Main Menu (Mobile)- Block

Main Menu - Block

custom | custom

Search Results

filters_region_cap | custom

Filter

facetapi-Q2b17qCsTdECvJIqZJgYMaGsr8vANl1n | block

Associated Lab

facetapi-W9JlIB1X0bjs93n1Alu3wHJQTTgDCBGe | block
facetapi-61yz1V0li8B1bixrCWxdAe2aYiEXdhd0 | block
facetapi-PV5lg7xuz68EAY8eakJzrcmwtdGEnxR0 | block
general_search_page-panel_pane_1 | views_panes

2647 Janelia Publications

Showing 1-10 of 2647 results
04/23/25 | Whole-body simulation of realistic fruit fly locomotion with deep reinforcement learning
Roman Vaxenburg , Igor Siwanowicz , Josh Merel , Alice A Robie , Carmen Morrow , Guido Novati , Zinovia Stefanidi , Gwyneth M Card , Michael B Reiser , Matthew M Botvinick , Kristin M Branson , Yuval Tassa , Srinivas C Turaga
Nature. 2025 Apr 23:. doi: 10.1038/s41586-025-09029-4

The body of an animal influences how its nervous system generates behavior1. Accurately modeling the neural control of sensorimotor behavior requires an anatomically detailed biomechanical representation of the body. Here, we introduce a whole-body model of the fruit fly Drosophila melanogaster in a physics simulator. Designed as a general-purpose framework, our model enables the simulation of diverse fly behaviors, including both terrestrial and aerial locomotion. We validate its versatility by replicating realistic walking and flight behaviors. To support these behaviors, we develop new phenomenological models for fluid and adhesion forces. Using data-driven, end-to-end reinforcement learning we train neural network controllers capable of generating naturalistic locomotion along complex trajectories in response to high-level steering commands. Additionally, we show the use of visual sensors and hierarchical motor control, training a high-level controller to reuse a pre-trained low-level flight controller to perform visually guided flight tasks. Our model serves as an open-source platform for studying the neural control of sensorimotor behavior in an embodied context.

 

Preprint: www.biorxiv.org/content/early/2024/03/14/2024.03.11.584515

View Publication Page
04/22/25 | TEMI: Tissue Expansion Mass Spectrometry Imaging
Zhang H, Ding L, Hu A, Shi X, Huang P, Lu H, Tillberg PW, Wang MC, Li L
Nat Methods. 2025 Apr 22:. doi: 10.1101/2025.02.22.639343

The spatial distribution of diverse biomolecules in multicellular organisms is essential for their physiological functions. High-throughput in situ mapping of biomolecules is crucial for both basic and medical research, and requires high scanning speed, spatial resolution, and chemical sensitivity. Here, we developed a Tissue Expansion method compatible with matrix-assisted laser desorption/ionization Mass spectrometry Imaging (TEMI). TEMI reaches single-cell spatial resolution without sacrificing voxel throughput and enables the profiling of hundreds of biomolecules, including lipids, metabolites, peptides (proteins), and N-glycans. Using TEMI, we mapped the spatial distribution of biomolecules across various mammalian tissues and uncovered metabolic heterogeneity in tumors. TEMI can be easily adapted and broadly applied in biological and medical research, to advance spatial multi-omics profiling.

Preprint: 10.1101/2025.02.22.639343

View Publication Page
04/21/25 | Abstract 2420: Deep learning enables automated detection of circulating tumor cell-immune cell interactions with prognostic insights in cancer
Sun Y, Squires JR, Hoffmann A, Zhang Y, Minor A, Singh A, Scholten D, Mao C, Luo Y, Fang D, Gradishar WJ, Cristofanilli M, Stringer C, Liu H
Cancer Research. 2025 Apr 21;85:2420-2420. doi: 10.1158/1538-7445.AM2025-2420

