1
|
Gong X, Wang S, Yuan J, Ji J, Zhao R, Huang J, Li B, Zhai Y, Zhong Y, Zheng Y, Jiang Q. Ferrocene-derived magnetic fiber-particles from diesel exhaust: enhanced pulmonary toxicity via Bach1-SAT1-polyamine depletion. J Nanobiotechnology 2025; 23:324. [PMID: 40301891 PMCID: PMC12039003 DOI: 10.1186/s12951-025-03397-9] [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: 12/23/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND AND AIM Magnetic nanoparticles are key components of air pollution. The combustion of diesel engine fuels, especially with ferrocene doping to reduce emissions, may increase exposure to these particles and related health risks. This study aimed to reveal the generation and characterization of ferrocene-derived magnetic particles (FMP) in ferrocene-doped diesel exhaust, and to investigate its toxicities and associated mechanisms in an avian model. METHODS FMP was observed in ferrocene-doped diesel exhaust particles, and extracted with neodymium magnets. Extracted FMP was characterized, and exposed to hatchling chickens via aerosol inhalation. Pulmonary toxicities were assessed with pathological and molecular methods. Associated mechanisms were investigated with RNA-seq, in vitro cell culture, and in vivo gene silencing. RESULTS FMP was characterized to be fibrous, magnetic iron-containing carbon particles. Extracted FMP could directly induce pulmonary toxicity. Mechanistic investigations revealed molecular mechanism associated with ferroptosis via Bach1, SAT1 and polyamines depletion, and further confirmed with ferroptosis inhibitor treatment, Bach1 inhibitor treatment, supplementation of polyamines or SAT1 silencing. CONCLUSIONS Ferrocene doping could result in formation of magnetic particles in diesel exhaust. For the first time, magnetic fiber-like particles were extracted from ferrocene-doped DE particles, which is a potential source of magnetic particles in air pollution. To better balance emission control and health effects, further investigations are necessary.
Collapse
Affiliation(s)
- Xinxian Gong
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Siyi Wang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Junhua Yuan
- Department of Special Medicine, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jing Ji
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Rui Zhao
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jing Huang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Boyang Li
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Yunuo Zhai
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Yuxu Zhong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China.
| | - Qixiao Jiang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China.
| |
Collapse
|
2
|
Van Heest A, Wang Y, Zhang L, Phillips LA, Karsen SD, Nelson C, Knight HL, Perper SJ, O’Brien S, Clements M, Sun VZ, Goodearl A, Schwartz Sterman A, Mitra S. Quantitative Assessment of Pulmonary Fibrosis in a Murine Model via a Multimodal Imaging Workflow. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:85-94. [PMID: 40018646 PMCID: PMC11863149 DOI: 10.1021/cbmi.4c00065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 03/01/2025]
Abstract
Disease-recapitulating animal models are valuable tools in preclinical development for the study of compounds. In the case of fibrotic pulmonary diseases such as idiopathic pulmonary fibrosis (IPF), the bleomycin model of lung injury in the mouse is widely used. To evaluate bleomycin-induced changes in the lung, we employed a quantitative, multimodal approach. Using in vivo microcomputed tomography (μCT), we demonstrated radiographic changes associated with disease progression in aeration levels of the lung parenchyma. There exists an unmet need for a quantitative, high-resolution imaging probe to detect pulmonary fibrosis, particularly that can differentiate between inflammatory and fibrotic components of the disease. Matrix remodeling and overexpression of extracellular matrix (ECM) proteins such as collagen and fibronectin are hallmarks of organ fibrosis. A splice variant of fibronectin containing extra domain A (FnEDA) is of particular interest in fibrosis due to its high level of expression in diseased tissue, which is confirmed here using immunohistochemistry (IHC) in mouse and human lungs. An antibody against FnEDA was evaluated for use as an imaging tool, particularly by using in vivo single-photon emission computed tomography (SPECT) and ex vivo near-infrared (NIR) fluorescence imaging. These data were further corroborated with histological tissue staining and fibrosis quantitation based on a Modified Ashcroft (MA) score and a digital image analysis of whole slide lung tissue sections. The fusion of these different approaches represents a robust integrated workflow combining anatomical and molecular imaging technologies to enable the visualization and quantitation of disease activity and treatment response with an inhibitor of the TGFβ signaling pathway.
Collapse
Affiliation(s)
| | | | - Liang Zhang
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Lucy A. Phillips
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Samuel D. Karsen
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Christine Nelson
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Heather L. Knight
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Stuart J. Perper
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Stephen O’Brien
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Meghan Clements
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Victor Z. Sun
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Andrew Goodearl
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | | | - Soumya Mitra
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| |
Collapse
|
3
|
Goto T, Sano A, Onishi S, Hada N, Kimata R, Matsuo S, Oyama S, Kato A, Mizuno H, Yamazaki M. Optimized digital workflow for pathologist-grade evaluation in bleomycin-induced pulmonary fibrosis mouse model. Sci Rep 2025; 15:2331. [PMID: 39833349 PMCID: PMC11747197 DOI: 10.1038/s41598-025-86544-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disorder of unknown etiology, characterized by interstitial fibrosis of the lungs. Bleomycin-induced pulmonary fibrosis mouse model (BLM model) is a widely used animal model to evaluate therapeutic targets for IPF. Histopathological analysis of lung fibrosis is an important method for evaluating BLM model. However, this method requires expertise in recognizing complex visual patterns and is time-consuming, making the workflow difficult and inefficient. Therefore, we developed a new workflow for BLM model that reduces inter- and intra-observer variations and improves the evaluation process. We generated deep learning models for grading lung fibrosis that were able to achieve accuracy comparable to that of pathologists. These models incorporate complex image patterns and qualitative factors, such as collagen texture and distribution, potentially identifying drug candidates overlooked in evaluations based solely on simple area extraction. This deep learning-based fibrosis grade assessment has the potential to streamline drug development for pulmonary fibrosis by offering higher granularity and reproducibility in evaluating BLM model.
