1
|
Tornifoglio B, Hughes C, Digeronimo F, Guendouz Y, Johnston RD, Lally C. Imaging the microstructure of the arterial wall - ex vivo to in vivo potential. Acta Biomater 2025:S1742-7061(25)00346-0. [PMID: 40348073 DOI: 10.1016/j.actbio.2025.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Microstructural imaging enables researchers to visualise changes in the arterial wall, allowing for (i) a deeper understanding of the role of specific components in arterial mechanics, (ii) the observation of cellular responses, (iii) insights into pathological alterations in tissue microstructure, and/or (iv) advancements in tissue engineering aimed at replicating healthy native tissue. In this prospective review, we present various imaging modalities spanning from ex vivo to in vivo applications within arterial tissue. The pros, cons, and sensitivities of these modalities are highlighted. By consolidating the latest advancements in microstructural imaging of arterial tissue, the authors aim for this paper to serve as a guide for researchers designing experiments at various stages. Furthermore, the integration of non-invasive, non-destructive imaging techniques into studies provides an additional layer of microstructural information, enhancing scientific findings, improving our understanding of disease, and potentially enabling earlier or more effective diagnostic capabilities. STATEMENT OF SIGNIFICANCE: Imaging the specific microstructural components of the arterial wall provides critical insights into vascular biology, mechanics, and pathology. It enables the visualisation of key structural components and their roles in arterial function, supports the analysis of cell-matrix interactions, and reveals microarchitectural changes associated with disease progression. This level of specificity also informs the design of biomimetic materials and scaffolds in tissue engineering, facilitating the replication of native arterial properties. By synthesising recent developments in microstructural imaging techniques, this paper serves as a reference for investigators designing experiments across a range of vascular research applications. Moreover, the incorporation of non-invasive, non-destructive imaging methods offers a means to acquire detailed microstructural data without compromising tissue integrity. This enhances the interpretability and translational potential of findings, deepens our understanding of vascular disease mechanisms, and may ultimately contribute to the development of earlier and more precise diagnostic approaches.
Collapse
Affiliation(s)
- B Tornifoglio
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland.
| | - C Hughes
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - F Digeronimo
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Y Guendouz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - R D Johnston
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - C Lally
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Ireland.
| |
Collapse
|
2
|
Yang Y, Mao HM, Huang SG, Guo WL. A magnetic resonance image-based deep learning radiomics nomogram for hepatocyte cytokeratin 7 expression: application to predict cholestasis progression in children with pancreaticobiliary maljunction. Pediatr Radiol 2025; 55:1164-1177. [PMID: 40186654 DOI: 10.1007/s00247-025-06225-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Hepatocyte cytokeratin 7 (CK7) is a reliable marker for evaluating the severity of cholestasis in chronic cholestatic cholangiopathies. However, there is currently no noninvasive test available to assess the status of hepatocyte CK7 in pancreaticobiliary maljunction patients. OBJECTIVE We aimed to develop a deep learning radiomics nomogram using magnetic resonance images (MRIs) to preoperatively identify the hepatocyte CK7 status and assess cholestasis progression in patients with pancreaticobiliary maljunction. MATERIALS AND METHODS In total, 180 pancreaticobiliary maljunction patients were retrospectively enrolled and were randomly divided into a training cohort (n = 144) and a validation cohort (n = 36). CK7 status was determined through immunohistochemical analysis. Pyradiomics and pretrained ResNet50 were used to extract radiomics and deep learning features, respectively. To construct the radiomics and deep learning signature, feature selection methods including the minimum redundancy-maximum relevance and least absolute shrinkage and selection operator were employed. The integrated deep learning radiomics nomogram model was constructed by combining the imaging signatures and valuable clinical feature. RESULTS The deep learning signature exhibited superior predictive performance compared with the radiomics signature, as evidenced by the higher area under the curve (AUC) values in validation cohort (0.92 vs. 0.81). Further, the deep learning radiomics nomogram, which incorporated the radiomics signature, deep learning signature, and Komi classification, demonstrated excellent predictive ability for CK7 expression, with AUC value of 0.95 in the validation cohort. CONCLUSION The proposed deep learning radiomics nomogram exhibits promising performance in accurately identifying hepatic CK7 expression, thus facilitating prediction of cholestasis progression and perhaps earlier initiation of treatment in pancreaticobiliary maljunction children.
Collapse
Affiliation(s)
- Yang Yang
- Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu Province, 215028, China
| | - Hui-Min Mao
- Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu Province, 215028, China
| | - Shun-Gen Huang
- Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu Province, 215028, China
| | - Wan-Liang Guo
- Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu Province, 215028, China.
| |
Collapse
|
3
|
Guareschi EE, Nicholls PK, Tobe SS, Magni PA. Taphonomy and diagenesis of submerged bone: An experimental approach. Forensic Sci Int 2025; 370:112416. [PMID: 40054340 DOI: 10.1016/j.forsciint.2025.112416] [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: 01/14/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 05/02/2025]
Abstract
Bone taphonomy and diagenesis contribute to anthropological analysis in forensic investigations by attempting to reconstruct the relationship between human cadaveric remains and their postmortem depositional environment. The rare aquatic taphonomic experiments have been delivering conflicting results on the influence of time and the environment on the decay of bone and teeth, especially considering that the main diagenetic processes can lead to fragmentation, progressive dissolution or fossilization. The aim of this experimental, quantitative, randomized and controlled 2-year study was to analyse the taphonomy and diagenesis of submerged terrestrial mammalian bones to achieve a more accurate estimation of both the post-mortem interval (PMI) and the post-mortem submersion interval (PMSI) in the short term. Three parameters of bone diagenesis, the Oxford Histological Index (OHI), the total porosity and the collagen content of cortical bone were analysed by MicroCT Scan, bright-field Light Microscopy (Picrosirius Red stain), Scanning Electron Microscopy (SEM) and Laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) on 75 sheep femurs and tibias placed in four distinct types of environment (natural saltwater, natural freshwater, an artificial seawater solution and exposed to the air) vs. non-exposed controls. LA-ICP-MS was soon discontinued because no measurable changes of the elemental profiles could be detected. Multivariate statistical analysis was applied to the collected data. The macroscopical preservation was consistently excellent (OHI=5). The total porosity and the degradation of collagen were greater underwater than in subaerial exposure, whereas demineralization zones and bioerosion tunnelling appeared after 12 months in the air-exposed samples only. Underwater, the continuous movement, the correlated abrasion by sand and sediment and the constant alkaline pH (≥ 8) can explain the progressive removal of the mineral component and the subsequent exposure of collagen to bioeroders and chemical hydrolysis. On land, the same process occurs at a slower rate on account of the seasonality of the water flow, however, the action of the more abundant and diversified species of bioeroding microorganisms appears more efficient. Despite some limitations, this study indicates that three parameters of bone diagenesis can predict the depositional environment of terrestrial mammalian bone characterized by a PMI and/or PMSI of at least 12 months.
Collapse
Affiliation(s)
- Edda E Guareschi
- Curtin Medical School, Curtin University, Bentley, Western Australia 6102, Australia; Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, Western Australia 6150, Australia.
| | - Philip K Nicholls
- Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, Western Australia 6150, Australia
| | - Shanan S Tobe
- Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, Western Australia 6150, Australia
| | - Paola A Magni
- Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, Western Australia 6150, Australia; The UWA Oceans Institute and School of Engineering, The University of Western Australia, Perth, Western Australia 6009, Australia
| |
Collapse
|
4
|
Bertacchi M, Theiß S, Ahmed A, Eibl M, Loubat A, Maharaux G, Phromkrasae W, Chakrabandhu K, Camgöz A, Antonaci M, Schaaf CP, Studer M, Laugsch M. Unravelling the conundrum of nucleolar NR2F1 localization using antibody-based approaches in vitro and in vivo. Commun Biol 2025; 8:594. [PMID: 40204944 PMCID: PMC11982218 DOI: 10.1038/s42003-025-07985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
As a transcription factor, NR2F1 regulates spatiotemporal gene expression in the nucleus particularly during development. Aberrant NR2F1 causes the rare neurodevelopmental disorder Bosch-Boonstra-Schaaf Optic Atrophy Syndrome. In addition, altered NR2F1 expression is frequently observed in various cancers and is considered a prognostic marker or potential therapeutic target. NR2F1 has been found in both the nucleus and nucleoli, suggesting a non-canonical and direct role in the latter compartment. Hence, we studied this phenomenon employing various in vitro and in vivo models using different antibody-dependent approaches. Examination of seven commonly used anti-NR2F1 antibodies in different human cancer and stem cells as well as in wild type and null mice revealed that NR2F1 nucleolar localization is artificial and has no functional role. Our subsequent comparative analysis demonstrated which anti-NR2F1 antibody best fits which approach. The data allow for correct data interpretation and underline the need to optimize any antibody-mediated technique.
Collapse
Affiliation(s)
- Michele Bertacchi
- Université Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose (iBV), 06108, Nice, France.
| | - Susanne Theiß
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Ayat Ahmed
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Michael Eibl
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Agnès Loubat
- Université Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose (iBV), 06108, Nice, France
| | - Gwendoline Maharaux
- Université Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose (iBV), 06108, Nice, France
| | - Wanchana Phromkrasae
- Université Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose (iBV), 06108, Nice, France
| | - Krittalak Chakrabandhu
- Université Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose (iBV), 06108, Nice, France
| | - Aylin Camgöz
- Hopp Children's Cancer Center (KITZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Marco Antonaci
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | | | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose (iBV), 06108, Nice, France
| | - Magdalena Laugsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
5
|
Smallridge MW, Aktepe TE, Coppo MJC, Vaz PK, Diaz-Méndez A, Murray CM, Segal G, Devlin JM, Hartley CA. Three-dimensional exploration of the chicken embryo, a comparative study of light sheet and histological visualisation. PLoS One 2025; 20:e0320483. [PMID: 40168291 PMCID: PMC11960958 DOI: 10.1371/journal.pone.0320483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
Ultramicroscopy has offered new avenues into the visualisation of tissues within animal models, providing three-dimensional visualisation through the use of light sheet fluorescence microscopy. This study aimed to develop and apply an optical tissue clearing method to investigate the application of light sheet fluorescence microscopy to image late-stage chicken embryos, and compare anatomical visualisation to traditional histological staining. Seventeen-day old specific pathogen free embryos were collected, fixed, and sectioned. Haematoxylin and eosin stained sections were prepared for histology, while light sheet imaging required the tissues to be optically clear. For this, an ethyl cinnamate-based method was utilised, allowing for acquisition of clear, unobstructed three-dimensional images of significant organ structures and systems using only autofluorescence. The use of established histological techniques provided anatomical mapping of structures between familiar histology images and the three-dimensional light sheet images. Rendering of organs using light sheet imaging provided contextual insights into the surrounding tissues and physiological architecture of major organ structures and systems. This was most apparent through the identification of the pulmonary vein and rendering of a volumetric projection of the vasculature branching within the lung and the subsequent merging of vasculature into the left side of the heart. Overall, the visualisation of the chicken embryo was enhanced by combining traditional histology with the information gained by three-dimensional light sheet fluorescence microscopy.
Collapse
Affiliation(s)
- M. W. Smallridge
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
| | - T. E. Aktepe
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
| | - M. J. C. Coppo
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
- Facultad de Ciencias de la Vida, Escuela de Medicina Veterinaria, Universidad Andres Bello, Concepcion, Biobio, Chile
| | - P. K. Vaz
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
| | - A. Diaz-Méndez
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
| | - C. M. Murray
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
| | - G. Segal
- Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Victoria, Australia
| | - J. M. Devlin
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
| | - C. A. Hartley
- Faculty of Science, Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, The University of Melbourne, Melbourne, Victoria, Australia,
| |
Collapse
|
6
|
Tkachev S, Brosalov V, Kit O, Maksimov A, Goncharova A, Sadyrin E, Dalina A, Popova E, Osipenko A, Voloshin M, Karnaukhov N, Timashev P. Unveiling Another Dimension: Advanced Visualization of Cancer Invasion and Metastasis via Micro-CT Imaging. Cancers (Basel) 2025; 17:1139. [PMID: 40227647 PMCID: PMC11988112 DOI: 10.3390/cancers17071139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Invasion and metastasis are well-known hallmarks of cancer, with metastatic disease accounting for 60% to 90% of cancer-related deaths [...].
Collapse
Affiliation(s)
- Sergey Tkachev
- Institute for Regenerative Medicine, Sechenov University, 119992 Moscow, Russia
| | | | - Oleg Kit
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia
| | - Alexey Maksimov
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia
| | - Anna Goncharova
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia
| | - Evgeniy Sadyrin
- Laboratory of Mechanics of Biocompatible Materials, Don State Technical University, 344003 Rostov-on-Don, Russia
| | - Alexandra Dalina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Elena Popova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, 115682 Moscow, Russia
| | - Anton Osipenko
- Department of Pharmacology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Mark Voloshin
- A.S. Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Nikolay Karnaukhov
- A.S. Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
- Institute of Clinical Morphology and Digital Pathology, Sechenov University, 119991 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 119992 Moscow, Russia
| |
Collapse
|
7
|
Al-Jawadri AMH, Karami Z, Haririan I, Akrami M, Gholami M. Development of a bio-inspired phagocytic stable nanoghost with anti-inflammatory properties for management of inflammation in ulcerative colitis. J Drug Target 2025:1-15. [PMID: 40022643 DOI: 10.1080/1061186x.2025.2474644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND New bio-mimetic approaches are needed to develop effective delivery systems for inflammation regulation in chronic diseases like ulcerative colitis, avoiding fast clearance by immune system. The cell membrane-coated nanoparticle with a therapeutic payload has been considered as a promising delivery system to address the requirement. METHODS Here, Glibenclamide (GLY)-loaded PLGA nanoparticles (NPs) were constructed by a single emulsion procedure and camouflaged by a layer of monocyte membrane using the extrusion technique to fabricate bio-mimetic nanoghosts (NGs), followed by physiochemical and biological characterizations. RESULTS Upon coating the NPs by the membrane, the hydrodynamic size and zeta potential of NGs was changed. The formation of the shell compartment with diameter of about 15.5nm around NP core was confirmed by TEM. The expression levels of NLRP3, IL-1β, IL-18, caspase-1, TNF-α and IL-6 were decreased upon the NGs treatment. The lower cellular internalization of the NGs exhibited potential for improved circulation stability against macrophage phagocytosis. Treatment of acetic acid-induced UC with NGs exhibited healing of the mucosal lining in the colon tissue. CONCLUSION The monocyte membrane-coated NPs with a sulfonylurea derivatives payload can be considered as an excellent biologically inspired candidate for management of inflammatory diseases like UC via inflammation regulation.