Circulating tumor cells (CTCs) are critical biomarkers for predicting therapy response and survival in breast cancer patients. Multicellular CTC clusters exhibit enhanced metastatic potential, yet their detection and characterization are constrained by low frequency in blood samples and reliance on labor-intensive manual analysis. Advancing these methods could significantly improve prognostic evaluation and therapeutic strategies.Leveraging FDA-approved CellSearch technology and single-cell sequencing, we analyzed 2, 853 blood specimens, longitudinally collected from 1358 patients with advanced cancer (breast, prostate, etc) and other diseases. Integrating machine learning and deep learning tools, we developed a novel CTCpose platform to automate detection and analysis of CTCs, immune cells, and their interactions. Using artificial intelligence (AI)-driven image analysis, we extracted over 270 cellular and nuclear features including intensity, morphometry, fourier shape, gradient/edge, and haralick of cytokeratin, CD45, and DAPI expression patterns, enabling precise characterization of CTCs, white blood cells (WBCs), CTC clusters, and their interactions with immune cells (WBCs).The CTCpose platform enabled automated identification of CTCs, WBCs, homotypic CTC clusters, heterogenous CTC-WBC clusters, and immune cell clusters, providing comprehensive insights into cell morphology, biomarker expression, and spatial organization. These features correlated with patient survival, disease progression, and treatment response. Our findings highlight the clinical significance of CTC-immune cell interactions and dynamic alterations of CTCs (singles and clusters) and underscore their potential in stratifying patients into distinct risk categories.This study demonstrates the transformative potential of deep learning in overcoming limitations of traditional CTC detection methods and integrating imaging data with large cohorts of patient data. By automating and enhancing the analysis of CTC-immune cell interactions, we present a robust framework for developing predictive models with direct clinical relevance. This work opens avenues for personalized treatment strategies, underscoring the impact of AI in advancing precision oncology.Yuanfei Sun, Joshua R. Squires, Andrew Hoffmann, Youbin Zhang, Allegra Minor, Anmol Singh, David Scholten, Chengsheng Mao, Yuan Luo, Deyu Fang, William J. Gradishar, Massimo Cristofanilli, Carsen Stringer, Huiping Liu. Deep learning enables automated detection of circulating tumor cell-immune cell interactions with prognostic insights in cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2025; Part 1 (Regular Abstracts); 2025 Apr 25-30; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2025;85(8_Suppl_1):Abstract nr 2420.

View Publication Page
04/20/25 | FilaBuster: A Strategy for Rapid, Specific, and Spatiotemporally Controlled Intermediate Filament Disassembly
Moore AS, Krug T, Hansen SB, Ludlow AV, Grimm JB, Ayala AX, Plutkis SE, Wang N, Goldman RD, Medalia O, Lavis LD, Weitz DA, Lippincott-Schwartz J
bioRxiv. 2025 Apr 20:. doi: 10.1101/2025.04.20.649718

Intermediate filaments (IFs) play key roles in cellular mechanics, signaling, and organization, but tools for their rapid, selective disassembly remain limited. Here, we introduce FilaBuster, a photochemical approach for efficient and spatiotemporally controlled IF disassembly in living cells. FilaBuster uses a three-step strategy: (1) targeting HaloTag to IFs, (2) labeling with a covalent photosensitizer ligand, and (3) light-induced generation of localized reactive oxygen species to trigger filament disassembly. This modular strategy applies broadly across IF subtypes—including vimentin, GFAP, desmin, peripherin, and keratin 18—and is compatible with diverse dyes and imaging platforms. Using vimentin IFs as a model system, we establish a baseline implementation in which vimentin-HaloTag labeled with a photosensitizer HaloTag ligand triggers rapid and specific IF disassembly upon light activation. We then refine this approach by (i) expanding targeting strategies to include a vimentin nanobody-HaloTag fusion, (ii) broadening the range of effective photosensitizers, and (iii) optimizing irradiation parameters to enable precise spatial control over filament disassembly. Together, these findings position FilaBuster as a robust platform for acute, selective, and spatiotemporally precise disassembly of IF networks, enabling new investigations into their structural and functional roles in cell physiology and disease.

View Publication Page
04/09/25 | Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment.
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N
Cell Syst. 2025 Apr 09:101261. doi: 10.1016/j.cels.2025.101261

Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.