Collapse
Affiliation(s)
- Toshiki Goto
- Research Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Akira Sano
- ExaWizards Inc., 4-2-8 Shibaura, Minato-ku, Tokyo, 108-0023, Japan.
| | - Shinichi Onishi
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Natsuko Hada
- Research Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Rui Kimata
- ExaWizards Inc., 4-2-8 Shibaura, Minato-ku, Tokyo, 108-0023, Japan
| | - Saori Matsuo
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Sohei Oyama
- Research Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Atsuhiko Kato
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Hideaki Mizuno
- Research Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Masaki Yamazaki
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, 216 Totsuka-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan.
| |
Collapse
|
4
|
Karz A, Coudray N, Bayraktar E, Galbraith K, Jour G, Shadaloey AAS, Eskow N, Rubanov A, Navarro M, Moubarak R, Baptiste G, Levinson G, Mezzano V, Alu M, Loomis C, Lima D, Rubens A, Jilaveanu L, Tsirigos A, Hernando E. MetFinder: A Tool for Automated Quantitation of Metastatic Burden in Histological Sections From Preclinical Models. Pigment Cell Melanoma Res 2025; 38:e13195. [PMID: 39254030 PMCID: PMC11948878 DOI: 10.1111/pcmr.13195] [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: 02/08/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024]
Abstract
As efforts to study the mechanisms of melanoma metastasis and novel therapeutic approaches multiply, researchers need accurate, high-throughput methods to evaluate the effects on tumor burden resulting from specific interventions. We show that automated quantification of tumor content from whole slide images is a compelling solution to assess in vivo experiments. In order to increase the outflow of data collection from preclinical studies, we assembled a large dataset with annotations and trained a deep neural network for the quantitative analysis of melanoma tumor content on histopathological sections of murine models. After assessing its performance in segmenting these images, the tool obtained consistent results with an orthogonal method (bioluminescence) of measuring metastasis in an experimental setting. This AI-based algorithm, made freely available to academic laboratories through a web-interface called MetFinder, promises to become an asset for melanoma researchers and pathologists interested in accurate, quantitative assessment of metastasis burden.
Collapse
Affiliation(s)
- Alcida Karz
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Nicolas Coudray
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, NY 10016
- Department of Cell Biology, NYU School of Medicine; New York, NY, USA
| | - Erol Bayraktar
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Kristyn Galbraith
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
| | - George Jour
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
| | - Arman Alberto Sorin Shadaloey
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Nicole Eskow
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Andrey Rubanov
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Maya Navarro
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Rana Moubarak
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Gillian Baptiste
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Grace Levinson
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| | - Valeria Mezzano
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016
| | - Mark Alu
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016
| | - Cynthia Loomis
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016
| | - Daniel Lima
- Research Software Engineering Core, Medical Center Information Technology Department, NYU Langone Health, New York, NY 10016
| | - Adam Rubens
- Research Software Engineering Core, Medical Center Information Technology Department, NYU Langone Health, New York, NY 10016
| | - Lucia Jilaveanu
- Department of Medicine, Yale University, New Haven, CT 06510
| | - Aristotelis Tsirigos
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, NY 10016
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016
| |
Collapse
|
5
|
Reininger D, Fundel-Clemens K, Mayr CH, Wollin L, Laemmle B, Quast K, Nickolaus P, Herrmann FE. PDE4B inhibition by nerandomilast: Effects on lung fibrosis and transcriptome in fibrotic rats and on biomarkers in human lung epithelial cells. Br J Pharmacol 2024; 181:4766-4781. [PMID: 39183442 DOI: 10.1111/bph.17303] [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: 02/22/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND AND PURPOSE The PDE4 family is considered a prime target for therapeutic intervention in several fibro-inflammatory diseases. We have investigated the molecular mechanisms of nerandomilast (BI 1015550), a preferential PDE4B inhibitor. EXPERIMENTAL APPROACH In addition to clinically relevant parameters of idiopathic pulmonary fibrosis (IPF; lung function measurement/high-resolution computed tomography scan/AI-Ashcroft score), whole-lung homogenates from a therapeutic male Wistar rat model of pulmonary fibrosis were analysed by next-generation sequencing (NGS). Data were matched with public domain data derived from human IPF samples to investigate how well the rat model reflected human IPF. We scored the top counter-regulated genes following treatment with nerandomilast in human single cells and validated disease markers discovered in the rat model using a human disease-relevant in vitro assay of IPF. KEY RESULTS Nerandomilast improved the decline of lung function parameters in bleomycin-treated animals. In the NGS study, most transcripts deregulated by bleomycin treatment were normalised by nerandomilast treatment. Most notably, a significant number of deregulated transcripts that were identified in human IPF disease were also found in the animal model and reversed by nerandomilast. Mapping to single-cell data revealed the strongest effects on mesenchymal, epithelial and endothelial cell populations. In a primary human epithelial cell culture system, several disease-related (bio)markers were inhibited by nerandomilast in a concentration-dependent manner. CONCLUSIONS AND IMPLICATIONS This study further supports the available knowledge about the anti-inflammatory/antifibrotic mechanisms of nerandomilast and provides novel insights into the mode of action and signalling pathways influenced by nerandomilast treatment of lung fibrosis.