Collapse
Affiliation(s)
| | - Zahra Karami
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Akrami
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research, Tehran, Iran
| |
Collapse
|
8
|
Kirya P, Mestre‐Farrera A, Yang J, Poulikakos LV. Leveraging Optical Anisotropy of the Morpho Butterfly Wing for Quantitative, Stain-Free, and Contact-Free Assessment of Biological Tissue Microstructures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407728. [PMID: 39811986 PMCID: PMC11937990 DOI: 10.1002/adma.202407728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/27/2024] [Indexed: 01/16/2025]
Abstract
Changes in the density and organization of fibrous biological tissues often accompany the progression of serious diseases ranging from fibrosis to neurodegenerative diseases, heart disease and cancer. However, challenges in cost, complexity, or precision faced by existing imaging methodologies and materials pose barriers to elucidating the role of tissue microstructure in disease. Here, we leverage the intrinsic optical anisotropy of the Morpho butterfly wing and introduce Morpho-Enhanced Polarized Light Microscopy (MorE-PoL), a stain- and contact-free imaging platform that enhances and quantifies the birefringent material properties of fibrous biological tissues. We develop a mathematical model, based on Jones calculus, which describes fibrous tissue density and organization. As a representative example, we analyzed collagen-dense and collagen-sparse human breast cancer tissue sections and leverage our technique to assess the microstructural properties of distinct regions of interest. We compare our results with conventional Hematoxylin and Eosin (H&E) staining procedures and second harmonic generation (SHG) microscopy for fibrillar collagen detection. Our findings demonstrate that our MorE-PoL technique provides a robust, quantitative, and accessible route toward analyzing biological tissue microstructures, with great potential for application to a broad range of biological materials.
Collapse
Affiliation(s)
- Paula Kirya
- Department of Mechanical and Aerospace EngineeringProgram of Materials Science and EngineeringUniversity of California San Diego9500 Gilman DriveLa JollaCA92093USA
| | - Aida Mestre‐Farrera
- Department of PharmacologyMoores Cancer CenterUniversity of California San Diego3855 Health Sciences DriveLa JollaCA92093USA
- Department of PediatricsUniversity of California San Diego9500 Gilman DriveLa JollaCA92093USA
| | - Jing Yang
- Department of PharmacologyMoores Cancer CenterUniversity of California San Diego3855 Health Sciences DriveLa JollaCA92093USA
- Department of PediatricsUniversity of California San Diego9500 Gilman DriveLa JollaCA92093USA
| | - Lisa V. Poulikakos
- Department of Mechanical and Aerospace EngineeringProgram of Materials Science and EngineeringUniversity of California San Diego9500 Gilman DriveLa JollaCA92093USA
| |
Collapse
|
9
|
Upadhye AR, Cintron E, Zhang J, Coleman J, Kolluru C, Jenkins MW, Wilson D, Pelot NA, Shoffstall AJ. Phosphotungstic Acid Staining to Visualize the Vagus Nerve Perineurium Using Micro-CT. J Neuroimaging 2025; 35:e70040. [PMID: 40207700 PMCID: PMC11984074 DOI: 10.1111/jon.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Peripheral nerve stimulation is approved by the US Food and Drug Administration for treating various disorders, but it is often limited by side effects, highlighting the need for a clear understanding of fascicular and fiber organization to design selective therapies. Micro-CT imaging of contrast-stained nerves enables the visualization of tissue microstructures, such as the fascicular perineurium and vasculature. In this work, we evaluated phosphotungstic acid (PTA) as a contrast agent and assessed its compatibility with downstream histology. METHODS Human vagus nerve samples were collected from three embalmed cadavers and subjected to three different staining methods, followed by micro-CT imaging: Lugol's iodine, osmium tetroxide, and PTA. Contrast ratios of adjacent tissue microstructures (perineurium, interfascicular epineurium, and fascicle) were quantified for each stain and compared. We further developed a pipeline to optimize micro-CT scan acquisition parameters based on objective metrics for sharpness, noise, and pixel saturation. The PTA-stained samples underwent subsequent histological processing and staining with hematoxylin and eosin, Masson's trichrome, and immunohistochemistry and were assessed for tissue degradation. RESULTS PTA enhanced the visualization of perineurium, providing high contrast ratios compared to iodine and osmium tetroxide. Optimized scanning parameters for PTA-stained nerves (55 kV and 109 µA) effectively balanced noise and sharpness. While we found that PTA is generally nondestructive for downstream histology, higher concentrations and longer exposure could alter the optical density of nuclei and affect stain differentiation in special stains. CONCLUSION PTA serves as a valuable micro-CT contrast agent for nerve imaging, effective in visualizing the perineurium with minimal impact on histological integrity.
Collapse
Affiliation(s)
- Aniruddha R. Upadhye
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
- APT CenterLouis Stokes Cleveland VA Medical CenterClevelandOhioUSA
| | - Eleana Cintron
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Jichu Zhang
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Jennifer Coleman
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Chaitanya Kolluru
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Michael W. Jenkins
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - David Wilson
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Nicole A. Pelot
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Andrew J. Shoffstall
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
- APT CenterLouis Stokes Cleveland VA Medical CenterClevelandOhioUSA
| |
Collapse
|
10
|
Kurelac I, Sollazzo M, De Luise M, Nanetti F, Lanteri L, D’Angelo L, Cavina B, Corrà S, Miglietta S, Milioni S, Luppi E, Iommarini L, Di Costanzo S, Ricciardi AM, Coluccelli S, Maloberti T, Grillini M, Coadă CA, Perrone AM, De Iaco P, de Biase D, Ragazzi M, Gasparre G, Porcelli AM. Immunomagnetic enrichment coupled to PAX8/TP53 molecular pathology approach increases sensitivity in the detection of ovarian cancer cells in ascites. Front Mol Biosci 2025; 12:1537407. [PMID: 40051502 PMCID: PMC11882402 DOI: 10.3389/fmolb.2025.1537407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is one of the deadliest malignancies in female population and the cause of 70% of all ovarian cancer-related deaths. Among its hallmarks, the fluid accumulation in the peritoneal cavity, or ascites, is a peculiar pathological sign during late stages and in recurrent patients. Besides cancer cells, ascitic fluids contain a heterogeneous cellular composition, representing a precious source to dissect molecular mechanisms underlying invasion and metastatization or find new biomarkers to predict therapy response. However, malignant cells are often a minority population in ascites making the detection and analysis of cancer cells a challenge. Here we propose a combinatorial approach for the detection of malignant cells in OC ascites based on TP53 deep sequencing and PAX8 cytological staining. In addition, we improve the procedure by implementing a cancer cell enrichment step, increasing the sensitivity in the detection of neoplastic fraction and potentiating downstream research and diagnostics applications.
Collapse
Affiliation(s)
- Ivana Kurelac
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Manuela Sollazzo
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
- Centre for Applied Biomedical Research, University of Bologna, Bologna, Italy
| | - Monica De Luise
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesca Nanetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Laura Lanteri
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Luigi D’Angelo
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Beatrice Cavina
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Simona Corrà
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Stefano Miglietta
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Sara Milioni
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Elena Luppi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Luisa Iommarini
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
- Centre for Applied Biomedical Research, University of Bologna, Bologna, Italy
| | - Stella Di Costanzo
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Sara Coluccelli
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Marco Grillini
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Camelia Alexandra Coadă
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Anna Myriam Perrone
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pierandrea De Iaco
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Moira Ragazzi
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Giuseppe Gasparre
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Centre for Applied Biomedical Research, University of Bologna, Bologna, Italy
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche, University of Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| |
Collapse
|
11
|
Juybari J, Hamilton J, Chen C, Khalil A, Zhu Y. Context-guided segmentation for histopathologic cancer segmentation. Sci Rep 2025; 15:5404. [PMID: 39948139 PMCID: PMC11825859 DOI: 10.1038/s41598-025-86428-7] [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: 07/30/2024] [Accepted: 01/10/2025] [Indexed: 02/16/2025] Open
Abstract
Microscopic inspection of histologically stained tissue is considered as the gold standard for cancer diagnosis. This research is inspired by the practices of pathologists who analyze diagnostic samples by zooming in and out. We propose a dual-encoder model that simultaneously evaluates two views of the tissue at different levels of magnification. The lower magnification view provides contextual information for a target area, while the higher magnification view provides detailed information. The model consists of two encoder branches that consider both detail and context resolutions of the target area concurrently for binary pixel-wise segmentation. We introduce a unique weight initialization for the cross-attention between the context and detail feature tensors, allowing the model to incorporate contextual information. Our design is evaluated using the Camelyon16 dataset of sentinel lymph node tissue and cancer. The results demonstrate the benefit of including context regions when segmenting for cancer, with an improvement in AUC ranging from 0.31 to 0.92% and an improvement in cancer Dice score ranging from 4.09% to 6.81% compared to single detailed input models.
Collapse
Affiliation(s)
- Jeremy Juybari
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, 04469, USA
- DEAL Lab, Department of Electrical and Computer Engineering, University of Maine, Orono, 04469, USA
| | - Josh Hamilton
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, 04469, USA
| | - Chaofan Chen
- School of Computing and Information Science, University of Maine, Orono, 04469, USA
| | - Andre Khalil
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, 04469, USA
| | - Yifeng Zhu
- DEAL Lab, Department of Electrical and Computer Engineering, University of Maine, Orono, 04469, USA.
| |
Collapse
|
12
|
Mandal S, Motganhalli Ravikumar R, Tannert A, Urbanek A, Guliev RR, Naumann M, Coldewey SM, Dahmen U, Carvalho L, Bastião Silva L, Neugebauer U. Qualitative comparison of decalcifiers for mouse bone cryosections for subsequent biophotonic analysis. Sci Rep 2025; 15:1153. [PMID: 39774725 PMCID: PMC11707355 DOI: 10.1038/s41598-024-84330-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Bone tissue, with its complex structure, often necessitates decalcification of the hard tissue for ex vivo morphological studies. The choice of a suitable decalcification method plays a crucial role in preserving desired features and ensuring compatibility with diverse imaging techniques. The search for a universal decalcification method that is suitable for a range of biophotonic analyses remains an ongoing challenge. In this study, we systematically assessed five standard bone decalcification protocols, encompassing strong mineralic acids (3% and 5% nitric acid), a commercially available formulation of hydrochloric and formic acid), as well as weak organic acids (5% trichloroacetic acid and 8% formic acid), and a chelating agent (25% ethylenediamine-tetraacetic acid) with varying decalcification durations, using mouse long bones as our experimental model. Our imaging analysis panel included classical histological staining (Hematoxylin and Eosin, H&E), immunofluorescence staining, and label-free Raman microspectroscopic imaging. We used cryosections instead of paraffin sections since paraffin interferes with tissue Raman signals. This approach is not as commonly used as it is more prone to handling artifacts, but is the preferred method for subsequent Raman analysis. Decalcification efficacy was evaluated based on various qualitative and some quantitative imaging parameters by 2-3 independent observers. Our systematic approach revealed that the chelating agent, when used for 24 h, optimally preserved bone features and, thus, would be the ideal decalcifying agent for comprehensive subsequent analysis. However, the choice of decalcifier and the ideal decalcification duration may vary depending on the type and thickness of bone, necessitating tailored adjustments to meet specific experimental requirements.
Collapse
Affiliation(s)
- Shibarjun Mandal
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745, Jena, Germany
| | - Ramya Motganhalli Ravikumar
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745, Jena, Germany
| | - Astrid Tannert
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Annett Urbanek
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745, Jena, Germany
| | - Rustam R Guliev
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745, Jena, Germany
| | - Max Naumann
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Sina M Coldewey
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747, Jena, Germany
- Septomics Research Center, Jena University Hospital, 07745, Jena, Germany
| | - Uta Dahmen
- Experimental Surgery, Clinic for General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany
| | - Lina Carvalho
- Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, Coimbra, 3004-504, Portugal
| | | | - Ute Neugebauer
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745, Jena, Germany.
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany.
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743, Jena, Germany.
| |
Collapse
|
13
|
Daly S, Bulovaite E, Handa A, Morris K, Muresan L, Adams C, Kaizuka T, Kitching A, Spark A, Chant G, O′Holleran K, Grant SGN, Horrocks MH, Lee SF. 3D Super-Resolution Imaging of PSD95 Reveals an Abundance of Diffuse Protein Supercomplexes in the Mouse Brain. ACS Chem Neurosci 2025; 16:40-51. [PMID: 39702971 PMCID: PMC11697326 DOI: 10.1021/acschemneuro.4c00684] [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: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
PSD95 is an abundant scaffolding protein that assembles multiprotein complexes controlling synaptic physiology and behavior. Confocal microscopy has previously shown that PSD95 is enriched in the postsynaptic terminals of excitatory synapses and two-dimensional (2D) super-resolution microscopy further revealed that it forms nanoclusters. In this study, we utilized three-dimensional (3D) super-resolution microscopy to examine the nanoarchitecture of PSD95 in the mouse brain, characterizing the spatial arrangement of over 8 million molecules. While we were able to identify molecular arrangements that have been previously reported, imaging in 3D allowed us to classify these with higher accuracy. Furthermore, 3D super-resolution microscopy enabled the quantification of protein levels, revealing that an abundance of PSD95 molecules existed outside of synapses as a diffuse population of supercomplexes, containing multiple copies of PSD95. Further analysis of the supercomplexes containing two units identified two populations: one that had PSD95 molecules separated by 39 ± 2 nm, and a second with a separation of 94 ± 27 nm. The finding that there exists supercomplexes containing two PSD95 units outside of the synapse suggests that supercomplexes containing multiple protein copies assemble outside the synapse and then integrate into the synapse to form a supramolecular nanocluster architecture.
Collapse
Affiliation(s)
- Sam Daly
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - Edita Bulovaite
- Genes
to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, U.K.
| | - Anoushka Handa
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - Katie Morris
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | - Leila Muresan
- Cambridge
Advanced Imaging Centre, University of Cambridge, Cambridge CB2 3DY, U.K.
| | - Candace Adams
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | - Takeshi Kaizuka
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | | | | | - Gregory Chant
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - Kevin O′Holleran
- Cambridge
Advanced Imaging Centre, University of Cambridge, Cambridge CB2 3DY, U.K.
