1
|
Zhu E, Li YR, Margolis S, Wang J, Wang K, Zhang Y, Wang S, Park J, Zheng C, Yang L, Chu A, Zhang Y, Gao L, Hsiai TK. Frontiers in artificial intelligence-directed light-sheet microscopy for uncovering biological phenomena and multi-organ imaging. VIEW 2024; 5:20230087. [PMID: 39478956 PMCID: PMC11521201 DOI: 10.1002/viw.20230087] [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: 04/02/2024] [Accepted: 07/18/2024] [Indexed: 11/02/2024] Open
Abstract
Light-sheet fluorescence microscopy (LSFM) introduces fast scanning of biological phenomena with deep photon penetration and minimal phototoxicity. This advancement represents a significant shift in 3-D imaging of large-scale biological tissues and 4-D (space + time) imaging of small live animals. The large data associated with LSFM requires efficient imaging acquisition and analysis with the use of artificial intelligence (AI)/machine learning (ML) algorithms. To this end, AI/ML-directed LSFM is an emerging area for multi-organ imaging and tumor diagnostics. This review will present the development of LSFM and highlight various LSFM configurations and designs for multi-scale imaging. Optical clearance techniques will be compared for effective reduction in light scattering and optimal deep-tissue imaging. This review will further depict a diverse range of research and translational applications, from small live organisms to multi-organ imaging to tumor diagnosis. In addition, this review will address AI/ML-directed imaging reconstruction, including the application of convolutional neural networks (CNNs) and generative adversarial networks (GANs). In summary, the advancements of LSFM have enabled effective and efficient post-imaging reconstruction and data analyses, underscoring LSFM's contribution to advancing fundamental and translational research.
Collapse
Affiliation(s)
- Enbo Zhu
- Department of Bioengineering, UCLA, California, 90095, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, California, 90095, USA
- Department of Medicine, Greater Los Angeles VA Healthcare System, California, 90073, USA
- Department of Microbiology, Immunology & Molecular Genetics, UCLA, California, 90095, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, UCLA, California, 90095, USA
| | - Samuel Margolis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, California, 90095, USA
| | - Jing Wang
- Department of Bioengineering, UCLA, California, 90095, USA
| | - Kaidong Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, California, 90095, USA
- Department of Medicine, Greater Los Angeles VA Healthcare System, California, 90073, USA
| | - Yaran Zhang
- Department of Bioengineering, UCLA, California, 90095, USA
| | - Shaolei Wang
- Department of Bioengineering, UCLA, California, 90095, USA
| | - Jongchan Park
- Department of Bioengineering, UCLA, California, 90095, USA
| | - Charlie Zheng
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, California, 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, UCLA, California, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, California, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, California, 90095, USA
- Molecular Biology Institute, UCLA, California, 90095, USA
| | - Alison Chu
- Division of Neonatology and Developmental Biology, Department of Pediatrics, David Geffen School of Medicine, UCLA, California, 90095, USA
| | - Yuhua Zhang
- Doheny Eye Institute, Department of Ophthalmology, UCLA, California, 90095, USA
| | - Liang Gao
- Department of Bioengineering, UCLA, California, 90095, USA
| | - Tzung K. Hsiai
- Department of Bioengineering, UCLA, California, 90095, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, California, 90095, USA
- Department of Medicine, Greater Los Angeles VA Healthcare System, California, 90073, USA
| |
Collapse
|
2
|
Almasian M, Saberigarakani A, Zhang X, Lee B, Ding Y. Light-Sheet Imaging to Reveal Cardiac Structure in Rodent Hearts. J Vis Exp 2024:10.3791/66707. [PMID: 38619234 PMCID: PMC11027943 DOI: 10.3791/66707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Light-sheet microscopy (LSM) plays a pivotal role in comprehending the intricate three-dimensional (3D) structure of the heart, providing crucial insights into fundamental cardiac physiology and pathologic responses. We hereby delve into the development and implementation of the LSM technique to elucidate the micro-architecture of the heart in mouse models. The methodology integrates a customized LSM system with tissue clearing techniques, mitigating light scattering within cardiac tissues for volumetric imaging. The combination of conventional LSM with image stitching and multiview deconvolution approaches allows for the capture of the entire heart. To address the inherent trade-off between axial resolution and field of view (FOV), we further introduce an axially swept light-sheet microscopy (ASLM) method to minimize out-of-focus light and uniformly illuminate the heart across the propagation direction. In the meanwhile, tissue clearing methods such as iDISCO enhance light penetration, facilitating the visualization of deep structures and ensuring a comprehensive examination of the myocardium throughout the entire heart. The combination of the proposed LSM and tissue clearing methods presents a promising platform for researchers in resolving cardiac structures in rodent hearts, holding great potential for the understanding of cardiac morphogenesis and remodeling.