Collapse
Affiliation(s)
- Dennis Reininger
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Katrin Fundel-Clemens
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christoph H Mayr
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Lutz Wollin
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Baerbel Laemmle
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Karsten Quast
- Global Clinical Development & Operations, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Peter Nickolaus
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Franziska Elena Herrmann
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
6
|
Ségard BD, Kimura K, Matsuoka Y, Imamura T, Ikeda A, Iwamiya T. Quantification of fibrosis extend and airspace availability in lung: A semi-automatic ImageJ/Fiji toolbox. PLoS One 2024; 19:e0298015. [PMID: 38421996 PMCID: PMC10903859 DOI: 10.1371/journal.pone.0298015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 01/17/2024] [Indexed: 03/02/2024] Open
Abstract
The evaluation of the structural integrity of mechanically dynamic organs such as lungs is critical for the diagnosis of numerous pathologies and the development of therapies. This task is classically performed by histology experts in a qualitative or semi-quantitative manner. Automatic digital image processing methods appeared in the last decades, and although immensely powerful, tools are highly specialized and lack the versatility required in various experimental designs. Here, a set of scripts for the image processing software ImageJ/Fiji to easily quantify fibrosis extend and alveolar airspace availability in Sirius Red or Masson's trichrome stained samples is presented. The toolbox consists in thirteen modules: sample detection, particles filtration (automatic and manual), border definition, air ducts identification, air ducts walls definition, parenchyma extraction, MT-staining specific pre-processing, fibrosis detection, fibrosis particles filtration, airspace detection, and visualizations (tissue only or tissue and airspace). While the process is largely automated, critical parameters are accessible to the user for increased adaptability. The modularity of the protocol allows for its adjustment to alternative experimental settings. Fibrosis and airspace can be combined as an evaluation of the structural integrity of the organ. All settings and intermediate states are saved to ensure reproducibility. These new analysis scripts allow for a rapid quantification of fibrosis and airspace in a large variety of experimental settings.
Collapse
Affiliation(s)
| | - Kodai Kimura
- Research and Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
| | - Yuimi Matsuoka
- Research and Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
| | - Tomomi Imamura
- Research and Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
| | - Ayana Ikeda
- Research and Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
| | - Takahiro Iwamiya
- Research and Development Department, Metcela Inc., Kawasaki, Kanagawa, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| |
Collapse
|
7
|
Wohnhaas CT, Baßler K, Watson CK, Shen Y, Leparc GG, Tilp C, Heinemann F, Kind D, Stierstorfer B, Delić D, Brunner T, Gantner F, Schultze JL, Viollet C, Baum P. Monocyte-derived alveolar macrophages are key drivers of smoke-induced lung inflammation and tissue remodeling. Front Immunol 2024; 15:1325090. [PMID: 38348034 PMCID: PMC10859862 DOI: 10.3389/fimmu.2024.1325090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Smoking is a leading risk factor of chronic obstructive pulmonary disease (COPD), that is characterized by chronic lung inflammation, tissue remodeling and emphysema. Although inflammation is critical to COPD pathogenesis, the cellular and molecular basis underlying smoking-induced lung inflammation and pathology remains unclear. Using murine smoke models and single-cell RNA-sequencing, we show that smoking establishes a self-amplifying inflammatory loop characterized by an influx of molecularly heterogeneous neutrophil subsets and excessive recruitment of monocyte-derived alveolar macrophages (MoAM). In contrast to tissue-resident AM, MoAM are absent in homeostasis and characterized by a pro-inflammatory gene signature. Moreover, MoAM represent 46% of AM in emphysematous mice and express markers causally linked to emphysema. We also demonstrate the presence of pro-inflammatory and tissue remodeling associated MoAM orthologs in humans that are significantly increased in emphysematous COPD patients. Inhibition of the IRAK4 kinase depletes a rare inflammatory neutrophil subset, diminishes MoAM recruitment, and alleviates inflammation in the lung of cigarette smoke-exposed mice. This study extends our understanding of the molecular signaling circuits and cellular dynamics in smoking-induced lung inflammation and pathology, highlights the functional consequence of monocyte and neutrophil recruitment, identifies MoAM as key drivers of the inflammatory process, and supports their contribution to pathological tissue remodeling.
Collapse
Affiliation(s)
- Christian T. Wohnhaas
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Kevin Baßler
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Carolin K. Watson
- Immunology & Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Yang Shen
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Germán G. Leparc
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Cornelia Tilp
- Immunology & Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Fabian Heinemann
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David Kind
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Birgit Stierstorfer
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Denis Delić
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Brunner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Florian Gantner
- Department of Biology, University of Konstanz, Konstanz, Germany
- Translational Medicine & Clinical Pharmacology, C. H. Boehringer Sohn AG & Co. KG, Biberach, Germany
| | - Joachim L. Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and University of Bonn, Bonn, Germany
| | - Coralie Viollet
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Patrick Baum
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
8
|
Handa T. The potential role of artificial intelligence in the clinical practice of interstitial lung disease. Respir Investig 2023; 61:702-710. [PMID: 37708636 DOI: 10.1016/j.resinv.2023.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/26/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023]
Abstract
Artificial intelligence (AI) is being widely applied in the field of medicine, in areas such as drug discovery, diagnostic support, and assistance with medical practice. Among these, medical imaging is an area where AI is expected to make a significant contribution. In Japan, as of November 2022, 23 AI medical devices have received regulatory approval; all these devices are related to image analysis. In interstitial lung diseases, technologies have been developed that use AI to analyze high-resolution computed tomography and pathological images, and gene expression patterns in tissue taken from transbronchial lung biopsies to assist in the diagnosis of idiopathic pulmonary fibrosis. Some of these technologies are already being used in clinical practice in the United States. AI is expected to reduce the burden on physicians, improve reproducibility, and advance personalized medicine. Obtaining sufficient data for diseases with a small number of patients is difficult. Additionally, certain issues must be addressed in order for AI to be applied in healthcare. These issues include taking responsibility for the AI results output, updating software after the launch of technology, and adapting to new imaging technologies. Establishing research infrastructures such as large-scale databases and common platforms is important for the development of AI technology: their use requires an understanding of the characteristics and limitations of the systems. CLINICAL TRIAL REGISTRATION: Not applicable.