- ZOMP, Maxwell
Centre, JJ Thomson Avenue, Cambridge CB3 0HE, U.K.
| | - Seth G. N. Grant
- Genes
to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, U.K.
| | - Mathew H. Horrocks
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | - Steven F. Lee
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| |
Collapse
|
14
|
Walia A, Ortmann AJ, Lefler S, Holden TA, Puram SV, Herzog JA, Buchman CA. Electrocochleography-Based Tonotopic Map: I. Place Coding of the Human Cochlea With Hearing Loss. Ear Hear 2025; 46:253-264. [PMID: 39233326 PMCID: PMC11649476 DOI: 10.1097/aud.0000000000001579] [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] [Indexed: 09/06/2024]
Abstract
OBJECTIVES Due to the challenges of direct in vivo measurements in humans, previous studies of cochlear tonotopy primarily utilized human cadavers and animal models. This study uses cochlear implant electrodes as a tool for intracochlear recordings of acoustically evoked responses to achieve two primary goals: (1) to map the in vivo tonotopy of the human cochlea, and (2) to assess the impact of sound intensity and the creation of an artificial "third window" on this tonotopic map. DESIGN Fifty patients with hearing loss received cochlear implant electrode arrays. Postimplantation, pure-tone acoustic stimuli (0.25 to 4 kHz) were delivered, and electrophysiological responses were recorded from all 22 electrode contacts. The analysis included fast Fourier transformation to determine the amplitude of the first harmonic, indicative of predominantly outer hair cell activity, and tuning curves to identify the best frequency (BF) electrode. These measures, coupled with postoperative imaging for precise electrode localization, facilitated the construction of an in vivo frequency-position function. The study included a specific examination of 2 patients with auditory neuropathy spectrum disorder (ANSD), with preserved cochlear function as assessed by present distortion-product otoacoustic emissions, to determine the impact of sound intensity on the frequency-position map. In addition, the electrophysiological map was recorded in a patient undergoing a translabyrinthine craniotomy for vestibular schwannoma removal, before and after creating an artificial third window, to explore whether an experimental artifact conducted in cadaveric experiments, as was performed in von Békésy landmark experiments, would produce a shift in the frequency-position map. RESULTS A significant deviation from the Greenwood model was observed in the electrophysiological frequency-position function, particularly at high-intensity stimulations. In subjects with hearing loss, frequency tuning, and BF location remained consistent across sound intensities. In contrast, ANSD patients exhibited Greenwood-like place coding at low intensities (~40 dB SPL) and a basal shift in BF location at higher intensities (~70 dB SPL or greater). Notably, creating an artificial "third-window" did not alter the frequency-position map. CONCLUSIONS This study successfully maps in vivo tonotopy of human cochleae with hearing loss, demonstrating a near-octave shift from traditional frequency-position maps. In patients with ANSD, representing more typical cochlear function, intermediate intensity levels (~70 to 80 dB SPL) produced results similar to high-intensity stimulation. These findings highlight the influence of stimulus intensity on the cochlear operational point in subjects with hearing loss. This knowledge could enhance cochlear implant programming and improve auditory rehabilitation by more accurately aligning electrode stimulation with natural cochlear responses.
Collapse
Affiliation(s)
- Amit Walia
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| | - Amanda J. Ortmann
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| | - Shannon Lefler
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| | - Timothy A. Holden
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| | - Sidharth V. Puram
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| | - Jacques A. Herzog
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| | - Craig A. Buchman
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| |
Collapse
|
15
|
Orellana F, Grassi A, Nuss KM, Wahl P, Neels A, Zaffagnini S, Parrilli A. Spatial and temporal evaluation of iodine uptake and radiodensity in meniscus tissue using contrast-enhanced micro-CT. Heliyon 2024; 10:e41080. [PMID: 39759317 PMCID: PMC11696653 DOI: 10.1016/j.heliyon.2024.e41080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Rationale and objective The visualization of soft tissues, like the meniscus, through X-ray micro-computed tomography (micro-CT), requires the use of contrast agents (CAs). While other studies have investigated CA diffusion in fibrocartilagineous tissues, this work aimed to optimize iodine staining protocols for meniscal tissue that improve their visualization by micro-CT. Specific objectives included evaluating the diffusion of CAs within meniscal samples over time, assessing volume changes due to staining, and identifying the iodine ions absorbed by the tissue. Materials and methods Water-based and PBS-based Lugol solutions (KI3) were used to stain sheep and pig menisci for 24 days. Samples were scanned using micro-CT at different time points (0, 1, 4, 8, 12, 16, 20, and 24 days) to monitor CA diffusion and volume changes. Micro-CT provided three-dimensional (3D) visualization of iodine distribution and quantification of volume changes and radiodensity in the menisci. Additionally, UV-visible spectroscopy (UV-vis) analyses were performed to determine the uptake of iodine ions by the meniscus. Results Results indicated volumetric shrinkage and increased radiodensity within the first days of staining, with diffusion primarily occurring from the periphery of the meniscus. UV-visible spectroscopy identified two iodide ions in the CA solution (I- and I3 -) and revealed a preferential absorption of the triiodide ion (I3 -). Conclusion This study demonstrated the utility of iodine-based CAs and micro-CT technique for visualizing and investigating the spatial and temporal iodine diffusion within the meniscal tissue of sheep and pigs. The findings of this study have important implications for using iodine-based CAs in imaging analyses of the meniscus and offer potentially valuable insights into the diffusion patterns of iodine in fibrocartilagineous tissues.
Collapse
Affiliation(s)
- Federica Orellana
- Empa – Swiss Federal Laboratories for Materials Science and Technology, 8600, Dübendorf, Switzerland
- Department of Chemistry, University of Fribourg, 1700, Fribourg, Switzerland
| | - Alberto Grassi
- IRCCS - Rizzoli Orthopaedic Institute, 40136, Bologna, Italy
| | - Katja M. Nuss
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Peter Wahl
- Faculty of Medicine, University of Bern, 3008, Bern, Switzerland
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, 8401, Winterthur, Switzerland
| | - Antonia Neels
- Empa – Swiss Federal Laboratories for Materials Science and Technology, 8600, Dübendorf, Switzerland
- Department of Chemistry, University of Fribourg, 1700, Fribourg, Switzerland
| | | | - Annapaola Parrilli
- Empa – Swiss Federal Laboratories for Materials Science and Technology, 8600, Dübendorf, Switzerland
| |
Collapse
|
16
|
Zhong M, He H, Ni P, Huang C, Zhang T, Chen W, Liu L, Wang C, Jiang X, Pu L, Yuan T, Liang J, Fan Y, Zhang X. Semi-quantitative scoring criteria based on multiple staining methods combined with machine learning to evaluate residual nuclei in decellularized matrix. Regen Biomater 2024; 12:rbae147. [PMID: 39886363 PMCID: PMC11780845 DOI: 10.1093/rb/rbae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 02/01/2025] Open
Abstract
The detection of residual nuclei in decellularized extracellular matrix (dECM) biomaterials is critical for ensuring their quality and biocompatibility. However, current evaluation methods have limitations in addressing impurity interference and providing intelligent analysis. In this study, we utilized four staining techniques-hematoxylin-eosin staining, acetocarmine staining, the Feulgen reaction and 4',6-diamidino-2-phenylindole staining-to detect residual nuclei in dECM biomaterials. Each staining method was quantitatively evaluated across multiple parameters, including area, perimeter and grayscale values, to establish a semi-quantitative scoring system for residual nuclei. These quantitative data were further employed as learning indicators in machine learning models designed to automatically identify residual nuclei. The experimental results demonstrated that no single staining method alone could accurately differentiate between nuclei and impurities. In this study, a semi-quantitative scoring table was developed. With this table, the accuracy of determining whether a single suspicious point is a cell nucleus has reached over 98%. By combining four staining methods, false positives caused by impurity contamination were eliminated. The automatic recognition model trained based on nuclear parameter features reached the optimal index of the model after several iterations of training in 172 epochs. The trained artificial intelligence model achieved a recognition accuracy of over 90% for detecting residual nuclei. The use of multidimensional parameters, integrated with machine learning, significantly improved the accuracy of identifying nuclear residues in dECM slices. This approach provides a more reliable and objective method for evaluating dECM biomaterials, while also increasing detection efficiency.
Collapse
Affiliation(s)
- Meng Zhong
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Hongwei He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Panxianzhi Ni
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Can Huang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Tianxiao Zhang
- Neo Modulus (Suzhou) Medical Technology Co., Ltd, Suzhou 215163, China
| | - Weiming Chen
- Neo Modulus (Suzhou) Medical Technology Co., Ltd, Suzhou 215163, China
| | - Liming Liu
- Kemoshen AI Lab, Shanghai Kemosheng Medical Technology Co., Ltd, Shanghai 201700, China
| | - Changfeng Wang
- Kemoshen AI Lab, Shanghai Kemosheng Medical Technology Co., Ltd, Shanghai 201700, China
| | - Xin Jiang
- Sichuan Testing Center for Biomaterials and Medical Devices Co., Ltd, Chengdu 610064, China
| | - Linyun Pu
- Sichuan Testing Center for Biomaterials and Medical Devices Co., Ltd, Chengdu 610064, China
| | - Tun Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Sichuan Testing Center for Biomaterials and Medical Devices Co., Ltd, Chengdu 610064, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Sichuan Testing Center for Biomaterials and Medical Devices Co., Ltd, Chengdu 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
17
|
Murchan P, Ó Broin P, Baird AM, Sheils O, P Finn S. Deep feature batch correction using ComBat for machine learning applications in computational pathology. J Pathol Inform 2024; 15:100396. [PMID: 39398947 PMCID: PMC11470259 DOI: 10.1016/j.jpi.2024.100396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Background Developing artificial intelligence (AI) models for digital pathology requires large datasets from multiple sources. However, without careful implementation, AI models risk learning confounding site-specific features in datasets instead of clinically relevant information, leading to overestimated performance, poor generalizability to real-world data, and potential misdiagnosis. Methods Whole-slide images (WSIs) from The Cancer Genome Atlas (TCGA) colon (COAD), and stomach adenocarcinoma datasets were selected for inclusion in this study. Patch embeddings were obtained using three feature extraction models, followed by ComBat harmonization. Attention-based multiple instance learning models were trained to predict tissue-source site (TSS), as well as clinical and genetic attributes, using raw, Macenko normalized, and Combat-harmonized patch embeddings. Results TSS prediction achieved high accuracy (AUROC > 0.95) with all three feature extraction models. ComBat harmonization significantly reduced the AUROC for TSS prediction, with mean AUROCs dropping to approximately 0.5 for most models, indicating successful mitigation of batch effects (e.g., CCL-ResNet50 in TCGA-COAD: Pre-ComBat AUROC = 0.960, Post-ComBat AUROC = 0.506, p < 0.001). Clinical attributes associated with TSS, such as race and treatment response, showed decreased predictability post-harmonization. Notably, the prediction of genetic features like MSI status remained robust after harmonization (e.g., MSI in TCGA-COAD: Pre-ComBat AUROC = 0.667, Post-ComBat AUROC = 0.669, p=0.952), indicating the preservation of true histological signals. Conclusion ComBat harmonization of deep learning-derived histology features effectively reduces the risk of AI models learning confounding features in WSIs, ensuring more reliable performance estimates. This approach is promising for the integration of large-scale digital pathology datasets.
Collapse
Affiliation(s)
- Pierre Murchan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D08 W9RT, Ireland
- The SFI Centre for Research Training in Genomics Data Science, Dublin, Ireland
| | - Pilib Ó Broin
- The SFI Centre for Research Training in Genomics Data Science, Dublin, Ireland
- School of Mathematical & Statistical Sciences, University of Galway, Galway H91 TK33, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D02 A440, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D02 A440, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D08 W9RT, Ireland
- Department of Histopathology, St. James's Hospital, James's Street, Dublin D08 X4RX, Ireland
| |
Collapse
|
18
|
Samset Hoem K, Lillerovde Ørstenvik H, Varhaugvik AE, Tveten AK. In vitro cultivation of adherent cells on microscope slides for downstream applications. MethodsX 2024; 13:102847. [PMID: 39101125 PMCID: PMC11294713 DOI: 10.1016/j.mex.2024.102847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
In vitro studies with cultured cells are often conducted as an important part of basic research. Adherent cells are typically cultivated in flasks or trays, for which cell staining and subsequent visualization become impractical. We here present a simple step-by-step method for growing adherent cells directly on glass microscope slides, using low-cost equipment readily available in most laboratories. Most parameters such as type of microscope slide (e.g. surface coating), cell seeding concentrations and incubation times can be adjusted according to cell line characteristics and experimental aims, reflecting the methods' flexibility. Through our experiments, microscope slides proved to provide an acceptable surface for cell adhesion and growth of the tested cell lines, as well as being robust and functional with respect to downstream procedures. The method can potentially be combined with different techniques for visualization of experimental effects, such as histological staining methods, fluorescent staining, and immunochemistry. In our method development we have successfully cultivated three different cell lines directly on microscope slides - Atlantic salmon kidney cells (ASK), rainbow trout gill cells (RTgill-W1), and human cancerous lung cells (A549) - and subjected them to various experimental treatments. Finally, as proof-of-concept we provide examples of successful histological staining of the fixed cells. Experimental design in short:•Cultivate cells and calculate cell concentration•Seed a small volume of growth medium with an appropriate number of cells on microscope slide in an area confined by hydrophobic marker•Let cells adhere over night before adding more growth medium or directly conducting experiments and fixing cells for downstream applications.
Collapse
Affiliation(s)
| | | | - Anne Elin Varhaugvik
- Department of Biological Sciences Aalesund, Faculty of Natural Sciences, Norwegian University of Science and Technology, Larsgardsvegen 2, Aalesund 6009, Norway
| | - Ann-Kristin Tveten
- Department of Biological Sciences Aalesund, Faculty of Natural Sciences, Norwegian University of Science and Technology, Larsgardsvegen 2, Aalesund 6009, Norway
| |
Collapse
|
19
|
Lampe M, Dietrich B, Wnetrzak J, Waring T, Lycett G, Merino MM, Adams DJ, Marcello M. A biocompatible supramolecular hydrogel mesh for sample stabilization in light microscopy and nanoscopy. Sci Rep 2024; 14:29232. [PMID: 39587170 PMCID: PMC11589135 DOI: 10.1038/s41598-024-76661-x] [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: 02/02/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Most embedding media for live and fixed samples were not designed for microscopy and have issues including long polymerization times, peak of toxicity toward the sample during the sol-gel transition, and irreversibility of this transition. Gels derived from biological sources are widely used in microscopy, but their precise composition is ill-defined and can vary between batches. Non-physiological temperatures and/or specific enzymatic solutions are often needed to revert the gel back to the sol state to allow sample recovery. Recovering the sample undamaged is important for multiple purposes, from the ability to release a living organism back into its environment and re-observe it at a later stage, to interrogating the sample once freed from the gel after imaging. We describe a supramolecular hydrogel that enables the observation of small living organisms using light microscopy, with simple sample recovery through vigorous pipetting with water. The organisms can be recovered alive and capable of further development into adulthood, which represents a significant advancement, as most other matrices require release conditions such as heating, the addition of chemicals, or mechanical disruption, which can damage or kill the embedded organisms. Furthermore, the gel is compatible with super-resolution multi-colour STED nanoscopy.