View Publication Page
04/08/25 | Glutamate indicators with increased sensitivity and tailored deactivation rates
Podgorski K, Aggarwal A, Negrean A, Chen Y, Iyer R, Reep D, Liu A, Palutla A, Xie M, Maclennan B, Hagihara K, Kinsey L, Sun J, Yao P, Zheng J, Tsang A, Tsegaye G, Zhang Y, Patel R, Hasseman J
Research Square. 2025 Apr 8:. doi: 10.21203/rs.3.rs-6257403/v1

Identifying the input-output operations of neurons requires measurements of synaptic transmission simultaneously at many of a neuron’s thousands of inputs in the intact brain. To facilitate this goal, we engineered and screened 3365 variants of the fluorescent protein glutamate indicator iGluSnFR3 in neuron culture, and selected variants in the mouse visual cortex. Two variants have high sensitivity, fast activation (< 2 ms) and deactivation times tailored for recording large populations of synapses (iGluSnFR4s, 153 ms) or rapid dynamics (iGluSnFR4f, 26 ms). By imaging action-potential evoked signals on axons and visually-evoked signals on dendritic spines, we show that iGluSnFR4s/4f primarily detect local synaptic glutamate with single-vesicle sensitivity. The indicators detect a wide range of naturalistic synaptic transmission, including in the vibrissal cortex layer 4 and in hippocampal CA1 dendrites. iGluSnFR4 increases the sensitivity and scale (4s) or speed (4f) of tracking information flow in neural networks in vivo.

View Publication Page
04/17/25 | Mitochondrial complexity is regulated at ER-mitochondria contact sites via PDZD8-FKBP8 tethering.
Nakamura K, Aoyama-Ishiwatari S, Nagao T, Paaran M, Obara CJ, Sakurai-Saito Y, Johnston J, Du Y, Suga S, Tsuboi M, Nakakido M, Tsumoto K, Kishi Y, Gotoh Y, Kwak C, Rhee H, Seo JK, Kosako H, Potter C, Carragher B, Lippincott-Schwartz J, Polleux F, Hirabayashi Y
Nat Commun. 2025 Apr 17;16(1):3401. doi: 10.1038/s41467-025-58538-3

Mitochondria-ER membrane contact sites (MERCS) represent a fundamental ultrastructural feature underlying unique biochemistry and physiology in eukaryotic cells. The ER protein PDZD8 is required for the formation of MERCS in many cell types, however, its tethering partner on the outer mitochondrial membrane (OMM) is currently unknown. Here we identify the OMM protein FKBP8 as the tethering partner of PDZD8 using a combination of unbiased proximity proteomics, CRISPR-Cas9 endogenous protein tagging, Cryo-electron tomography, and correlative light-electron microscopy. Single molecule tracking reveals highly dynamic diffusion properties of PDZD8 along the ER membrane with significant pauses and captures at MERCS. Overexpression of FKBP8 is sufficient to narrow the ER-OMM distance, whereas independent versus combined deletions of these two proteins demonstrate their interdependence for MERCS formation. Furthermore, PDZD8 enhances mitochondrial complexity in a FKBP8-dependent manner. Our results identify a novel ER-mitochondria tethering complex that regulates mitochondrial morphology in mammalian cells.

 

Preprint: 10.1101/2025.02.22.639343

View Publication Page
04/15/25 | Wnt/β-catenin signalling assists cell fate decision making in the early mouse embryo
Lilao-Garzón J, Corujo-Simon E, Vinyoles M, Fischer SC, Guillén J, Balayo T, Muñoz-Descalzo S
bioRxiv. 2025 Apr 15:. doi: 10.1101/2025.04.09.647220

Cell fate choice is a key event happening during preimplantation mouse development. From embryonic day 3.5 (E3.5) to E4.5, the inner cell mass (ICM) differentiates into epiblast (Epi, NANOG expressing cells) and primitive endoderm (PrE, GATA6, SOX17 and/or GATA4 expressing cells). The mechanism by which ICM cells differentiate into Epi cells and PrE cells remains partially unknown. FGF/ERK has been proposed as the main signalling pathway for this event, but it does not explain co-expression of NANOG and GAT6 or how the cell fate choice is initiated.