Collapse
Affiliation(s)
- Tomohiro Handa
- Department of Advanced Medicine for Respiratory Failure and Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
9
|
Luchian A, Cepeda KT, Harwood R, Murray P, Wilm B, Kenny S, Pregel P, Ressel L. Quantifying acute kidney injury in an Ischaemia-Reperfusion Injury mouse model using deep-learning-based semantic segmentation in histology. Biol Open 2023; 12:bio059988. [PMID: 37642317 PMCID: PMC10537956 DOI: 10.1242/bio.059988] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
This study focuses on ischaemia-reperfusion injury (IRI) in kidneys, a cause of acute kidney injury (AKI) and end-stage kidney disease (ESKD). Traditional kidney damage assessment methods are semi-quantitative and subjective. This study aims to use a convolutional neural network (CNN) to segment murine kidney structures after IRI, quantify damage via CNN-generated pathological measurements, and compare this to conventional scoring. The CNN was able to accurately segment the different pathological classes, such as Intratubular casts and Tubular necrosis, with an F1 score of over 0.75. Some classes, such as Glomeruli and Proximal tubules, had even higher statistical values with F1 scores over 0.90. The scoring generated based on the segmentation approach statistically correlated with the semiquantitative assessment (Spearman's rank correlation coefficient=0.94). The heatmap approach localised the intratubular necrosis mainly in the outer stripe of the outer medulla, while the tubular casts were also present in more superficial or deeper portions of the cortex and medullary areas. This study presents a CNN model capable of segmenting multiple classes of interest, including acute IRI-specific pathological changes, in a whole mouse kidney section and can provide insights into the distribution of pathological classes within the whole mouse kidney section.
Collapse
Affiliation(s)
- Andreea Luchian
- Department of Veterinary Anatomy Physiology and Pathology, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, CH64 7TE, UK
| | - Katherine Trivino Cepeda
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
- Centre for Pre-clinical Imaging, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7TX, UK
| | - Rachel Harwood
- Department of Paediatric Surgery, Alder Hey in the Park, Liverpool, L14 5AB, UK
| | - Patricia Murray
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
- Centre for Pre-clinical Imaging, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7TX, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
- Centre for Pre-clinical Imaging, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7TX, UK
| | - Simon Kenny
- Department of Paediatric Surgery, Alder Hey in the Park, Liverpool, L14 5AB, UK
| | - Paola Pregel
- Department of Veterinary Sciences, University of Turin, Turin, 8-10124, Italy
| | - Lorenzo Ressel
- Department of Veterinary Anatomy Physiology and Pathology, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, CH64 7TE, UK
| |
Collapse
|
10
|
Viswanathan VS, Toro P, Corredor G, Mukhopadhyay S, Madabhushi A. The state of the art for artificial intelligence in lung digital pathology. J Pathol 2022; 257:413-429. [PMID: 35579955 PMCID: PMC9254900 DOI: 10.1002/path.5966] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
Lung diseases carry a significant burden of morbidity and mortality worldwide. The advent of digital pathology (DP) and an increase in computational power have led to the development of artificial intelligence (AI)-based tools that can assist pathologists and pulmonologists in improving clinical workflow and patient management. While previous works have explored the advances in computational approaches for breast, prostate, and head and neck cancers, there has been a growing interest in applying these technologies to lung diseases as well. The application of AI tools on radiology images for better characterization of indeterminate lung nodules, fibrotic lung disease, and lung cancer risk stratification has been well documented. In this article, we discuss methodologies used to build AI tools in lung DP, describing the various hand-crafted and deep learning-based unsupervised feature approaches. Next, we review AI tools across a wide spectrum of lung diseases including cancer, tuberculosis, idiopathic pulmonary fibrosis, and COVID-19. We discuss the utility of novel imaging biomarkers for different types of clinical problems including quantification of biomarkers like PD-L1, lung disease diagnosis, risk stratification, and prediction of response to treatments such as immune checkpoint inhibitors. We also look briefly at some emerging applications of AI tools in lung DP such as multimodal data analysis, 3D pathology, and transplant rejection. Lastly, we discuss the future of DP-based AI tools, describing the challenges with regulatory approval, developing reimbursement models, planning clinical deployment, and addressing AI biases. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Paula Toro
- Department of PathologyCleveland ClinicClevelandOHUSA
| | - Germán Corredor
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| | | | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| |
Collapse
|
11
|
Arlova A, Jin C, Wong-Rolle A, Chen ES, Lisle C, Brown GT, Lay N, Choyke PL, Turkbey B, Harmon S, Zhao C. Artificial Intelligence-based Tumor Segmentation in Mouse Models of Lung Adenocarcinoma. J Pathol Inform 2022; 13:100007. [PMID: 35242446 PMCID: PMC8860735 DOI: 10.1016/j.jpi.2022.100007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mouse models are highly effective for studying the pathophysiology of lung adenocarcinoma and evaluating new treatment strategies. Treatment efficacy is primarily determined by the total tumor burden measured on excised tumor specimens. The measurement process is time-consuming and prone to human errors. To address this issue, we developed a novel deep learning model to segment lung tumor foci on digitally scanned hematoxylin and eosin (H&E) histology slides. METHODS Digital slides of 239 mice from 9 experimental cohorts were split into training (n=137), validation (n=37), and testing cohorts (n=65). Image patches of 500×500 pixels were extracted from 5× and 10× magnifications, along with binary masks of expert annotations representing ground-truth tumor regions. Deep learning models utilizing DeepLabV3+ and UNet architectures were trained for binary segmentation of tumor foci under varying stain normalization conditions. The performance of algorithm segmentation was assessed by Dice Coefficient, and detection was evaluated by sensitivity and positive-predictive value (PPV). RESULTS The best model on patch-based validation was DeepLabV3+ using a Resnet-50 backbone, which achieved Dice 0.890 and 0.873 on validation and testing cohort, respectively. This result corresponded to 91.3 Sensitivity and 51.0 PPV in the validation cohort and 93.7 Sensitivity and 51.4 PPV in the testing cohort. False positives could be reduced 10-fold with thresholding artificial intelligence (AI) predicted output by area, without negative impact on Dice Coefficient. Evaluation at various stain normalization strategies did not demonstrate improvement from the baseline model. CONCLUSIONS A robust AI-based algorithm for detecting and segmenting lung tumor foci in the pre-clinical mouse models was developed. The output of this algorithm is compatible with open-source software that researchers commonly use.