Collapse
Affiliation(s)
- Marko Lampe
- Advanced Light Microscopy Facility, EMBL, Meyerhofstr. 1, 69117, Heidelberg, Germany
| | - Bart Dietrich
- School of Chemistry, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Joanna Wnetrzak
- Centre for Cell Imaging, Institute of Systems, Molecular and Integrative Biology, Liverpool, L69 7ZB, UK
| | - Tom Waring
- Centre for Cell Imaging, Institute of Systems, Molecular and Integrative Biology, Liverpool, L69 7ZB, UK
| | - Gareth Lycett
- Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Marisa M Merino
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, Liverpool, L69 7ZB, UK
| | - Dave J Adams
- School of Chemistry, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Marco Marcello
- Centre for Cell Imaging, Institute of Systems, Molecular and Integrative Biology, Liverpool, L69 7ZB, UK.
| |
Collapse
|
20
|
Laguna-Castro S, Salminen A, Arponen O, Hannula M, Rinta-Kiikka I, Hyttinen J, Tolonen T. Micro-computed Tomography in the Evaluation of Eosin-stained Axillary Lymph Node Biopsies of Females Diagnosed with Breast Cancer. Sci Rep 2024; 14:28237. [PMID: 39548163 PMCID: PMC11568233 DOI: 10.1038/s41598-024-79060-4] [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: 01/09/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Histopathological investigation of metastasis in core needle axillary lymph node (ALN) biopsies is crucial for the prognosis and treatment planning of breast cancer patients. Biopsies are typically sliced and evaluated as two-dimensional (2D) images. Biopsy sampling errors and the limited view provided by 2D histology are leading factors contributing to false-negative results in the preoperative detection of metastatic lymph nodes and underestimation of metastatic foci.In this proof-of-concept study, we aim to explore the technical feasibility and the potential capacities of tridimensional (3D) X-ray micro-computed tomography imaging to expedite error detection, enhancement of histopathological accuracy, and precise measurement of metastatic lesion on ALN core needle biopsies of two breast cancer patients. Our self-developed micro-CT protocol uses eosin for the first time, a common histological dye, to enhance 3D architecture of ALNs. Performed analysis on the images of the ALN biopsies involves cancer tissue segmentation, swift biopsy evaluation, and measurement of the metastatic longest diameter and deposit volume.The eosin micro-CT protocol shows potential for an improved tumor deposit estimates, offering additional clinical value compared to standard 2D histology, however, further studies for validating this method are needed.
Collapse
Affiliation(s)
- Santiago Laguna-Castro
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland.
- BioMediTech Unit, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland.
| | - Annukka Salminen
- Department of Radiology, Tampere University Hospital, Tampere, 33520, Finland
| | - Otso Arponen
- Department of Radiology, Tampere University Hospital, Tampere, 33520, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
| | - Markus Hannula
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
- BioMediTech Unit, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
| | - Irina Rinta-Kiikka
- Department of Radiology, Tampere University Hospital, Tampere, 33520, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
| | - Jari Hyttinen
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
- BioMediTech Unit, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
| | - Teemu Tolonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
- Department of Pathology, Fimlab Laboratories, Tampere, 33520, Finland
| |
Collapse
|
21
|
Rodrigues-Jesus J, Canadas-Sousa A, Santos M, Oliveira P, Figueira AC, Marrinhas C, Petrucci GN, Gregório H, Tinoco F, Goulart A, Felga H, Vilhena H, Dias-Pereira P. Level of Necrosis in Feline Mammary Tumors: How to Quantify, Why and for What Purpose? Animals (Basel) 2024; 14:3280. [PMID: 39595332 PMCID: PMC11591325 DOI: 10.3390/ani14223280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Necrosis is a common finding in human and animal neoplasms. The percentage of tumor necrosis is included in tumor grading schemes in veterinary oncology; however, evaluation methods are often overlooked. Different studies have assessed the prognostic value of tumor necrosis in feline mammary tumors with contradictory results, which could be related to methodologic variability. In this study, a comprehensive evaluation of tumor necrosis in feline mammary tumors (FMTs) was conducted, by applying a semi-quantitative and a quantitative methodology for assessing necrosis. The interobserver agreement, the relationship with clinicopathological characteristics and the prognostic value of necrosis were analyzed in 154 FMT cases. Although subjectivity in the assessment of necrosis existed, an almost perfect agreement (weighted quadratic k = 0.851) between two observers was obtained. Furthermore, there was a significant positive correlation between the semi-quantitative and quantitative methods. Necrosis was more common and more extensive in malignant tumors than in their benign counterparts. Despite the non-significant results in the survival analysis, extensive necrosis was significantly associated with aggressive clinicopathological features, such as higher histological grade, high mitotic count and lymphovascular invasion. Our results support the potential relevance of necrosis in FMT.
Collapse
Affiliation(s)
- Joana Rodrigues-Jesus
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, 4050-313 Porto, Portugal; (J.R.-J.); (A.C.-S.)
| | - Ana Canadas-Sousa
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, 4050-313 Porto, Portugal; (J.R.-J.); (A.C.-S.)
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, 3020-210 Coimbra, Portugal; (A.C.F.); (C.M.); (H.V.)
| | - Marta Santos
- Department of Microscopy, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, 4050-313 Porto, Portugal;
| | - Pedro Oliveira
- Department of Populations Studies, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, 4050-313 Porto, Portugal;
| | - Ana Catarina Figueira
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, 3020-210 Coimbra, Portugal; (A.C.F.); (C.M.); (H.V.)
- OneVet Veterinary University Hospital of Coimbra (HVUC), 3020-210 Coimbra, Portugal
| | - Carla Marrinhas
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, 3020-210 Coimbra, Portugal; (A.C.F.); (C.M.); (H.V.)
- OneVet Veterinary Hospital of Baixo Vouga (HVBV), 3750-742 Águeda, Portugal
| | - Gonçalo N. Petrucci
- OneVet Veterinary Hospital of Porto (HVP), 4250-475 Porto, Portugal;
- Department of Animal and Veterinary Sciences, University Institute for Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal;
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Hugo Gregório
- Department of Animal and Veterinary Sciences, University Institute for Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal;
- AniCura Veterinary Hospital Centre (CHV), 4100-320 Porto, Portugal
| | - Flora Tinoco
- Dra. Flora Tinoco Veterinary Clinic, 4475-498 Maia, Portugal;
| | | | - Helena Felga
- Clínica dos Gatos Veterinary Clinic, 4100-207 Porto, Portugal;
| | - Hugo Vilhena
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, 3020-210 Coimbra, Portugal; (A.C.F.); (C.M.); (H.V.)
- OneVet Veterinary University Hospital of Coimbra (HVUC), 3020-210 Coimbra, Portugal
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Associate Laboratory of Animal and Veterinary Sciences AL4AnimaLS, 1300-477 Lisbon, Portugal
| | - Patrícia Dias-Pereira
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, 4050-313 Porto, Portugal; (J.R.-J.); (A.C.-S.)
| |
Collapse
|
22
|
Maris L, Göker M, Debacker JM, De Man K, Van den Broeck B, Van Dorpe J, Van de Vijver K, Keereman V, Vanhove C. Method for co-registration of high-resolution specimen PET-CT with histopathology to improve insight into radiotracer distributions. EJNMMI Phys 2024; 11:85. [PMID: 39400788 PMCID: PMC11473743 DOI: 10.1186/s40658-024-00681-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND As the spatial resolution of positron emission tomography (PET) scanners improves, understanding of radiotracer distributions in tissues at high resolutions is important. Hence, we propose a method for co-registration of high-resolution ex vivo specimen PET images, combined with computed tomography (CT) images, and the corresponding specimen histopathology. METHODS We applied our co-registration method to breast cancer (BCa) specimens of patients who were preoperatively injected with 0.8 MBq/kg [18 F]fluorodeoxyglucose ([18F]FDG). The method has two components. First, we used an image acquisition scheme that minimises and tracks tissue deformation: (1) We acquired sub-millimetre (micro)-PET-CT images of ±2 mm-thick lamellas of the fresh specimens, enclosed in tissue cassettes. (2) We acquired micro-CT images of the same lamellas after formalin fixation to visualise tissue deformation. (3) We obtained 1 hematoxylin and eosin (H&E) stained histopathology section per lamella of which we captured a digital whole slide image (WSI). Second, we developed an automatic co-registration algorithm to improve the alignment between the micro-PET-CT images and WSIs, guided by the micro-CT of the fixated lamellas. To estimate the spatial co-registration error, we calculated the distance between corresponding microcalcifications in the micro-CTs and WSIs. The co-registered images allowed to study standardised uptake values (SUVs) of different breast tissues, as identified on the WSIs by a pathologist. RESULTS We imaged 22 BCa specimens, 13 cases of invasive carcinoma of no special type (NST), 6 of invasive lobular carcinoma (ILC), and 3 of ductal carcinoma in situ (DCIS). While the cassette framework minimised tissue deformation, the best alignment between the micro-PET-CT images and WSIs was achieved after deformable co-registration. We found an overall average co-registration error of 0.74 ± 0.17 mm between the micro-PET images and WSIs. (Pre)malignant tissue (including NST, ILC, and DCIS) generally showed higher SUVs than healthy tissue (including healthy glandular, connective, and adipose tissue). As expected, inflamed tissue and skin also showed high uptake. CONCLUSIONS We developed a method to co-register micro-PET-CT images of surgical specimens and WSIs with an accuracy comparable to the spatial resolution of the micro-PET images. While currently, we only applied this method to BCa specimens, we believe this method is applicable to a wide range of specimens and radiotracers, providing insight into distributions of (new) radiotracers in human malignancies at a sub-millimetre resolution.
Collapse
Affiliation(s)
- Luna Maris
- Department of Electronics and Information Systems, Medical Image and Signal Processing, Ghent University, Ghent, Belgium.
- Clinical Department, XEOS Medical, Ghent, Belgium.
| | - Menekse Göker
- Department of Gynaecology, Ghent University Hospital, Ghent, Belgium
| | - Jens M Debacker
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Nuclear Medicine, UZ Brussel, Brussels, Belgium
- Department of Head and Skin, Head and Neck Surgery Research Group, Ghent University, Ghent, Belgium
| | - Kathia De Man
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bliede Van den Broeck
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Koen Van de Vijver
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Vincent Keereman
- Department of Electronics and Information Systems, Medical Image and Signal Processing, Ghent University, Ghent, Belgium
- Clinical Department, XEOS Medical, Ghent, Belgium
| | - Christian Vanhove
- Department of Electronics and Information Systems, Medical Image and Signal Processing, Ghent University, Ghent, Belgium
- INFINITY Lab, Ghent University, Ghent, Belgium
| |
Collapse
|
23
|
Kanwal N, Khoraminia F, Kiraz U, Mosquera-Zamudio A, Monteagudo C, Janssen EAM, Zuiverloon TCM, Rong C, Engan K. Equipping computational pathology systems with artifact processing pipelines: a showcase for computation and performance trade-offs. BMC Med Inform Decis Mak 2024; 24:288. [PMID: 39375719 PMCID: PMC11457387 DOI: 10.1186/s12911-024-02676-z] [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: 07/24/2024] [Accepted: 09/09/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Histopathology is a gold standard for cancer diagnosis. It involves extracting tissue specimens from suspicious areas to prepare a glass slide for a microscopic examination. However, histological tissue processing procedures result in the introduction of artifacts, which are ultimately transferred to the digitized version of glass slides, known as whole slide images (WSIs). Artifacts are diagnostically irrelevant areas and may result in wrong predictions from deep learning (DL) algorithms. Therefore, detecting and excluding artifacts in the computational pathology (CPATH) system is essential for reliable automated diagnosis. METHODS In this paper, we propose a mixture of experts (MoE) scheme for detecting five notable artifacts, including damaged tissue, blur, folded tissue, air bubbles, and histologically irrelevant blood from WSIs. First, we train independent binary DL models as experts to capture particular artifact morphology. Then, we ensemble their predictions using a fusion mechanism. We apply probabilistic thresholding over the final probability distribution to improve the sensitivity of the MoE. We developed four DL pipelines to evaluate computational and performance trade-offs. These include two MoEs and two multiclass models of state-of-the-art deep convolutional neural networks (DCNNs) and vision transformers (ViTs). These DL pipelines are quantitatively and qualitatively evaluated on external and out-of-distribution (OoD) data to assess generalizability and robustness for artifact detection application. RESULTS We extensively evaluated the proposed MoE and multiclass models. DCNNs-based MoE and ViTs-based MoE schemes outperformed simpler multiclass models and were tested on datasets from different hospitals and cancer types, where MoE using (MobileNet) DCNNs yielded the best results. The proposed MoE yields 86.15 % F1 and 97.93% sensitivity scores on unseen data, retaining less computational cost for inference than MoE using ViTs. This best performance of MoEs comes with relatively higher computational trade-offs than multiclass models. Furthermore, we apply post-processing to create an artifact segmentation mask, a potential artifact-free RoI map, a quality report, and an artifact-refined WSI for further computational analysis. During the qualitative evaluation, field experts assessed the predictive performance of MoEs over OoD WSIs. They rated artifact detection and artifact-free area preservation, where the highest agreement translated to a Cohen Kappa of 0.82, indicating substantial agreement for the overall diagnostic usability of the DCNN-based MoE scheme. CONCLUSIONS The proposed artifact detection pipeline will not only ensure reliable CPATH predictions but may also provide quality control. In this work, the best-performing pipeline for artifact detection is MoE with DCNNs. Our detailed experiments show that there is always a trade-off between performance and computational complexity, and no straightforward DL solution equally suits all types of data and applications. The code and HistoArtifacts dataset can be found online at Github and Zenodo , respectively.
Collapse
Affiliation(s)
- Neel Kanwal
- Department of Electrical Engineering and Computer Science, University of Stavanger, 4021, Stavanger, Norway.
| | - Farbod Khoraminia
- Department of Urology, University Medical Center Rotterdam, Erasmus MC Cancer Institute, 1035 GD, Rotterdam, The Netherlands
| | - Umay Kiraz
- Department of Pathology, Stavanger University Hospital, 4011, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021, Stavanger, Norway
| | - Andrés Mosquera-Zamudio
- Department of Pathology, INCLIVA Biomedical Research Institute, and University of Valencia, 46010, Valencia, Spain
| | - Carlos Monteagudo
- Department of Pathology, INCLIVA Biomedical Research Institute, and University of Valencia, 46010, Valencia, Spain
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, 4011, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021, Stavanger, Norway
| | - Tahlita C M Zuiverloon
- Department of Urology, University Medical Center Rotterdam, Erasmus MC Cancer Institute, 1035 GD, Rotterdam, The Netherlands
| | - Chunming Rong
- Department of Electrical Engineering and Computer Science, University of Stavanger, 4021, Stavanger, Norway
| | - Kjersti Engan
- Department of Electrical Engineering and Computer Science, University of Stavanger, 4021, Stavanger, Norway.
| |
Collapse
|
24
|
Cheng Q, Zhao W, Song X, Jin T. Machine-learning and scRNA-Seq-based diagnostic and prognostic models illustrating survival and therapy response of lung adenocarcinoma. Genes Immun 2024; 25:356-366. [PMID: 39075270 DOI: 10.1038/s41435-024-00289-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/31/2024]
Abstract
Lung cancer is a major cause accounting for cancer-related mortalities, with lung adenocarcinoma (LUAD) being the most prevalent subtype. Given the high clinical and cellular heterogeneities of LUAD, accurate diagnosis and prognosis are crucial to avoid overdiagnosis and overtreatment. Taking full advantage of scRNA-Seq data to resolve the tumor heterogeneities, we explored the overall landscape of LUAD microenvironment. Utilizing the stage-specific tumor cell markers, we have developed highly accurate diagnostic and prognostic models with elevated sensitivity and specificity. The diagnostic model, developed through random forest algorithms with a thirteen-gene signature, achieved an accuracy of 96.4% and an AUC of 0.993. These metrics were further demonstrated by benchmarking with available models and scoring systems in independent cohorts. Concurrently, the prognostic model, formulated via Cox regression with a six-gene signature, effectively predicted overall survival, with elevated risk scores associated with increased fractions of cancer-associated fibroblasts, and higher likelihood of immune escape and T-cell exclusion. Subsequently, two nomograms were developed to predict survival and drug responses, facilitating their integration into clinical practice. Overall, this study underscores the potential of our models for efficient, rapid, and cost-effective diagnosis and prognosis of LUAD, adaptable to multiple expression profiling platforms and quantification methods.