In this study, we investigate whether Wnt/β-catenin signalling also plays a role. To this end, we use two in vitro models based on inducible GATA6 expression: one in 2D, and another in 3D, namely ICM organoids. By combining these in vitro models with in vivo mouse embryos, chemical and classical genetics, and quantitative 3D immunofluorescence analyses, we propose a dual role for Wnt/β-catenin signalling.

We find that β-catenin, acting alongside FGF/ERK signalling, helps to guide the cell fate choice towards PrE. Additionally, by regulating GATA6 and GATA4 stability, β-catenin further facilitates this choice. To summarise, we observe that pathway activation promotes PrE differentiation, while its inhibition stalls it.

SUMMARY STATEMENT Wnt/β-catenin signalling promotes PrE fate in mouse preimplantation embryos.

View Publication Page
04/15/25 | Bio-inspired 3D-printed phantom: Encoding cellular heterogeneity for characterization of quantitative phase imaging
Sylvia Desissaire , Michał Ziemczonok , Tigrane Cantat-Moltrecht , Arkadiusz Kuś , Guillaume Godefroy , Lionel Hervé , Chiara Paviolo , Wojciech Krauze , Cédric Allier , Ondrej Mandula , Małgorzata Kujawińska
Measurement. 2025 Apr 15;247:116765. doi: 10.1016/j.measurement.2025.116765

Quantitative phase imaging (QPI) has proven to be a valuable tool for advanced biological and pharmacological research, providing phase information for the study of cell features and physiology in label-free conditions. The next step for QPI to become a gold standard is the quantitative assessment of the phase gradients over the different microscopy setups. Given the large variety of QPI systems, a systematic comparison is a challenging task, and requires a calibration target representative of the living samples. In this paper, we introduce a tailor-made 3D-printed phantom derived from phase images of eukaryotic cells. It comprises typical morphologies and optical thicknesses found in biological cultures and is characterized with digital holographic microscopy (reference measurements). The performance of three different full field QPI optical systems, in terms of optical path difference and dry mass accuracy, were evaluated. This phantom opens up other possibilities for the validation of reconstruction algorithms and post-processing routines, and paves the way for calibration targets designed ad hoc for specific biological questions.

View Publication Page
04/07/25 | Far-red fluorescent genetically encoded calcium ion indicators.
Dalangin R, Jia BZ, Qi Y, Aggarwal A, Sakoi K, Drobizhev M, Molina RS, Patel R, Abdelfattah AS, Zheng J, Reep D, Hasseman JP, GENIE Project Team , Zhao Y, Wu J, Podgorski K, Tebo AG, Schreiter ER, Hughes TE, Terai T, Paquet M, Megason SG, Cohen AE, Shen Y, Campbell RE
Nat Commun. 2025 Apr 07;16(1):3318. doi: 10.1038/s41467-025-58485-z

Genetically encoded calcium ion (Ca) indicators (GECIs) are widely-used molecular tools for functional imaging of Ca dynamics and neuronal activities with single-cell resolution. Here we report the design and development of two far-red fluorescent GECIs, FR-GECO1a and FR-GECO1c, based on the monomeric far-red fluorescent proteins mKelly1 and mKelly2. FR-GECOs have excitation and emission maxima at ~596 nm and ~644 nm, respectively, display large responses to Ca in vitro (ΔF/F = 6 for FR-GECO1a, 18 for FR-GECO1c), are bright under both one-photon and two-photon illumination, and have high affinities (apparent K = 29 nM for FR-GECO1a, 83 nM for FR-GECO1c) for Ca. FR-GECOs offer sensitive and fast detection of single action potentials in neurons, and enable in vivo all-optical manipulation and measurement of cellular activities in combination with optogenetic actuators.

Preprint: https://doi.org/10.1101/2020.11.12.380089

View Publication Page