Collapse
Affiliation(s)
- Alena Arlova
- Artificial Intelligence Resource, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Chengcheng Jin
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Abigail Wong-Rolle
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Eric S. Chen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - G. Thomas Brown
- Artificial Intelligence Resource, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Nathan Lay
- Artificial Intelligence Resource, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Peter L. Choyke
- Artificial Intelligence Resource, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Baris Turkbey
- Artificial Intelligence Resource, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Stephanie Harmon
- Artificial Intelligence Resource, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Chen Zhao
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
12
|
Le HQ, Hill MA, Kollak I, Keck M, Schroeder V, Wirth J, Skronska‐Wasek W, Schruf E, Strobel B, Stahl H, Herrmann FE, Campos AR, Li J, Quast K, Knebel D, Viollet C, Thomas MJ, Lamb D, Garnett JP. An EZH2-dependent transcriptional complex promotes aberrant epithelial remodelling after injury. EMBO Rep 2021; 22:e52785. [PMID: 34224201 PMCID: PMC8339687 DOI: 10.15252/embr.202152785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/05/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Unveiling the molecular mechanisms of tissue remodelling following injury is imperative to elucidate its regenerative capacity and aberrant repair in disease. Using different omics approaches, we identified enhancer of zester homolog 2 (EZH2) as a key regulator of fibrosis in injured lung epithelium. Epithelial injury drives an enrichment of nuclear transforming growth factor-β-activated kinase 1 (TAK1) that mediates EZH2 phosphorylation to facilitate its liberation from polycomb repressive complex 2 (PRC2). This process results in the establishment of a transcriptional complex of EZH2, RNA-polymerase II (POL2) and nuclear actin, which orchestrates aberrant epithelial repair programmes. The liberation of EZH2 from PRC2 is accompanied by an EZH2-EZH1 switch to preserve H3K27me3 deposition at non-target genes. Loss of epithelial TAK1, EZH2 or blocking nuclear actin influx attenuates the fibrotic cascade and restores respiratory homeostasis. Accordingly, EZH2 inhibition significantly improves outcomes in a pulmonary fibrosis mouse model. Our results reveal an important non-canonical function of EZH2, paving the way for new therapeutic interventions in fibrotic lung diseases.
Collapse
Affiliation(s)
- Huy Q Le
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Matthew A Hill
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
- University of BathBathUK
| | - Ines Kollak
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Martina Keck
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Victoria Schroeder
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Johannes Wirth
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Wioletta Skronska‐Wasek
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Eva Schruf
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Benjamin Strobel
- Drug Discovery SciencesBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Heiko Stahl
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Franziska E Herrmann
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | | | - Jun Li
- Immunology and Respiratory Disease Research DepartmentBoehringer Ingelheim Pharmaceuticals, IncRidgefieldCTUSA
| | - Karsten Quast
- Global Computational Biology and Digital SciencesBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Dagmar Knebel
- Global Computational Biology and Digital SciencesBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Coralie Viollet
- Global Computational Biology and Digital SciencesBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Matthew J Thomas
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
- University of BathBathUK
| | - David Lamb
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - James P Garnett
- Lung Repair & Regeneration DepartmentBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
- Translational and Clinical Research InstituteNewcastle UniversityNewcastleUK
| |
Collapse
|
13
|
Testa LC, Jule Y, Lundh L, Bertotti K, Merideth MA, O'Brien KJ, Nathan SD, Venuto DC, El-Chemaly S, Malicdan MCV, Gochuico BR. Automated Digital Quantification of Pulmonary Fibrosis in Human Histopathology Specimens. Front Med (Lausanne) 2021; 8:607720. [PMID: 34211981 PMCID: PMC8240807 DOI: 10.3389/fmed.2021.607720] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is characterized by abnormal interstitial extracellular matrix and cellular accumulations. Methods quantifying fibrosis severity in lung histopathology samples are semi-quantitative, subjective, and analyze only portions of sections. We sought to determine whether automated computerized imaging analysis shown to continuously measure fibrosis in mice could also be applied in human samples. A pilot study was conducted to analyze a small number of specimens from patients with Hermansky-Pudlak syndrome pulmonary fibrosis (HPSPF) or idiopathic pulmonary fibrosis (IPF). Digital images of entire lung histological serial sections stained with picrosirius red and alcian blue or anti-CD68 antibody were analyzed using dedicated software to automatically quantify fibrosis, collagen, and macrophage content. Automated fibrosis quantification based on parenchymal tissue density and fibrosis score measurements was compared to pulmonary function values or Ashcroft score. Automated fibrosis quantification of HPSPF lung explants was significantly higher than that of IPF lung explants or biopsies and was also significantly higher in IPF lung explants than in IPF biopsies. A high correlation coefficient was found between some automated quantification measurements and lung function values for the three sample groups. Automated quantification of collagen content in lung sections used for digital image analyses was similar in the three groups. CD68 immunolabeled cell measurements were significantly higher in HPSPF explants than in IPF biopsies. In conclusion, computerized image analysis provides access to accurate, reader-independent pulmonary fibrosis quantification in human histopathology samples. Fibrosis, collagen content, and immunostained cells can be automatically and individually quantified from serial sections. Robust automated digital image analysis of human lung samples enhances the available tools to quantify and study fibrotic lung disease.