Collapse
Affiliation(s)
- Qingyu Cheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Weidong Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Cellular Dynamics, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China.
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China.
- Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
25
|
Zhou J, Li X, Demeke D, Dinh TA, Yang Y, Janowczyk AR, Zee J, Holzman L, Mariani L, Chakrabarty K, Barisoni L, Hodgin JB, Lafata KJ. Characterization of arteriosclerosis based on computer-aided measurements of intra-arterial thickness. J Med Imaging (Bellingham) 2024; 11:057501. [PMID: 39398866 PMCID: PMC11466048 DOI: 10.1117/1.jmi.11.5.057501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 10/15/2024] Open
Abstract
Purpose Our purpose is to develop a computer vision approach to quantify intra-arterial thickness on digital pathology images of kidney biopsies as a computational biomarker of arteriosclerosis. Approach The severity of the arteriosclerosis was scored (0 to 3) in 753 arteries from 33 trichrome-stained whole slide images (WSIs) of kidney biopsies, and the outer contours of the media, intima, and lumen were manually delineated by a renal pathologist. We then developed a multi-class deep learning (DL) framework for segmenting the different intra-arterial compartments (training dataset: 648 arteries from 24 WSIs; testing dataset: 105 arteries from 9 WSIs). Subsequently, we employed radial sampling and made measurements of media and intima thickness as a function of spatially encoded polar coordinates throughout the artery. Pathomic features were extracted from the measurements to collectively describe the arterial wall characteristics. The technique was first validated through numerical analysis of simulated arteries, with systematic deformations applied to study their effect on arterial thickness measurements. We then compared these computationally derived measurements with the pathologists' grading of arteriosclerosis. Results Numerical validation shows that our measurement technique adeptly captured the decreasing smoothness in the intima and media thickness as the deformation increases in the simulated arteries. Intra-arterial DL segmentations of media, intima, and lumen achieved Dice scores of 0.84, 0.78, and 0.86, respectively. Several significant associations were identified between arteriosclerosis grade and pathomic features using our technique (e.g., intima-media ratio average [ τ = 0.52 , p < 0.0001 ]) through Kendall's tau analysis. Conclusions We developed a computer vision approach to computationally characterize intra-arterial morphology on digital pathology images and demonstrate its feasibility as a potential computational biomarker of arteriosclerosis.
Collapse
Affiliation(s)
- Jin Zhou
- Duke University, Department of Electrical and Computer Engineering, Durham, North Carolina, United States
| | - Xiang Li
- Duke University, Department of Electrical and Computer Engineering, Durham, North Carolina, United States
| | - Dawit Demeke
- University of Michigan, Department of Pathology, Ann Arbor, Michigan, United States
| | - Timothy A. Dinh
- University of Michigan, Department of Pathology, Ann Arbor, Michigan, United States
| | - Yingbao Yang
- University of Michigan, Department of Pathology, Ann Arbor, Michigan, United States
| | - Andrew R. Janowczyk
- Geneva University Hospitals, Department of Oncology, Division of Precision Oncology, Geneva, Switzerland
- Geneva University Hospitals, Department of Diagnostics, Division of Clinical Pathology, Geneva, Switzerland
- Emory University, Department of Biomedical Engineering, Atlanta, Georgia, United States
- Georgia Institute of Technology, Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Jarcy Zee
- University of Pennsylvania, Department of Biostatistics, Epidemiology, and Informatics, Philadelphia, Pennsylvania, United States
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Lawrence Holzman
- University of Pennsylvania, Department of Medicine, Renal-Electrolyte and Hypertension Division, Philadelphia, Pennsylvania, United States
| | - Laura Mariani
- University of Michigan, Department of Internal Medicine, Division of Nephrology, Ann Arbor, Michigan, United States
| | - Krishnendu Chakrabarty
- Arizona State University, School of Electrical, Computer and Energy Engineering, Tempe, Arizona, United States
| | - Laura Barisoni
- Duke University, Division of Artificial Intelligence and Computational Pathology, Department of Pathology, Durham, North Carolina, United States
- Duke University, Division of Nephrology Department of Medicine, Durham, North Carolina, United States
| | - Jeffrey B. Hodgin
- University of Michigan, Department of Pathology, Ann Arbor, Michigan, United States
| | - Kyle J. Lafata
- Duke University, Department of Electrical and Computer Engineering, Durham, North Carolina, United States
- Duke University, Division of Artificial Intelligence and Computational Pathology, Department of Pathology, Durham, North Carolina, United States
- Duke University, Department of Radiology, Durham, North Carolina, United States
- Duke University, Department of Radiation Oncology, Durham, North Carolina, United States
| |
Collapse
|
26
|
Suurmond CE, Leeuwenburgh SCG, van den Beucken JJJP. Modelling bone metastasis in spheroids to study cancer progression and screen cisplatin efficacy. Cell Prolif 2024; 57:e13693. [PMID: 38899562 PMCID: PMC11503253 DOI: 10.1111/cpr.13693] [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: 04/09/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Most bone metastases are caused by primary breast or prostate cancer cells settling in the bone microenvironment, affecting normal bone physiology and function and reducing 5-year survival rates to 10% and 6%, respectively. To expedite clinical availability of novel and effective bone metastases treatments, reliable and predictive in vitro models are urgently required to screen for novel therapies as current in vitro 2D planar mono-culture models do not accurately predict the clinical efficacy. We herein engineered a novel human in vitro 3D co-culture model based on spheroids to study dynamic cellular quantities of (breast or prostate) cancer cells and human bone marrow stromal cells and screen chemotherapeutic efficacy and specificity of the common anticancer drug cisplatin. Bone metastatic spheroids (BMSs) were formed rapidly within 24 h, while the morphology of breast versus prostate cancer BMS differed in terms of size and circularity upon prolonged culture periods. Prestaining cell types prior to BMS formation enabled confocal imaging and quantitative image analysis of in-spheroid cellular dynamics for up to 7 days of BMS culture. We found that cancer cells in BMS proliferated faster and were less susceptible to cisplatin treatment compared to 2D control cultures. Based on these findings and the versatility of our methodology, BMS represent a feasible 3D in vitro model for screening of new bone cancer metastases therapies.
Collapse
|
27
|
Stricker A, Fretwurst T, Abdullayeva A, Bosshardt D, Aghaloo T, Duttenhöfer F, Cordaro L, Nelson K, Gross C. Vitality of autologous retromolar bone grafts for alveolar ridge augmentation after a 3-months healing period: A prospective histomorphometrical analysis. Clin Oral Implants Res 2024; 35:1151-1162. [PMID: 38847078 DOI: 10.1111/clr.14306] [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: 12/28/2023] [Revised: 04/30/2024] [Accepted: 05/18/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVES The incorporation of retromolar bone grafts used for alveolar ridge augmentation is not well understood. This prospective observational study aims to supply histomorphometrical data from bone graft biopsies taken at the time of retrieval and after a 3-month healing period using patient-matched biopsies. MATERIALS AND METHODS In 17 patients, trephine biopsies of the graft were acquired at the time of graft retrieval and after a 3-month healing period. The biopsies were compared histomorphometrically regarding the number of osteocytes, appearance of osteocyte lacunae, quantity, surface area, and activity of the Haversian canals. RESULTS All grafts appeared clinically stable after screw removal and 17 implants were placed. Histomorphometric analysis revealed no significant difference in the number of osteocytes (p = .413), osteocyte lacunae (p = .611), the ratio of filled/empty osteocyte lacunae (p = .467) and active Haversian canals (p = .495) between the biopsies retrieved after a 3-months healing period with those at the time of grafting. The only significant difference was noted in the mean surface area of the Haversian canals (p = .002). Specifically, the grafts post 3-month healing showed a significantly larger mean area (0.069 mm2) compared to the time of grafting (0.029 mm2). CONCLUSION This study demonstrates, compared to other data, a high rate of vital structures in retromolar bone block grafts after 3 months of healing, exhibiting the same histological features in comparison to the biopsies from the native alveolar ridge. Standard histomorphometrical parameters, e.g., the amount of filled or empty osteocyte lacunae for the description of the vitality of the graft need to be reappraised.
Collapse
Affiliation(s)
- Andres Stricker
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Tobias Fretwurst
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Arzu Abdullayeva
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Dieter Bosshardt
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
- Robert K. Schenk Laboratory of Oral Histology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Tara Aghaloo
- Section of Oral and Maxillofacial Surgery, UCLA School of Dentistry, Los Angeles, California, USA
| | - Fabian Duttenhöfer
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Luca Cordaro
- Department of Periodontics and Prosthodontics, Policlinico Umberto I, Eastman Dental Hospital, Rome, Italy
| | - Katja Nelson
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Christian Gross
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
28
|
Jurgas A, Wodzinski M, D'Amato M, van der Laak J, Atzori M, Müller H. Improving quality control of whole slide images by explicit artifact augmentation. Sci Rep 2024; 14:17847. [PMID: 39090284 PMCID: PMC11294620 DOI: 10.1038/s41598-024-68667-2] [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: 02/23/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
The problem of artifacts in whole slide image acquisition, prevalent in both clinical workflows and research-oriented settings, necessitates human intervention and re-scanning. Overcoming this challenge requires developing quality control algorithms, that are hindered by the limited availability of relevant annotated data in histopathology. The manual annotation of ground-truth for artifact detection methods is expensive and time-consuming. This work addresses the issue by proposing a method dedicated to augmenting whole slide images with artifacts. The tool seamlessly generates and blends artifacts from an external library to a given histopathology dataset. The augmented datasets are then utilized to train artifact classification methods. The evaluation shows their usefulness in classification of the artifacts, where they show an improvement from 0.10 to 0.01 AUROC depending on the artifact type. The framework, model, weights, and ground-truth annotations are freely released to facilitate open science and reproducible research.
Collapse
Affiliation(s)
- Artur Jurgas
- AGH University of Krakow, Faculty of Electrical Engineering, Automatics, Computer Science and Biomedical Engineering, 30059, Krakow, Poland.
- University of Applied Sciences Western Switzerland (HES-SO), Institute of Informatics, 3960, Sierre, Switzerland.
| | - Marek Wodzinski
- AGH University of Krakow, Faculty of Electrical Engineering, Automatics, Computer Science and Biomedical Engineering, 30059, Krakow, Poland
- University of Applied Sciences Western Switzerland (HES-SO), Institute of Informatics, 3960, Sierre, Switzerland
| | - Marina D'Amato
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Manfredo Atzori
- University of Applied Sciences Western Switzerland (HES-SO), Institute of Informatics, 3960, Sierre, Switzerland
- Department of Neuroscience, University of Padova, Padua, Italy
| | - Henning Müller
- University of Applied Sciences Western Switzerland (HES-SO), Institute of Informatics, 3960, Sierre, Switzerland
- Medical Faculty, University of Geneva, Geneva, Switzerland
| |
Collapse
|
29
|
Lehner A, Hoffmann L, Rampp S, Coras R, Paulsen F, Frischknecht R, Hamer H, Walther K, Brandner S, Hofer W, Pieper T, Reisch L, Bien CG, Blumcke I. Age-dependent increase of perineuronal nets in the human hippocampus and precocious aging in epilepsy. Epilepsia Open 2024; 9:1372-1381. [PMID: 38845524 PMCID: PMC11296138 DOI: 10.1002/epi4.12963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 08/03/2024] Open
Abstract
OBJECTIVE Perineuronal nets (PNN) are specialized extracellular matrix (ECM) components of the central nervous system, frequently accumulating at the surface of inhibitory GABAergic interneurons. While an altered distribution of PNN has been observed in neurological disorders including Alzheimer's disease, schizophrenia and epilepsy, their anatomical distribution also changes during physiological brain maturation and aging. Such an age-dependent shift was experimentally associated also with hippocampal engram formation during brain maturation. Our aim was to histopathologically assess PNN in the hippocampus of adult and pediatric patients with temporal lobe epilepsy (TLE) compared to age-matched post-mortem control subjects and to compare PNN-related changes with memory impairment observed in our patient cohort. METHODS Sixty-six formalin-fixed and paraffin-embedded tissue specimens of the human hippocampus were retrieved from the European Epilepsy Brain Bank. Twenty-nine patients had histopathologically confirmed hippocampal sclerosis (HS), and eleven patients suffered from TLE without HS. PNN were immunohistochemically visualized using an antibody directed against aggrecan and manually counted from hippocampus subfields and the subiculum. RESULTS PNN density increased with age in both human controls and TLE patients. However, their density was significantly higher in all HS patients compared to age-matched controls. Intriguingly, TLE patients presented presurgically with better memory when their hippocampal PNN density was higher (p < 0.05). SIGNIFICANCE Our results were compatible with age-dependent ECM specialization in the human hippocampus and its precocious aging in the epileptic condition. These observations confirm recent experimental animal models and also support the notion that PNN play a role in memory formation in the human brain. PLAIN LANGUAGE SUMMARY "Perineuronal nets" (PNN) are a specialized compartment of the extracellular matrix (ECM), especially surrounding highly active neurons of the mammalian brain. There is evidence that PNN play a role in memory formation, brain maturation, and in some pathologies like Alzheimer's disease, schizophrenia or epilepsy. In this study, we investigated the role of PNN in patients suffering from drug-resistant focal epilepsy compared to controls. We found that with increasing age, more neurons are surrounded by PNN. Similarly, all epilepsy patients but especially patients with better memory performance also had more PNN. This study raises further interest in studying ECM molecules in the human brain under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Annika Lehner
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| | - Lucas Hoffmann
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| | - Stefan Rampp
- Department of NeuroradiologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Department of NeurosurgeryUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
| | - Roland Coras
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, FAU Erlangen‐NürnbergErlangenGermany
| | - Renato Frischknecht
- Department of Biology, Animal PhysiologyFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Hajo Hamer
- Epilepsy Center, Department of NeurologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
| | - Katrin Walther
- Epilepsy Center, Department of NeurologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
| | - Sebastian Brandner
- Department of NeurosurgeryUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Department of NeurosurgeryKlinikum FürthGermany
| | - Wiebke Hofer
- Department of Psychology/Neuropsychology, Center for Pediatric Neurology, Neurorehabilitation, and EpileptologySchön Klinik VogtareuthGermany
| | - Tom Pieper
- Center for Pediatric Neurology, Neurorehabilitation, and EpileptologySchön Klinik VogtareuthGermany
| | - Lea‐Marie Reisch
- Department of Epileptology (Krankenhaus Mara), Medical SchoolBielefeld UniversityBielefeldGermany
| | - Christian G. Bien
- Department of Epileptology (Krankenhaus Mara), Medical SchoolBielefeld UniversityBielefeldGermany
| | - Ingmar Blumcke
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| |
Collapse
|
30
|
Xie Z, Zeinstra N, Kirby MA, Le NM, Murry CE, Zheng Y, Wang RK. Quantifying Microvascular Structure in Healthy and Infarcted Rat Hearts Using Optical Coherence Tomography Angiography. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2878-2887. [PMID: 38568757 PMCID: PMC11341234 DOI: 10.1109/tmi.2024.3381934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Myocardial infarction (MI) is a life-threatening medical emergency resulting in coronary microvascular dysregulation and heart muscle damage. One of the primary characteristics of MI is capillary loss, which plays a significant role in the progression of this cardiovascular condition. In this study, we utilized optical coherence tomography angiography (OCTA) to image coronary microcirculation in fixed rat hearts, aiming to analyze coronary microvascular impairment post-infarction. Various angiographic metrics are presented to quantify vascular features, including the vessel area density, vessel complexity index, vessel tortuosity index, and flow impairment. Pathological differences identified from OCTA analysis are corroborated with histological analysis. The quantitative assessments reveal a significant decrease in microvascular density in the capillary-sized vessels and an enlargement for the arteriole/venule-sized vessels. Further, microvascular tortuosity and complexity exhibit an increase after myocardial infarction. The results underscore the feasibility of using OCTA to offer qualitative microvascular details and quantitative metrics, providing insights into coronary vascular network remodeling during disease progression and response to therapy.