Collapse
Affiliation(s)
- Milad Almasian
- Department of Bioengineering, The University of Texas at Dallas
| | | | - Xinyuan Zhang
- Department of Bioengineering, The University of Texas at Dallas
| | - Brian Lee
- Department of Bioengineering, The University of Texas at Dallas
| | - Yichen Ding
- Department of Bioengineering, The University of Texas at Dallas; Center for Imaging and Surgical Innovation, The University of Texas at Dallas; Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center;
| |
Collapse
|
3
|
Brooks HL, de Castro Brás LE, Brunt KR, Sylvester MA, Parvatiyar MS, Sirish P, Bansal SS, Sule R, Eadie AL, Knepper MA, Fenton RA, Lindsey ML, DeLeon-Pennell KY, Gomes AV. Guidelines on antibody use in physiology research. Am J Physiol Renal Physiol 2024; 326:F511-F533. [PMID: 38234298 PMCID: PMC11208033 DOI: 10.1152/ajprenal.00347.2023] [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: 10/30/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 01/19/2024] Open
Abstract
Antibodies are one of the most used reagents in scientific laboratories and are critical components for a multitude of experiments in physiology research. Over the past decade, concerns about many biological methods, including those that use antibodies, have arisen as several laboratories were unable to reproduce the scientific data obtained in other laboratories. The lack of reproducibility could be largely attributed to inadequate reporting of detailed methods, no or limited verification by authors, and the production and use of unvalidated antibodies. The goal of this guideline article is to review best practices concerning commonly used techniques involving antibodies, including immunoblotting, immunohistochemistry, and flow cytometry. Awareness and integration of best practices will increase the rigor and reproducibility of these techniques and elevate the quality of physiology research.
Collapse
Affiliation(s)
- Heddwen L Brooks
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | | | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Megan A Sylvester
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona, United States
| | - Michelle S Parvatiyar
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California, United States
| | - Shyam S Bansal
- Department of Cellular and Molecular Physiology, Heart and Vascular Institute, Pennsylvania State University Hershey Medical Center, Hershey, Pennsylvania, United States
| | - Rasheed Sule
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, United States
| | - Ashley L Eadie
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Merry L Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
- Research Service, Nashville Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, School of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Research Service, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, United States
| |
Collapse
|
4
|
Zhang X, Saberigarakani A, Almasian M, Hassan S, Nekkanti M, Ding Y. 4D Light-sheet Imaging of Zebrafish Cardiac Contraction. J Vis Exp 2024:10.3791/66263. [PMID: 38251787 PMCID: PMC10939705 DOI: 10.3791/66263] [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: 01/23/2024] Open
Abstract
Zebrafish is an intriguing model organism known for its remarkable cardiac regeneration capacity. Studying the contracting heart in vivo is essential for gaining insights into structural and functional changes in response to injuries. However, obtaining high-resolution and high-speed 4-dimensional (4D, 3D spatial + 1D temporal) images of the zebrafish heart to assess cardiac architecture and contractility remains challenging. In this context, an in-house light-sheet microscope (LSM) and customized computational analysis are used to overcome these technical limitations. This strategy, involving LSM system construction, retrospective synchronization, single cell tracking, and user-directed analysis, enables one to investigate the micro-structure and contractile function across the entire heart at the single-cell resolution in the transgenic Tg(myl7:nucGFP) zebrafish larvae. Additionally, we are able to further incorporate microinjection of small molecule compounds to induce cardiac injury in a precise and controlled manner. Overall, this framework allows one to track physiological and pathophysiological changes, as well as the regional mechanics at the single-cell level during cardiac morphogenesis and regeneration.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Bioengineering, The University of Texas at Dallas
| | | | - Milad Almasian
- Department of Bioengineering, The University of Texas at Dallas
| | - Sohail Hassan
- Department of Bioengineering, The University of Texas at Dallas
| | - Manasa Nekkanti
- Department of Bioengineering, The University of Texas at Dallas
| | - Yichen Ding
