell states and circuits that shape synapses, myelin repair, and neuronal survival in the CNS milieu. We will discuss how mapping and functionally testing these CNS-resident and infiltrating immune populations inform new strategies for cell engineering and cell-targeted intervention. Examples include approaches to reprogram microglia toward protective phenotypes, to tune myeloid and lymphoid trafficking across CNS barriers, and to design immune cells that act as local sensors and effectors of neuroinflammation. By connecting clinical experience in MS and NMOSD with a deeper understanding of CNS immune ecology, this talk will outline how targeting the right cell in the right compartment may yield more effective and safer treatments—not only for classical neuroimmune diseases but also for a broader range of neurological disorders, including neurodegenerative and psychiatric disorders. |
(Talk 14) 17:05Sam A. Goldenワシントン大学Preclinical tools for bridging brain, body, and behaviorComing Soon |
(Talk 15) 17:25Andre Berndtワシントン大学![]() Accelerating Fluorescent Sensor Engineering for Neuroscience with Machine Learning and High-Throughput PlatformsGenetically encoded fluorescent sensors have become indispensable tools for visualizing neural activity and biochemical signaling in living systems. My laboratory integrates machine learning, structure-guided design, and high-throughput screening to develop next-generation fluorescent sensors optimized for in vivo imaging. Using these combined strategies, we engineer and refine sensors to achieve improved brightness, kinetics, and spectral properties, which are suitable for multiplexed recording. Our work spans multiple molecular targets and modalities, including calcium, reactive oxygen species (ROS), steroid hormones, and endogenous opioids. In addition, we are developing sensors with fluorescence lifetime readouts, which provide quantitative measurements that are less sensitive to artifacts such as expression level or motion. The integration of machine learning models with experimental screening enables rapid identification of functional variants from large mutational libraries, thereby accelerating sensor design and improving sensor performance. The combination of predictive computational models and automated screening platforms provides a scalable framework for engineering sensors that can visualize complex signaling processes across the brain and body. |