Collapse
|
31
|
Mi H, Sivagnanam S, Ho WJ, Zhang S, Bergman D, Deshpande A, Baras AS, Jaffee EM, Coussens LM, Fertig EJ, Popel AS. Computational methods and biomarker discovery strategies for spatial proteomics: a review in immuno-oncology. Brief Bioinform 2024; 25:bbae421. [PMID: 39179248 PMCID: PMC11343572 DOI: 10.1093/bib/bbae421] [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: 05/29/2024] [Revised: 07/11/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
Advancements in imaging technologies have revolutionized our ability to deeply profile pathological tissue architectures, generating large volumes of imaging data with unparalleled spatial resolution. This type of data collection, namely, spatial proteomics, offers invaluable insights into various human diseases. Simultaneously, computational algorithms have evolved to manage the increasing dimensionality of spatial proteomics inherent in this progress. Numerous imaging-based computational frameworks, such as computational pathology, have been proposed for research and clinical applications. However, the development of these fields demands diverse domain expertise, creating barriers to their integration and further application. This review seeks to bridge this divide by presenting a comprehensive guideline. We consolidate prevailing computational methods and outline a roadmap from image processing to data-driven, statistics-informed biomarker discovery. Additionally, we explore future perspectives as the field moves toward interfacing with other quantitative domains, holding significant promise for precision care in immuno-oncology.
Collapse
Affiliation(s)
- Haoyang Mi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Shamilene Sivagnanam
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
| | - Won Jin Ho
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Shuming Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Daniel Bergman
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Atul Deshpande
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Alexander S Baras
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Pathology, Johns Hopkins University School of Medicine, MD 21205, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Elizabeth M Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Lisa M Coussens
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR 97201, United States
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
| |
Collapse
|
32
|
Tajbakhsh K, Stanowska O, Neels A, Perren A, Zboray R. 3D Virtual Histopathology by Phase-Contrast X-Ray Micro-CT for Follicular Thyroid Neoplasms. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2670-2678. [PMID: 38437150 DOI: 10.1109/tmi.2024.3372602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Histological analysis is the core of follicular thyroid carcinoma (FTC) classification. The histopathological criteria of capsular and vascular invasion define malignancy and aggressiveness of FTC. Analysis of multiple sections is cumbersome and as only a minute tissue fraction is analyzed during histopathology, under-sampling remains a problem. Application of an efficient tool for complete tissue imaging in 3D would speed-up diagnosis and increase accuracy. We show that X-ray propagation-based imaging (XPBI) of paraffin-embedded tissue blocks is a valuable complementary method for follicular thyroid carcinoma diagnosis and assessment. It enables a fast, non-destructive and accurate 3D virtual histology of the FTC resection specimen. We demonstrate that XPBI virtual slices can reliably evaluate capsular invasions. Then we discuss the accessible morphological information from XPBI and their significance for vascular invasion diagnosis. We show 3D morphological information that allow to discern vascular invasions. The results are validated by comparing XPBI images with clinically accepted histology slides revised by and under supervision of two experienced endocrine pathologists.
Collapse
|
33
|
Howie RR, McKinney MM, Tataryn NM, Cole AL, Dupont WD, Yang TS, Gibson-Corley KN. Determination of Postmortem Interval in Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2024; 63:428-436. [PMID: 38471755 PMCID: PMC11270044 DOI: 10.30802/aalas-jaalas-23-000107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/14/2023] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Despite the major use of mice in biomedical research, little information is available with regard to identifying their postmortem changes and using that information to determine the postmortem interval (PMI), defined as the time after death. Both PMI and environmental conditions influence decomposition (autolysis and putrefaction) and other postmortem changes. Severe decomposition compromises lesion interpretation and disease detection and wastes limited pathology resources. The goal of this study was to assess postmortem changes in mice in room temperature cage conditions and under refrigeration at 4 °C to develop gross criteria for the potential value of further gross and histologic evaluation. We used 108 experimentally naïve C57BL/6 mice that were humanely euthanized and then allocated them into 2 experimental groups for evaluation of postmortem change: room temperature (20 to 22 °C) or refrigeration (4 °C). PMI assessments, including gross changes and histologic scoring, were performed at hours 0, 4, 8, and 12 and on days 1 to 14. Factors such as temperature, humidity, ammonia in the cage, and weight change were also documented. Our data indicates that carcasses held at room temperature decomposed faster than refrigerated carcasses. For most tissues, decomposition was evident by 12 h at room temperature as compared with 5 d under refrigeration. At room temperature, gross changes were present by day 2 as compared with day 7 under refrigeration. Mice at room temperature lost 0.78% of their baseline body weight per day as compared with 0.06% for refrigerated mice (95% CI for difference 0.67% to 0.76%, P < 0.0005). This study supports the consideration of temperature and PMI as important factors affecting the suitability of postmortem tissues for gross and histologic evaluation and indicates that storage of carcasses under refrigeration will significantly slow autolysis.
Collapse
Affiliation(s)
- Rachel R Howie
- Department of Pathology, Microbiology, and Immunology, Division of Comparative Medicine, Division of Animal Care, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael M McKinney
- Department of Pathology, Microbiology, and Immunology, Division of Comparative Medicine, Division of Animal Care, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nicholas M Tataryn
- Department of Pathology, Microbiology, and Immunology, Division of Comparative Medicine, Division of Animal Care, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Allysa L Cole
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - William D Dupont
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tzushan S Yang
- Department of Pathology, Microbiology, and Immunology, Division of Comparative Medicine, Division of Animal Care, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Division of Comparative Medicine, Division of Animal Care, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
34
|
Totty M, Hicks SC, Guo B. SpotSweeper: spatially-aware quality control for spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597765. [PMID: 38895212 PMCID: PMC11185656 DOI: 10.1101/2024.06.06.597765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Quality control (QC) is a crucial step to ensure the reliability and accuracy of the data obtained from RNA sequencing experiments, including spatially-resolved transcriptomics (SRT). Existing QC approaches for SRT that have been adopted from single-nucleus RNA sequencing (snRNA-seq) methods are confounded by spatial biology and are inappropriate for SRT data. In addition, no methods currently exist for identifying histological tissue artifacts unique to SRT. Here, we introduce SpotSweeper, spatially-aware QC methods for identifying local outliers and regional artifacts in SRT. SpotSweeper evaluates the quality of individual spots relative to their local neighborhood, thus minimizing bias due to biological heterogeneity, and uses multiscale methods to detect regional artifacts. Using SpotSweeper on publicly available data, we identified a consistent set of Visium barcodes/spots as systematically low quality and demonstrate that SpotSweeper accurately identifies two distinct types of regional artifacts, resulting in improved downstream clustering and marker gene detection for spatial domains.
Collapse
Affiliation(s)
- Michael Totty
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Boyi Guo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
35
|
Micuda A, Li H, Rask-Andersen H, Ladak HM, Agrawal SK. Morphologic Analysis of the Scala Tympani Using Synchrotron: Implications for Cochlear Implantation. Laryngoscope 2024; 134:2889-2897. [PMID: 38189807 DOI: 10.1002/lary.31263] [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: 09/30/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVES To use synchrotron radiation phase-contrast imaging (SR-PCI) to visualize and measure the morphology of the entire cochlear scala tympani (ST) and assess cochlear implant (CI) electrode trajectories. METHODS SR-PCI images were used to obtain geometric measurements of the cochlear scalar diameter and area at 5-degree increments in 35 unimplanted and three implanted fixed human cadaveric cochleae. RESULTS The cross-sectional diameter and area of the cochlea were found to decrease from the base to the apex. This study represents a wide variability in cochlear morphology and suggests that even in the smallest cochlea, the ST can accommodate a 0.4 mm diameter electrode up to 720°. Additionally, all lateral wall array trajectories were within the anatomically accommodating insertion zone. CONCLUSION This is the first study to use SR-PCI to visualize and quantify the entire ST morphology, from the round window to the apical tip, and assess the post-operative trajectory of electrodes. These high-resolution anatomical measurements can be used to inform the angular insertion depth that can be accommodated in CI patients, accounting for anatomical variability. LEVEL OF EVIDENCE N/A. Laryngoscope, 134:2889-2897, 2024.
Collapse
Affiliation(s)
- Ashley Micuda
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Hao Li
- Department of Surgical Sciences, Otorhinolaryngology and Head and Neck Surgery, Uppsala University, Uppsala, Sweden
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Otorhinolaryngology and Head and Neck Surgery, Uppsala University, Uppsala, Sweden
| | - Hanif M Ladak
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- School of Biomedical Engineering, Western University, London, Ontario, Canada
- Department of Otolaryngology-Head and Neck Surgery, Western University, London, Ontario, Canada
- Department of Electrical and Computer Engineering, Western University, London, Ontario, Canada
| | - Sumit K Agrawal
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- School of Biomedical Engineering, Western University, London, Ontario, Canada
- Department of Otolaryngology-Head and Neck Surgery, Western University, London, Ontario, Canada
- Department of Electrical and Computer Engineering, Western University, London, Ontario, Canada
| |
Collapse
|
36
|
Wodzinski M, Marini N, Atzori M, Müller H. RegWSI: Whole slide image registration using combined deep feature- and intensity-based methods: Winner of the ACROBAT 2023 challenge. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 250:108187. [PMID: 38657383 DOI: 10.1016/j.cmpb.2024.108187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/05/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND AND OBJECTIVE The automatic registration of differently stained whole slide images (WSIs) is crucial for improving diagnosis and prognosis by fusing complementary information emerging from different visible structures. It is also useful to quickly transfer annotations between consecutive or restained slides, thus significantly reducing the annotation time and associated costs. Nevertheless, the slide preparation is different for each stain and the tissue undergoes complex and large deformations. Therefore, a robust, efficient, and accurate registration method is highly desired by the scientific community and hospitals specializing in digital pathology. METHODS We propose a two-step hybrid method consisting of (i) deep learning- and feature-based initial alignment algorithm, and (ii) intensity-based nonrigid registration using the instance optimization. The proposed method does not require any fine-tuning to a particular dataset and can be used directly for any desired tissue type and stain. The registration time is low, allowing one to perform efficient registration even for large datasets. The method was proposed for the ACROBAT 2023 challenge organized during the MICCAI 2023 conference and scored 1st place. The method is released as open-source software. RESULTS The proposed method is evaluated using three open datasets: (i) Automatic Nonrigid Histological Image Registration Dataset (ANHIR), (ii) Automatic Registration of Breast Cancer Tissue Dataset (ACROBAT), and (iii) Hybrid Restained and Consecutive Histological Serial Sections Dataset (HyReCo). The target registration error (TRE) is used as the evaluation metric. We compare the proposed algorithm to other state-of-the-art solutions, showing considerable improvement. Additionally, we perform several ablation studies concerning the resolution used for registration and the initial alignment robustness and stability. The method achieves the most accurate results for the ACROBAT dataset, the cell-level registration accuracy for the restained slides from the HyReCo dataset, and is among the best methods evaluated on the ANHIR dataset. CONCLUSIONS The article presents an automatic and robust registration method that outperforms other state-of-the-art solutions. The method does not require any fine-tuning to a particular dataset and can be used out-of-the-box for numerous types of microscopic images. The method is incorporated into the DeeperHistReg framework, allowing others to directly use it to register, transform, and save the WSIs at any desired pyramid level (resolution up to 220k x 220k). We provide free access to the software. The results are fully and easily reproducible. The proposed method is a significant contribution to improving the WSI registration quality, thus advancing the field of digital pathology.