Collapse
Affiliation(s)
- Lauren C Testa
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | | | - Linnea Lundh
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | | | - Melissa A Merideth
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kevin J O'Brien
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Steven D Nathan
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA, United States
| | - Drew C Venuto
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA, United States
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - May Christine V Malicdan
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States.,Undiagnosed Diseases Program, Office of the Director, National Institutes of Health, Bethesda, MD, United States
| | - Bernadette R Gochuico
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
14
|
Jones MA, MacCuaig WM, Frickenstein AN, Camalan S, Gurcan MN, Holter-Chakrabarty J, Morris KT, McNally MW, Booth KK, Carter S, Grizzle WE, McNally LR. Molecular Imaging of Inflammatory Disease. Biomedicines 2021; 9:152. [PMID: 33557374 PMCID: PMC7914540 DOI: 10.3390/biomedicines9020152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/31/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory diseases include a wide variety of highly prevalent conditions with high mortality rates in severe cases ranging from cardiovascular disease, to rheumatoid arthritis, to chronic obstructive pulmonary disease, to graft vs. host disease, to a number of gastrointestinal disorders. Many diseases that are not considered inflammatory per se are associated with varying levels of inflammation. Imaging of the immune system and inflammatory response is of interest as it can give insight into disease progression and severity. Clinical imaging technologies such as computed tomography (CT) and magnetic resonance imaging (MRI) are traditionally limited to the visualization of anatomical information; then, the presence or absence of an inflammatory state must be inferred from the structural abnormalities. Improvement in available contrast agents has made it possible to obtain functional information as well as anatomical. In vivo imaging of inflammation ultimately facilitates an improved accuracy of diagnostics and monitoring of patients to allow for better patient care. Highly specific molecular imaging of inflammatory biomarkers allows for earlier diagnosis to prevent irreversible damage. Advancements in imaging instruments, targeted tracers, and contrast agents represent a rapidly growing area of preclinical research with the hopes of quick translation to the clinic.
Collapse
Affiliation(s)
- Meredith A. Jones
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (M.A.J.); (W.M.M.); (A.N.F.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - William M. MacCuaig
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (M.A.J.); (W.M.M.); (A.N.F.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA; (M.A.J.); (W.M.M.); (A.N.F.)
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - Seda Camalan
- Department of Internal Medicine, Wake Forest Baptist Health, Winston-Salem, NC 27157, USA; (S.C.); (M.N.G.)
| | - Metin N. Gurcan
- Department of Internal Medicine, Wake Forest Baptist Health, Winston-Salem, NC 27157, USA; (S.C.); (M.N.G.)
| | - Jennifer Holter-Chakrabarty
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Medicine, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Katherine T. Morris
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Molly W. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
| | - Kristina K. Booth
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Steven Carter
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Lacey R. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (J.H.-C.); (K.T.M.); (M.W.M.); (K.K.B.); (S.C.)
- Department of Surgery, University of Oklahoma, Oklahoma City, OK 73104, USA
| |
Collapse
|
15
|
Pischon H, Mason D, Lawrenz B, Blanck O, Frisk AL, Schorsch F, Bertani V. Artificial Intelligence in Toxicologic Pathology: Quantitative Evaluation of Compound-Induced Hepatocellular Hypertrophy in Rats. Toxicol Pathol 2021; 49:928-937. [PMID: 33397216 DOI: 10.1177/0192623320983244] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Digital pathology evolved rapidly, enabling more systematic usage of image analysis and development of artificial intelligence (AI) applications. Here, combined AI models were developed to evaluate hepatocellular hypertrophy in rat liver, using commercial AI-based software on hematoxylin and eosin-stained whole slide images. In a first approach, deep learning-based identification of critical tissue zones (centrilobular, midzonal, and periportal) enabled evaluation of region-specific cell size. Mean cytoplasmic area of hepatocytes was calculated via several sequential algorithms including segmentation in microanatomical structures (separation of sinusoids and vessels from hepatocytes), nuclear detection, and area measurements. An increase in mean cytoplasmic area could be shown in groups given phenobarbital, known to induce hepatocellular hypertrophy when compared to control groups, in multiple studies. Quantitative results correlated with the gold standard: observation and grading performed by board-certified veterinary pathologists, liver weights, and gene expression. Furthermore, as a second approach, we introduce for the first time deep learning-based direct detection of hepatocellular hypertrophy with similar results. Cell hypertrophy is challenging to pick up, particularly in milder cases. Additional evaluation of mean cytoplasmic area or direct detection of hypertrophy, combined with histopathological observations and liver weights, is expected to increase accuracy and repeatability of diagnoses and grading by pathologists.