- Department of Bioengineering, The University of Texas at Dallas; Center for Imaging and Surgical Innovation, The University of Texas at Dallas; Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center;
| |
Collapse
|
5
|
Gonzalez-Ramos S, Wang J, Cho JM, Zhu E, Park SK, In JG, Reddy ST, Castillo EF, Campen MJ, Hsiai TK. Integrating 4-D light-sheet fluorescence microscopy and genetic zebrafish system to investigate ambient pollutants-mediated toxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 902:165947. [PMID: 37543337 PMCID: PMC10659062 DOI: 10.1016/j.scitotenv.2023.165947] [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: 05/25/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/07/2023]
Abstract
Ambient air pollutants, including PM2.5 (aerodynamic diameter d ~2.5 μm), PM10 (d ~10 μm), and ultrafine particles (UFP: d < 0.1 μm) impart both short- and long-term toxicity to various organs, including cardiopulmonary, central nervous, and gastrointestinal systems. While rodents have been the principal animal model to elucidate air pollution-mediated organ dysfunction, zebrafish (Danio rerio) is genetically tractable for its short husbandry and life cycle to study ambient pollutants. Its electrocardiogram (ECG) resembles that of humans, and the fluorescent reporter-labeled tissues in the zebrafish system allow for screening a host of ambient pollutants that impair cardiovascular development, organ regeneration, and gut-vascular barriers. In parallel, the high spatiotemporal resolution of light-sheet fluorescence microscopy (LSFM) enables investigators to take advantage of the transparent zebrafish embryos and genetically labeled fluorescent reporters for imaging the dynamic cardiac structure and function at a single-cell resolution. In this context, our review highlights the integrated strengths of the genetic zebrafish system and LSFM for high-resolution and high-throughput investigation of ambient pollutants-mediated cardiac and intestinal toxicity.
Collapse
Affiliation(s)
- Sheila Gonzalez-Ramos
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA; Department of Bioengineering, School of Engineering & Applied Science, University of California, Los Angeles, CA, USA
| | - Jing Wang
- Department of Bioengineering, School of Engineering & Applied Science, University of California, Los Angeles, CA, USA
| | - Jae Min Cho
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Enbo Zhu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Seul-Ki Park
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Julie G In
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Degree Program, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Eliseo F Castillo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Tzung K Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA; Department of Bioengineering, School of Engineering & Applied Science, University of California, Los Angeles, CA, USA; Greater Los Angeles VA Healthcare System, Department of Medicine, Los Angeles, California, USA.
| |
Collapse
|
6
|
Campos Pamplona C, Moers C, Leuvenink HGD, van Leeuwen LL. Expanding the Horizons of Pre-Transplant Renal Vascular Assessment Using Ex Vivo Perfusion. Curr Issues Mol Biol 2023; 45:5437-5459. [PMID: 37504261 PMCID: PMC10378498 DOI: 10.3390/cimb45070345] [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: 06/01/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Recently, immense efforts have focused on improving the preservation of (sub)optimal donor organs by means of ex vivo perfusion, which enables the opportunity for organ reconditioning and viability assessment. However, there is still no biomarker that correlates with renal viability. Therefore, it is essential to explore new techniques for pre-transplant assessment of organ quality to guarantee successful long-term transplantation outcomes. The renal vascular compartment has received little attention in machine perfusion studies. In vivo, proper renal vascular and endothelial function is essential for maintaining homeostasis and long-term graft survival. In an ex vivo setting, little is known about vascular viability and its implications for an organ's suitability for transplant. Seeing that endothelial damage is the first step in a cascade of disruptions and maintaining homeostasis is crucial for positive post-transplant outcomes, further research is key to clarifying the (patho)physiology of the renal vasculature during machine perfusion. In this review, we aim to summarize key aspects of renal vascular physiology, describe the role of the renal vasculature in pathophysiological settings, and explain how ex vivo perfusion plays a role in either unveiling or targeting such processes. Additionally, we discuss potentially new vascular assessment tools during ex vivo renal perfusion.