Collapse
Affiliation(s)
- Marek Wodzinski
- Institute of Informatics, University of Applied Sciences Western Switzerland, Sierre, Switzerland; Department of Measurement and Electronics, AGH University of Kraków, Krakow, Poland.
| | - Niccolò Marini
- Institute of Informatics, University of Applied Sciences Western Switzerland, Sierre, Switzerland
| | - Manfredo Atzori
- Institute of Informatics, University of Applied Sciences Western Switzerland, Sierre, Switzerland; Department of Neuroscience, University of Padova, Padova, Italy
| | - Henning Müller
- Institute of Informatics, University of Applied Sciences Western Switzerland, Sierre, Switzerland; Medical Faculty, University of Geneva, Geneva, Switzerland
| |
Collapse
|
37
|
Ouyang X, Matt A, Wang F, Gracheva E, Migunova E, Rajamani S, Dubrovsky EB, Zhou C. Attention LSTM U-Net model for Drosophila melanogaster heart tube segmentation in optical coherence microscopy images. BIOMEDICAL OPTICS EXPRESS 2024; 15:3639-3653. [PMID: 38867790 PMCID: PMC11166423 DOI: 10.1364/boe.523364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 06/14/2024]
Abstract
Optical coherence microscopy (OCM) imaging of the Drosophila melanogaster (fruit fly) heart tube has enabled the non-invasive characterization of fly heart physiology in vivo. OCM generates large volumes of data, making it necessary to automate image analysis. Deep-learning-based neural network models have been developed to improve the efficiency of fly heart image segmentation. However, image artifacts caused by sample motion or reflections reduce the accuracy of the analysis. To improve the precision and efficiency of image data analysis, we developed an Attention LSTM U-Net model (FlyNet3.0), which incorporates an attention learning mechanism to track the beating fly heart in OCM images. The new model has improved the intersection over union (IOU) compared to FlyNet2.0 + with reflection artifacts from 86% to 89% and with movement from 81% to 89%. We also extended the capabilities of OCM analysis through the introduction of an automated, in vivo heart wall thickness measurement method, which has been validated on a Drosophila model of cardiac hypertrophy. This work will enable the comprehensive, non-invasive characterization of fly heart physiology in a high-throughput manner.
Collapse
Affiliation(s)
- Xiangping Ouyang
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Abigail Matt
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Fei Wang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Elena Gracheva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Ekaterina Migunova
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Saathvika Rajamani
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | | | - Chao Zhou
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
38
|
Galaz-Montoya JG. The advent of preventive high-resolution structural histopathology by artificial-intelligence-powered cryogenic electron tomography. Front Mol Biosci 2024; 11:1390858. [PMID: 38868297 PMCID: PMC11167099 DOI: 10.3389/fmolb.2024.1390858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 05/08/2024] [Indexed: 06/14/2024] Open
Abstract
Advances in cryogenic electron microscopy (cryoEM) single particle analysis have revolutionized structural biology by facilitating the in vitro determination of atomic- and near-atomic-resolution structures for fully hydrated macromolecular complexes exhibiting compositional and conformational heterogeneity across a wide range of sizes. Cryogenic electron tomography (cryoET) and subtomogram averaging are rapidly progressing toward delivering similar insights for macromolecular complexes in situ, without requiring tags or harsh biochemical purification. Furthermore, cryoET enables the visualization of cellular and tissue phenotypes directly at molecular, nanometric resolution without chemical fixation or staining artifacts. This forward-looking review covers recent developments in cryoEM/ET and related technologies such as cryogenic focused ion beam milling scanning electron microscopy and correlative light microscopy, increasingly enhanced and supported by artificial intelligence algorithms. Their potential application to emerging concepts is discussed, primarily the prospect of complementing medical histopathology analysis. Machine learning solutions are poised to address current challenges posed by "big data" in cryoET of tissues, cells, and macromolecules, offering the promise of enabling novel, quantitative insights into disease processes, which may translate into the clinic and lead to improved diagnostics and targeted therapeutics.
Collapse
Affiliation(s)
- Jesús G. Galaz-Montoya
- Department of Bioengineering, James H. Clark Center, Stanford University, Stanford, CA, United States
| |
Collapse
|
39
|
Browning L, Jesus C, Malacrino S, Guan Y, White K, Puddle A, Alham NK, Haghighat M, Colling R, Birks J, Rittscher J, Verrill C. Artificial Intelligence-Based Quality Assessment of Histopathology Whole-Slide Images within a Clinical Workflow: Assessment of 'PathProfiler' in a Diagnostic Pathology Setting. Diagnostics (Basel) 2024; 14:990. [PMID: 38786288 PMCID: PMC11120465 DOI: 10.3390/diagnostics14100990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/17/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Digital pathology continues to gain momentum, with the promise of artificial intelligence to aid diagnosis and for assessment of features which may impact prognosis and clinical management. Successful adoption of these technologies depends upon the quality of digitised whole-slide images (WSI); however, current quality control largely depends upon manual assessment, which is inefficient and subjective. We previously developed PathProfiler, an automated image quality assessment tool, and in this feasibility study we investigate its potential for incorporation into a diagnostic clinical pathology setting in real-time. A total of 1254 genitourinary WSI were analysed by PathProfiler. PathProfiler was developed and trained on prostate tissue and, of the prostate biopsy WSI, representing 46% of the WSI analysed, 4.5% were flagged as potentially being of suboptimal quality for diagnosis. All had concordant subjective issues, mainly focus-related, 54% severe enough to warrant remedial action which resulted in improved image quality. PathProfiler was less reliable in assessment of non-prostate surgical resection-type cases, on which it had not been trained. PathProfiler shows potential for incorporation into a digitised clinical pathology workflow, with opportunity for image quality improvement. Whilst its reliability in the current form appears greatest for assessment of prostate specimens, other specimen types, particularly biopsies, also showed benefit.
Collapse
Affiliation(s)
- Lisa Browning
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Christine Jesus
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Stefano Malacrino
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Yue Guan
- Department of Cellular Pathology, Royal Berkshire Hospital, Royal Berkshire NHS Foundation Trust, Reading RG1 5AN, UK
| | - Kieron White
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Alison Puddle
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | | | - Maryam Haghighat
- School of Electrical Engineering and Robotics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Richard Colling
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Jacqueline Birks
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Jens Rittscher
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Clare Verrill
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| |
Collapse
|
40
|
Maes A, Borgel O, Braconnier C, Balcaen T, Wevers M, Halbgebauer R, Huber-Lang M, Kerckhofs G. X-Ray-Based 3D Histopathology of the Kidney Using Cryogenic Contrast-Enhanced MicroCT. Int J Biomed Imaging 2024; 2024:3924036. [PMID: 38634014 PMCID: PMC11022514 DOI: 10.1155/2024/3924036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 04/19/2024] Open
Abstract
The kidney's microstructure, which comprises a highly convoluted tubular and vascular network, can only be partially revealed using classical 2D histology. Considering that the kidney's microstructure is closely related to its function and is often affected by pathologies, there is a need for powerful and high-resolution 3D imaging techniques to visualize the microstructure. Here, we present how cryogenic contrast-enhanced microCT (cryo-CECT) allowed 3D visualization of glomeruli, tubuli, and vasculature. By comparing different contrast-enhancing staining agents and freezing protocols, we found that the preferred sample preparation protocol was the combination of staining with 1:2 hafnium(IV)-substituted Wells-Dawson polyoxometalate and freezing by submersion in isopentane at -78°C. This optimized protocol showed to be highly sensitive, allowing to detect small pathology-induced microstructural changes in a mouse model of mild trauma-related acute kidney injury after thorax trauma and hemorrhagic shock. In summary, we demonstrated that cryo-CECT is an effective 3D histopathological tool that allows to enhance our understanding of kidney tissue microstructure and their related function.
Collapse
Affiliation(s)
- Arne Maes
- Department of Materials Engineering, KU Leuven, Heverlee, Belgium
- Biomechanics Lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Onno Borgel
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| | - Clara Braconnier
- Biomechanics Lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
| | - Tim Balcaen
- Biomechanics Lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- MolDesignS, Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Martine Wevers
- Department of Materials Engineering, KU Leuven, Heverlee, Belgium
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| | - Greet Kerckhofs
- Department of Materials Engineering, KU Leuven, Heverlee, Belgium
- Biomechanics Lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Prometheus, Division for Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Joshi S, Forjaz A, Han KS, Shen Y, Queiroga V, Xenes D, Matelsk J, Wester B, Barrutia AM, Kiemen AL, Wu PH, Wirtz D. Generative interpolation and restoration of images using deep learning for improved 3D tissue mapping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583909. [PMID: 38496512 PMCID: PMC10942457 DOI: 10.1101/2024.03.07.583909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The development of novel imaging platforms has improved our ability to collect and analyze large three-dimensional (3D) biological imaging datasets. Advances in computing have led to an ability to extract complex spatial information from these data, such as the composition, morphology, and interactions of multi-cellular structures, rare events, and integration of multi-modal features combining anatomical, molecular, and transcriptomic (among other) information. Yet, the accuracy of these quantitative results is intrinsically limited by the quality of the input images, which can contain missing or damaged regions, or can be of poor resolution due to mechanical, temporal, or financial constraints. In applications ranging from intact imaging (e.g. light-sheet microscopy and magnetic resonance imaging) to sectioning based platforms (e.g. serial histology and serial section transmission electron microscopy), the quality and resolution of imaging data has become paramount. Here, we address these challenges by leveraging frame interpolation for large image motion (FILM), a generative AI model originally developed for temporal interpolation, for spatial interpolation of a range of 3D image types. Comparative analysis demonstrates the superiority of FILM over traditional linear interpolation to produce functional synthetic images, due to its ability to better preserve biological information including microanatomical features and cell counts, as well as image quality, such as contrast, variance, and luminance. FILM repairs tissue damages in images and reduces stitching artifacts. We show that FILM can decrease imaging time by synthesizing skipped images. We demonstrate the versatility of our method with a wide range of imaging modalities (histology, tissue-clearing/light-sheet microscopy, magnetic resonance imaging, serial section transmission electron microscopy), species (human, mouse), healthy and diseased tissues (pancreas, lung, brain), staining techniques (IHC, H&E), and pixel resolutions (8 nm, 2 μm, 1mm). Overall, we demonstrate the potential of generative AI in improving the resolution, throughput, and quality of biological image datasets, enabling improved 3D imaging.
Collapse
Affiliation(s)
- Saurabh Joshi
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Kyu Sang Han
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Yu Shen
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Vasco Queiroga
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Daniel Xenes
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD
| | - Jordan Matelsk
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD
| | - Brock Wester
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD
| | - Arrate Munoz Barrutia
- Bioengineering Department, Universidad Carlos III de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ashley L. Kiemen
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
| | - Denis Wirtz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore MD
- The Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
42
|
Kanwal N, López-Pérez M, Kiraz U, Zuiverloon TCM, Molina R, Engan K. Are you sure it's an artifact? Artifact detection and uncertainty quantification in histological images. Comput Med Imaging Graph 2024; 112:102321. [PMID: 38199127 DOI: 10.1016/j.compmedimag.2023.102321] [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: 07/10/2023] [Revised: 11/08/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Modern cancer diagnostics involves extracting tissue specimens from suspicious areas and conducting histotechnical procedures to prepare a digitized glass slide, called Whole Slide Image (WSI), for further examination. These procedures frequently introduce different types of artifacts in the obtained WSI, and histological artifacts might influence Computational Pathology (CPATH) systems further down to a diagnostic pipeline if not excluded or handled. Deep Convolutional Neural Networks (DCNNs) have achieved promising results for the detection of some WSI artifacts, however, they do not incorporate uncertainty in their predictions. This paper proposes an uncertainty-aware Deep Kernel Learning (DKL) model to detect blurry areas and folded tissues, two types of artifacts that can appear in WSIs. The proposed probabilistic model combines a CNN feature extractor and a sparse Gaussian Processes (GPs) classifier, which improves the performance of current state-of-the-art artifact detection DCNNs and provides uncertainty estimates. We achieved 0.996 and 0.938 F1 scores for blur and folded tissue detection on unseen data, respectively. In extensive experiments, we validated the DKL model on unseen data from external independent cohorts with different staining and tissue types, where it outperformed DCNNs. Interestingly, the DKL model is more confident in the correct predictions and less in the wrong ones. The proposed DKL model can be integrated into the preprocessing pipeline of CPATH systems to provide reliable predictions and possibly serve as a quality control tool.
Collapse
Affiliation(s)
- Neel Kanwal
- Department of Electrical Engineering and Computer Science, University of Stavanger, 4021 Stavanger, Norway.
| | - Miguel López-Pérez
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain
| | - Umay Kiraz
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021 Stavanger, Norway
| | - Tahlita C M Zuiverloon
- Department of Urology, University Medical Center Rotterdam, Erasmus MC Cancer Institute, 1035 GD Rotterdam, The Netherlands
| | - Rafael Molina
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain
| | - Kjersti Engan
- Department of Electrical Engineering and Computer Science, University of Stavanger, 4021 Stavanger, Norway
| |
Collapse
|
43
|
Tan YC, Mustangin M, Rosli N, Wan Ahmad Kammal WSE, Md Isa N, Low TY, Lee PY, Chellappan DK, Jarmin R, Zuhdi Z, Azman A, Ian C, Yusof NM, Lim LC. EtOH-LN cryoembedding workflow to minimize freezing artifact in frozen tissues: A pilot study in preparing tissues compatible with mass spectrometry-based spatial proteomics application. Cryobiology 2024; 114:104843. [PMID: 38158171 DOI: 10.1016/j.cryobiol.2023.104843] [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: 07/06/2023] [Revised: 11/27/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Coolant-assisted liquid nitrogen (LN) flash freezing of frozen tissues has been widely adopted to preserve tissue morphology for histopathological annotations in mass spectrometry-based spatial proteomics techniques. However, existing coolants pose health risks upon inhalation and are expensive. To overcome this challenge, we present our pilot study by introducing the EtOH-LN workflow, which demonstrates the feasibility of using 95 % ethanol as a safer and easily accessible alternative to existing coolants for LN-based cryoembedding of frozen tissues. Our study reveals that both the EtOH-LN and LN-only cryoembedding workflows exhibit significantly reduced freezing artifacts compared to cryoembedding in cryostat (p < 0.005), while EtOH-LN (SD = 0.56) generates more consistent results compared to LN-only (SD = 1.29). We have modified a previously reported morphology restoration method to incorporate the EtOH-LN workflow, which successfully restored the tissue architecture from freezing artifacts (p < 0.05). Additional studies are required to validate the impact of the EtOH-LN workflow on the molecular profiles of tissues.
Collapse
Affiliation(s)
- Yong Chiang Tan
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia.
| | - Muaatamarulain Mustangin
- Department of Pathology, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Nurwahyuna Rosli
- Department of Pathology, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | | | - Nurismah Md Isa
- Department of Pathology, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia.
| | - Razman Jarmin
- Department of Surgery, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Zamri Zuhdi
- Department of Surgery, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Azlanudin Azman
- Department of Surgery, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Chik Ian
- Department of Surgery, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Nursuhadah Mohamed Yusof
- Department of Surgery, UKM Medical Centre (UKMMC), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Lay Cheng Lim
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia.
| |
Collapse
|
44
|
Li Y, Pillar N, Li J, Liu T, Wu D, Sun S, Ma G, de Haan K, Huang L, Zhang Y, Hamidi S, Urisman A, Keidar Haran T, Wallace WD, Zuckerman JE, Ozcan A. Virtual histological staining of unlabeled autopsy tissue. Nat Commun 2024; 15:1684. [PMID: 38396004 PMCID: PMC10891155 DOI: 10.1038/s41467-024-46077-2] [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: 08/05/2023] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Traditional histochemical staining of post-mortem samples often confronts inferior staining quality due to autolysis caused by delayed fixation of cadaver tissue, and such chemical staining procedures covering large tissue areas demand substantial labor, cost and time. Here, we demonstrate virtual staining of autopsy tissue using a trained neural network to rapidly transform autofluorescence images of label-free autopsy tissue sections into brightfield equivalent images, matching hematoxylin and eosin (H&E) stained versions of the same samples. The trained model can effectively accentuate nuclear, cytoplasmic and extracellular features in new autopsy tissue samples that experienced severe autolysis, such as COVID-19 samples never seen before, where the traditional histochemical staining fails to provide consistent staining quality. This virtual autopsy staining technique provides a rapid and resource-efficient solution to generate artifact-free H&E stains despite severe autolysis and cell death, also reducing labor, cost and infrastructure requirements associated with the standard histochemical staining.