Collapse
Affiliation(s)
- Hannah Pischon
- 483305Nuvisan Pharma Grafing GmbH, Bayer AG, Berlin, Germany.,Nuvisan ICB GmbH, Berlin, Germany
| | | | - Bettina Lawrenz
- 483305Nuvisan Pharma Grafing GmbH, Bayer AG, Wuppertal, Germany
| | - Olivier Blanck
- 55075Bayer CropScience SAS, Sophia Antipolis, Valbonne, France
| | - Anna-Lena Frisk
- 483305Nuvisan Pharma Grafing GmbH, Bayer AG, Berlin, Germany.,Janssen Pharmaceutica, Beerse, Belgium
| | | | - Valeria Bertani
- 55075Bayer CropScience SAS, Sophia Antipolis, Valbonne, France
| |
Collapse
|
16
|
Zingman I, Zippel N, Birk G, Eder S, Thomas L, Schönberger T, Stierstorfer B, Heinemann F. Deep Learning-Based Detection of Endothelial Tip Cells in the Oxygen-Induced Retinopathy Model. Toxicol Pathol 2020; 49:862-871. [PMID: 33896293 DOI: 10.1177/0192623320972964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Proliferative retinopathies, such as diabetic retinopathy and retinopathy of prematurity, are leading causes of vision impairment. A common feature is a loss of retinal capillary vessels resulting in hypoxia and neuronal damage. The oxygen-induced retinopathy model is widely used to study revascularization of an ischemic area in the mouse retina. The presence of endothelial tip cells indicates vascular recovery; however, their quantification relies on manual counting in microscopy images of retinal flat mount preparations. Recent advances in deep neural networks (DNNs) allow the automation of such tasks. We demonstrate a workflow for detection of tip cells in retinal images using the DNN-based Single Shot Detector (SSD). The SSD was designed for detection of objects in natural images. We adapt the SSD architecture and training procedure to the tip cell detection task and retrain the DNN using labeled tip cells in images of fluorescently stained retina flat mounts. Transferring knowledge from the pretrained DNN and extensive data augmentation reduced the amount of required labeled data. Our system shows a performance comparable to the human level, while providing highly consistent results. Therefore, such a system can automate counting of tip cells, a readout frequently used in retinopathy research, thereby reducing routine work for biomedical experts.
Collapse
Affiliation(s)
- Igor Zingman
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| | - Nina Zippel
- Cardiometabolic-Diseases Research, 417986Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| | - Gerald Birk
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| | - Sebastian Eder
- Cardiometabolic-Diseases Research, 417986Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| | - Leo Thomas
- Cardiometabolic-Diseases Research, 417986Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| | - Tanja Schönberger
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| | - Birgit Stierstorfer
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| | - Fabian Heinemann
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riß, Germany
| |
Collapse
|
17
|
Courtoy GE, Leclercq I, Froidure A, Schiano G, Morelle J, Devuyst O, Huaux F, Bouzin C. Digital Image Analysis of Picrosirius Red Staining: A Robust Method for Multi-Organ Fibrosis Quantification and Characterization. Biomolecules 2020; 10:biom10111585. [PMID: 33266431 PMCID: PMC7709042 DOI: 10.3390/biom10111585] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Current understanding of fibrosis remains incomplete despite the increasing burden of related diseases. Preclinical models are used to dissect the pathogenesis and dynamics of fibrosis, and to evaluate anti-fibrotic therapies. These studies require objective and accurate measurements of fibrosis. Existing histological quantification methods are operator-dependent, organ-specific, and/or need advanced equipment. Therefore, we developed a robust, minimally operator-dependent, and tissue-transposable digital method for fibrosis quantification. The proposed method involves a novel algorithm for more specific and more sensitive detection of collagen fibers stained by picrosirius red (PSR), a computer-assisted segmentation of histological structures, and a new automated morphological classification of fibers according to their compactness. The new algorithm proved more accurate than classical filtering using principal color component (red-green-blue; RGB) for PSR detection. We applied this new method on established mouse models of liver, lung, and kidney fibrosis and demonstrated its validity by evidencing topological collagen accumulation in relevant histological compartments. Our data also showed an overall accumulation of compact fibers concomitant with worsening fibrosis and evidenced topological changes in fiber compactness proper to each model. In conclusion, we describe here a robust digital method for fibrosis analysis allowing accurate quantification, pattern recognition, and multi-organ comparisons useful to understand fibrosis dynamics.
Collapse
Affiliation(s)
- Guillaume E. Courtoy
- IREC Imaging Platform (2IP), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Isabelle Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium
- Correspondence: (I.L.); (C.B.)
| | - Antoine Froidure
- Pole of Pneumology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Guglielmo Schiano
- Mechanisms of Inherited Kidney Diseases Group, University of Zurich, 8057 Zurich, Switzerland; (G.S.); (O.D.)
| | - Johann Morelle
- Pole of Nephrology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Olivier Devuyst
- Mechanisms of Inherited Kidney Diseases Group, University of Zurich, 8057 Zurich, Switzerland; (G.S.); (O.D.)
- Pole of Nephrology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Caroline Bouzin
- IREC Imaging Platform (2IP), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
- Correspondence: (I.L.); (C.B.)
| |
Collapse
|
18
|
Mäkelä K, Mäyränpää MI, Sihvo HK, Bergman P, Sutinen E, Ollila H, Kaarteenaho R, Myllärniemi M. Artificial intelligence identifies inflammation and confirms fibroblast foci as prognostic tissue biomarkers in idiopathic pulmonary fibrosis. Hum Pathol 2020; 107:58-68. [PMID: 33161029 DOI: 10.1016/j.humpath.2020.10.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/27/2020] [Accepted: 10/30/2020] [Indexed: 12/21/2022]
Abstract
A large number of fibroblast foci (FF) predict mortality in idiopathic pulmonary fibrosis (IPF). Other prognostic histological markers have not been identified. Artificial intelligence (AI) offers a possibility to quantitate possible prognostic histological features in IPF. We aimed to test the use of AI in IPF lung tissue samples by quantitating FF, interstitial mononuclear inflammation, and intra-alveolar macrophages with a deep convolutional neural network (CNN). Lung tissue samples of 71 patients with IPF from the FinnishIPF registry were analyzed by an AI model developed in the Aiforia® platform. The model was trained to detect tissue, air spaces, FF, interstitial mononuclear inflammation, and intra-alveolar macrophages with 20 samples. For survival analysis, cut-point values for high and low values of histological parameters were determined with maximally selected rank statistics. Survival was analyzed using the Kaplan-Meier method. A large area of FF predicted poor prognosis in IPF (p = 0.01). High numbers of interstitial mononuclear inflammatory cells and intra-alveolar macrophages were associated with prolonged survival (p = 0.01 and p = 0.01, respectively). Of lung function values, low diffusing capacity for carbon monoxide was connected to a high density of FF (p = 0.03) and a high forced vital capacity of predicted was associated with a high intra-alveolar macrophage density (p = 0.03). The deep CNN detected histological features that are difficult to quantitate manually. Interstitial mononuclear inflammation and intra-alveolar macrophages were novel prognostic histological biomarkers in IPF. Evaluating histological features with AI provides novel information on the prognostic estimation of IPF.
Collapse
Affiliation(s)
- Kati Mäkelä
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki and Heart and Lung Center, Helsinki University Hospital, FI-00290, Helsinki, Finland.
| | - Mikko I Mäyränpää
- Pathology, University of Helsinki and Helsinki University Hospital, FI-00290, Helsinki, Finland
| | | | - Paula Bergman
- Biostatistics Consulting, Department of Public Health, University of Helsinki and Helsinki University Hospital, FI-00290, Helsinki, Finland
| | - Eva Sutinen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki and Heart and Lung Center, Helsinki University Hospital, FI-00290, Helsinki, Finland
| | - Hely Ollila
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki and Heart and Lung Center, Helsinki University Hospital, FI-00290, Helsinki, Finland
| | - Riitta Kaarteenaho
- Research Unit of Internal Medicine, University of Oulu and Medical Research Center Oulu, Oulu University Hospital, FI-90014, Oulu, Finland
| | - Marjukka Myllärniemi
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki and Heart and Lung Center, Helsinki University Hospital, FI-00290, Helsinki, Finland
| |
Collapse
|
19
|
Heinemann F, Birk G, Stierstorfer B. Deep learning enables pathologist-like scoring of NASH models. Sci Rep 2019; 9:18454. [PMID: 31804575 PMCID: PMC6895116 DOI: 10.1038/s41598-019-54904-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and the progressive form of non-alcoholic steatohepatitis (NASH) are diseases of major importance with a high unmet medical need. Efficacy studies on novel compounds to treat NAFLD/NASH using disease models are frequently evaluated using established histological feature scores on ballooning, inflammation, steatosis and fibrosis. These features are assessed by a trained pathologist using microscopy and assigned discrete scores. We demonstrate how to automate these scores with convolutional neural networks (CNNs). Whole slide images of stained liver sections are analyzed using two different scales with four CNNs, each specialized for one of four histopathological features. A continuous value is obtained to quantify the extent of each feature, which can be used directly to provide a high resolution readout. In addition, the continuous values can be mapped to obtain the established discrete pathologist-like scores. The automated deep learning-based scores show good agreement with the trainer - a human pathologist.
Collapse
Affiliation(s)
- Fabian Heinemann
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riß, Germany.
| | - Gerald Birk
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riß, Germany
| | - Birgit Stierstorfer
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riß, Germany
| |
Collapse
|
20
|
Turner OC, Aeffner F, Bangari DS, High W, Knight B, Forest T, Cossic B, Himmel LE, Rudmann DG, Bawa B, Muthuswamy A, Aina OH, Edmondson EF, Saravanan C, Brown DL, Sing T, Sebastian MM. Society of Toxicologic Pathology Digital Pathology and Image Analysis Special Interest Group Article*: Opinion on the Application of Artificial Intelligence and Machine Learning to Digital Toxicologic Pathology. Toxicol Pathol 2019; 48:277-294. [DOI: 10.1177/0192623319881401] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Toxicologic pathology is transitioning from analog to digital methods. This transition seems inevitable due to a host of ongoing social and medical technological forces. Of these, artificial intelligence (AI) and in particular machine learning (ML) are globally disruptive, rapidly growing sectors of technology whose impact on the long-established field of histopathology is quickly being realized. The development of increasing numbers of algorithms, peering ever deeper into the histopathological space, has demonstrated to the scientific community that AI pathology platforms are now poised to truly impact the future of precision and personalized medicine. However, as with all great technological advances, there are implementation and adoption challenges. This review aims to define common and relevant AI and ML terminology, describe data generation and interpretation, outline current and potential future business cases, discuss validation and regulatory hurdles, and most importantly, propose how overcoming the challenges of this burgeoning technology may shape toxicologic pathology for years to come, enabling pathologists to contribute even more effectively to answering scientific questions and solving global health issues. [Box: see text]
Collapse
Affiliation(s)
- Oliver C. Turner
- Novartis, Novartis Institutes for Biomedical Research, Preclinical Safety, East Hanover, NJ, USA
| | - Famke Aeffner
- Amgen Inc, Research, Comparative Biology and Safety Sciences, San Francisco, CA, USA
| | | | - Wanda High
- High Preclinical Pathology Consulting, Rochester, NY, USA
| | - Brian Knight
- Boehringer Ingelheim Pharmaceuticals Incorporated, Nonclinical Drug Safety, Ridgefield, CT, USA
| | | | - Brieuc Cossic
- Roche, Pharmaceutical Research and Early Development (pRED), Roche Innovation Center, Basel, Switzerland
| | - Lauren E. Himmel
- Division of Animal Care, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | - Elijah F. Edmondson
- Pathology/Histotechnology Laboratory, Frederick National Laboratory for Cancer Research, NIH, Frederick, MD, USA
| | - Chandrassegar Saravanan
- Novartis, Novartis Institutes for Biomedical Research, Preclinical Safety, Cambridge, MA, USA
| | | | - Tobias Sing
- Novartis, Novartis Institutes for Biomedical Research, NIBR Informatics, Basel, Switzerland
| | - Manu M. Sebastian
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA
| |
Collapse
|