Collapse
Affiliation(s)
- Yuzhu Li
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| | - Nir Pillar
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| | - Jingxi Li
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| | - Tairan Liu
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| | - Di Wu
- Computer Science Department, University of California, Los Angeles, CA, 90095, USA
| | - Songyu Sun
- Computer Science Department, University of California, Los Angeles, CA, 90095, USA
| | - Guangdong Ma
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- School of Physics, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Kevin de Haan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| | - Luzhe Huang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| | - Yijie Zhang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| | - Sepehr Hamidi
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Anatoly Urisman
- Department of Pathology, University of California, San Francisco, CA, 94143, USA
| | - Tal Keidar Haran
- Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, 91120, Israel
| | - William Dean Wallace
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jonathan E Zuckerman
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Aydogan Ozcan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA, 90095, USA.
- Bioengineering Department, University of California, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA.
- Department of Surgery, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
45
|
Gouzou D, Taimori A, Haloubi T, Finlayson N, Wang Q, Hopgood JR, Vallejo M. Applications of machine learning in time-domain fluorescence lifetime imaging: a review. Methods Appl Fluoresc 2024; 12:022001. [PMID: 38055998 PMCID: PMC10851337 DOI: 10.1088/2050-6120/ad12f7] [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: 06/30/2023] [Revised: 09/25/2023] [Accepted: 12/06/2023] [Indexed: 12/08/2023]
Abstract
Many medical imaging modalities have benefited from recent advances in Machine Learning (ML), specifically in deep learning, such as neural networks. Computers can be trained to investigate and enhance medical imaging methods without using valuable human resources. In recent years, Fluorescence Lifetime Imaging (FLIm) has received increasing attention from the ML community. FLIm goes beyond conventional spectral imaging, providing additional lifetime information, and could lead to optical histopathology supporting real-time diagnostics. However, most current studies do not use the full potential of machine/deep learning models. As a developing image modality, FLIm data are not easily obtainable, which, coupled with an absence of standardisation, is pushing back the research to develop models which could advance automated diagnosis and help promote FLIm. In this paper, we describe recent developments that improve FLIm image quality, specifically time-domain systems, and we summarise sensing, signal-to-noise analysis and the advances in registration and low-level tracking. We review the two main applications of ML for FLIm: lifetime estimation and image analysis through classification and segmentation. We suggest a course of action to improve the quality of ML studies applied to FLIm. Our final goal is to promote FLIm and attract more ML practitioners to explore the potential of lifetime imaging.
Collapse
Affiliation(s)
- Dorian Gouzou
- Dorian Gouzou and Marta Vallejo are with Institute of Signals, Sensors and Systems, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, EH14 4AS, United Kingdom
| | - Ali Taimori
- Tarek Haloubi, Ali Taimori, and James R. Hopgood are with Institute for Imaging, Data and Communication, School of Engineering, University of Edinburgh, Edinburgh, EH9 3FG, United Kingdom
| | - Tarek Haloubi
- Tarek Haloubi, Ali Taimori, and James R. Hopgood are with Institute for Imaging, Data and Communication, School of Engineering, University of Edinburgh, Edinburgh, EH9 3FG, United Kingdom
| | - Neil Finlayson
- Neil Finlayson is with Institute for Integrated Micro and Nano Systems, School of Engineering, University ofEdinburgh, Edinburgh EH9 3FF, United Kingdom
| | - Qiang Wang
- Qiang Wang is with Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - James R Hopgood
- Tarek Haloubi, Ali Taimori, and James R. Hopgood are with Institute for Imaging, Data and Communication, School of Engineering, University of Edinburgh, Edinburgh, EH9 3FG, United Kingdom
| | - Marta Vallejo
- Dorian Gouzou and Marta Vallejo are with Institute of Signals, Sensors and Systems, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, EH14 4AS, United Kingdom
| |
Collapse
|
46
|
Tang H, Jiao J, Lin JD, Zhang X, Sun N. Detection of Large-Droplet Macrovesicular Steatosis in Donor Livers Based on Segment-Anything Model. J Transl Med 2024; 104:100288. [PMID: 37977550 DOI: 10.1016/j.labinv.2023.100288] [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: 07/27/2023] [Revised: 10/15/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
Liver transplantation is an effective treatment for end-stage liver disease, acute liver failure, and primary hepatic malignancy. However, the limited availability of donor organs remains a challenge. Severe large-droplet fat (LDF) macrovesicular steatosis, characterized by cytoplasmic replacement with large fat vacuoles, can lead to liver transplant complications. Artificial intelligence models, such as segmentation and detection models, are being developed to detect LDF hepatocytes. The Segment-Anything Model, utilizing the DEtection TRansformer architecture, has the ability to segment objects without prior knowledge of size or shape. We investigated the Segment-Anything Model's potential to detect LDF hepatocytes in liver biopsies. Pathologist-annotated specimens were used to evaluate model performance. The model showed high sensitivity but compromised specificity due to similarities with other structures. Filtering algorithms were developed to improve specificity. Integration of the Segment-Anything Model with rule-based algorithms accurately detected LDF hepatocytes. Improved diagnosis and treatment of liver diseases can be achieved through advancements in artificial intelligence algorithms for liver histology analysis.
Collapse
Affiliation(s)
- Haiming Tang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Jingjing Jiao
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Jian Denny Lin
- Department of Management Information System, College of Business, University of Houston Clear Lake, Houston, Texas
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut.
| | - Nanfei Sun
- Department of Management Information System, College of Business, University of Houston Clear Lake, Houston, Texas.
| |
Collapse
|
47
|
Scarpitti BT, Fan S, Lomax-Vogt M, Lutton A, Olesik JW, Schultz ZD. Accurate Quantification and Imaging of Cellular Uptake Using Single-Particle Surface-Enhanced Raman Scattering. ACS Sens 2024; 9:73-80. [PMID: 38100727 PMCID: PMC10958331 DOI: 10.1021/acssensors.3c01648] [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] [Indexed: 12/17/2023]
Abstract
Understanding the uptake, distribution, and stability of gold nanoparticles (NPs) in cells is of fundamental importance in nanoparticle sensors and therapeutic development. Single nanoparticle imaging with surface-enhanced Raman spectroscopy (SERS) measurements in cells is complicated by aggregation-dependent SERS signals, particle inhomogeneity, and limited single-particle brightness. In this work, we assess the single-particle SERS signals of various gold nanoparticle shapes and the role of silica encapsulation on SERS signals to develop a quantitative probe for single-particle level Raman imaging in living cells. We observe that silica-encapsulated gap-enhanced Raman tags (GERTs) provide an optimized probe that can be quantifiable per voxel in SERS maps of cells. This approach is validated by single-particle inductively coupled mass spectrometry (spICP-MS) measurements of NPs in cell lysate post-imaging. spICP-MS also provides a means of measuring the tag stability. This analytical approach can be used not only to quantitatively assess nanoparticle uptake on the cellular level (as in previous digital SERS methods) but also to reliably image the subcellular distribution and to assess the stability of NPs in cells.
Collapse
Affiliation(s)
- Brian T. Scarpitti
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Sanjun Fan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Madeleine Lomax-Vogt
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Anthony Lutton
- School of Earth Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - John W. Olesik
- School of Earth Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Zachary D. Schultz
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, 43210, USA
| |
Collapse
|
48
|
McKenzie AT, Thorn EL, Nnadi O, Wróbel B, Kendziorra E, Farrell K, Crary JF. Cryopreservation of brain cell structure: a review. FREE NEUROPATHOLOGY 2024; 5:35. [PMID: 39844781 PMCID: PMC11753176 DOI: 10.17879/freeneuropathology-2024-5883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/19/2024] [Indexed: 01/24/2025]
Abstract
Cryopreservation, the preservation of tissues at subzero temperatures, is a mainstay of brain banking that allows for the storage of brain tissue without the use of chemical fixatives. This is particularly important for molecular studies that are incompatible with tissue fixation. However, brain tissue is vulnerable to various forms of damage during the cryopreservation process, in particular due to the phase transition of water from a liquid to a solid state with the formation of ice crystals, which can disrupt cellular morphology. There is a critical need to characterize the effects of cryopreservation on brain cell structure at the microscopic level. In this review, we conducted a comprehensive literature search, identifying 97 studies that yielded 146 distinct observations of the effects of cryopreservation on neurohistology. We classified the reviewed studies into three main categories: cryofixation, freezing, and cryopreservation with cryoprotectants. Cryofixation techniques enable vitrification and excellent ultrastructural preservation of thin tissue samples but are limited in terms of the depth of tissue that can be preserved without ice artifacts. Freezing methods, particularly when applied to brain slices, can achieve rapid cooling rates that result in minimal ice artifacts detectable by light microscopy. Cryoprotectant-based approaches have the potential to reduce ice damage and achieve vitrification. For thin tissue samples, immersion in cryoprotectants has been found to be effective for structural preservation. However, for larger samples or the entire brain, perfusion of cryoprotectants is necessary to perform rapid distribution, and this has a more limited evidence base. In conclusion, while current cryopreservation methods can provide sufficient quality for some downstream applications, there is a need for improved techniques that enable the cryopreservation of larger brain tissue samples while maintaining excellent structural preservation.
Collapse
Affiliation(s)
| | - Emma L. Thorn
- Friedman Brain Institute, Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Oge Nnadi
- Brain Preservation Foundation, Ashburn, Virginia, USA
| | - Borys Wróbel
- European Institute for Brain Research, Amstelveen, The Netherlands
- BioPreservation Institute, Vancouver, Washington, USA
| | - Emil Kendziorra
- European Biostasis Foundation, Riehen, Canton of Basel-Stadt, Switzerland
| | - Kurt Farrell
- Friedman Brain Institute, Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John F. Crary
- Friedman Brain Institute, Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
49
|
Sanyal AJ, Jha P, Kleiner DE. Digital pathology for nonalcoholic steatohepatitis assessment. Nat Rev Gastroenterol Hepatol 2024; 21:57-69. [PMID: 37789057 DOI: 10.1038/s41575-023-00843-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 10/05/2023]
Abstract
Histological assessment of nonalcoholic fatty liver disease (NAFLD) has anchored knowledge development about the phenotypes of the condition, their natural history and their clinical course. This fact has led to the use of histological assessment as a reference standard for the evaluation of efficacy of drug interventions for nonalcoholic steatohepatitis (NASH) - the more histologically active form of NAFLD. However, certain limitations of conventional histological assessment systems pose challenges in drug development. These limitations have spurred intense scientific and commercial development of machine learning and digital approaches towards the assessment of liver histology in patients with NAFLD. This research field remains an area in rapid evolution. In this Perspective article, we summarize the current conventional assessment of NASH and its limitations, the use of specific digital approaches for histological assessment, and their application to the study of NASH and its response to therapy. Although this is not a comprehensive review, the leading tools currently used to assess therapeutic efficacy in drug development are specifically discussed. The potential translation of these approaches to support routine clinical assessment of NAFLD and an agenda for future research are also discussed.
Collapse
Affiliation(s)
- Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Prakash Jha
- Food and Drug Administration, Silver Spring, MD, USA
| | - David E Kleiner
- Post-Mortem Section Laboratory of Pathology Center for Cancer Research National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
Hacıhasanoglu E, Bambul Sıgırcı B, Usul G, Savlı TC. PD-L1 Assessment in Needle Core Biopsies of Non-Small Cell Lung Cancer: Interpathologist Agreement and Potential Associated Histopathological Features. Turk Patoloji Derg 2024; 40:37-44. [PMID: 37614090 PMCID: PMC10823782 DOI: 10.5146/tjpath.2023.01609] [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: 05/24/2023] [Accepted: 07/14/2023] [Indexed: 08/25/2023] Open
Abstract
OBJECTIVE Immune checkpoint inhibitors are used in the treatment of non-small cell lung cancer (NSCLC). Programmed cell death-ligand 1 (PD-L1) immunohistochemistry (IHC) assessed by pathologists is subject to interobserver variability. In advanced/metastatic disease and inoperable patients, PD-L1 assessment relies on biopsy specimens, commonly needle core biopsies (NCB). We aimed to determine the interobserver agreement for PD-L1 tumor proportion score (TPS) in NSCLC NCBs and identify histopathological features that may be related to interobserver variability. MATERIAL AND METHODS Sixty NSCLC NCBs with PD-L1 IHC were evaluated independently by four pathologists from different institutions. PD-L1 TPS was evaluated in three categories: no/low expression ( < 1%), intermediate expression (1%49%), and high expression (≥50%). Histological tumor type, necrosis, tumor-infiltrating lymphocytes, tumor length/percentage in the biopsy, and crush/squeeze artifact was evaluated. RESULTS The statistical analysis of the three PD-L1 TPS categories demonstrated moderate agreement (Fleiss Kappa 0.477) in the no/low category, fair agreement (Fleiss Kappa 0.390) in the intermediate category, and almost perfect agreement (Fleiss Kappa 0.952) in the high category. A significant correlation (p=0.003) was found between the crush/squeeze artifact in NCB and rate of discordant TPS categories. There was no significant correlation between pathologists' agreement in the TPS categories and histological tumor type, tumor length, tumor ratio, necrosis, and tumor-infiltrating lymphocytes. CONCLUSION Our results demonstrated moderate agreement among pathologists for the PD-L1 TPS 1% cut-off in NSCLC NCB, which is lower than that reported in resection materials. The presence of crush/squeeze artifact in NCBs is significantly related to the rate of discordant TPS categories, suggesting that PD-L1 assessment of pulmonary NCBs requires an awareness of this artifact.
Collapse
Affiliation(s)
- Ezgi Hacıhasanoglu
- Department of Pathology, 1Yeditepe University, School of Medicine, İstanbul, Turkey
| | - Buket Bambul Sıgırcı
- University of Health Sciences, Sisli Hamidiye Etfal Training Hospital, İstanbul, Turkey
| | - Gamze Usul
- Basaksehir Cam and Sakura City Hospital, İstanbul, Turkey
| | | |
Collapse
|