Collapse
Affiliation(s)
- Carolina Campos Pamplona
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Cyril Moers
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - L Leonie van Leeuwen
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
7
|
Zhang X, Almasian M, Hassan SS, Jotheesh R, Kadam VA, Polk AR, Saberigarakani A, Rahat A, Yuan J, Lee J, Carroll K, Ding Y. 4D Light-sheet imaging and interactive analysis of cardiac contractility in zebrafish larvae. APL Bioeng 2023; 7:026112. [PMID: 37351330 PMCID: PMC10283270 DOI: 10.1063/5.0153214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023] Open
Abstract
Despite ongoing efforts in cardiovascular research, the acquisition of high-resolution and high-speed images for the purpose of assessing cardiac contraction remains challenging. Light-sheet fluorescence microscopy (LSFM) offers superior spatiotemporal resolution and minimal photodamage, providing an indispensable opportunity for the in vivo study of cardiac micro-structure and contractile function in zebrafish larvae. To track the myocardial architecture and contractility, we have developed an imaging strategy ranging from LSFM system construction, retrospective synchronization, single cell tracking, to user-directed virtual reality (VR) analysis. Our system enables the four-dimensional (4D) investigation of individual cardiomyocytes across the entire atrium and ventricle during multiple cardiac cycles in a zebrafish larva at the cellular resolution. To enhance the throughput of our model reconstruction and assessment, we have developed a parallel computing-assisted algorithm for 4D synchronization, resulting in a nearly tenfold enhancement of reconstruction efficiency. The machine learning-based nuclei segmentation and VR-based interaction further allow us to quantify cellular dynamics in the myocardium from end-systole to end-diastole. Collectively, our strategy facilitates noninvasive cardiac imaging and user-directed data interpretation with improved efficiency and accuracy, holding great promise to characterize functional changes and regional mechanics at the single cell level during cardiac development and regeneration.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Milad Almasian
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Sohail S. Hassan
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Rosemary Jotheesh
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Vinay A. Kadam
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Austin R. Polk
- Department of Computer Science, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Alireza Saberigarakani
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Aayan Rahat
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Jie Yuan
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Juhyun Lee
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas 76019, USA
| | - Kelli Carroll
- Department of Biology, Austin College, Sherman, Texas 75090, USA
| | - Yichen Ding
- Author to whom correspondence should be addressed:. Tel.: 972–883-7217
| |
Collapse
|
8
|
Cho JM, Poon MLS, Zhu E, Wang J, Butcher JT, Hsiai T. Quantitative 4D imaging of biomechanical regulation of ventricular growth and maturation. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 26:100438. [PMID: 37424697 PMCID: PMC10327868 DOI: 10.1016/j.cobme.2022.100438] [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] [Indexed: 12/24/2022]
Abstract
Abnormal cardiac development is intimately associated with congenital heart disease. During development, a sponge-like network of muscle fibers in the endocardium, known as trabeculation, becomes compacted. Biomechanical forces regulate myocardial differentiation and proliferation to form trabeculation, while the molecular mechanism is still enigmatic. Biomechanical forces, including intracardiac hemodynamic flow and myocardial contractile force, activate a host of molecular signaling pathways to mediate cardiac morphogenesis. While mechanotransduction pathways to initiate ventricular trabeculation is well studied, deciphering the relative importance of hemodynamic shear vs. mechanical contractile forces to modulate the transition from trabeculation to compaction requires advanced imaging tools and genetically tractable animal models. For these reasons, the advent of 4-D multi-scale light-sheet imaging and complementary multiplex live imaging via micro-CT in the beating zebrafish heart and live chick embryos respectively. Thus, this review highlights the complementary animal models and advanced imaging needed to elucidate the mechanotransduction underlying cardiac ventricular development.
Collapse
Affiliation(s)
- Jae Min Cho
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA
- Department of Medicine, Greater Los Angeles VA Healthcare System
| | - Mong Lung Steve Poon
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University
| | - Enbo Zhu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA
- Department of Medicine, Greater Los Angeles VA Healthcare System
| | | | - Jonathan T. Butcher
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University
| | - Tzung Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA
- Department of Medicine, Greater Los Angeles VA Healthcare System
- Department of Bioengineering, UCLA
| |
Collapse
|
9
|
Sodimu O, Almasian M, Gan P, Hassan S, Zhang X, Liu N, Ding Y. Light sheet imaging and interactive analysis of the cardiac structure in neonatal mice. JOURNAL OF BIOPHOTONICS 2023; 16:e202200278. [PMID: 36624523 PMCID: PMC10192002 DOI: 10.1002/jbio.202200278] [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: 09/05/2022] [Revised: 11/25/2022] [Accepted: 12/24/2022] [Indexed: 05/17/2023]
Abstract
Light-sheet microscopy (LSM) enables us to strengthen the understanding of cardiac development, injury, and regeneration in mammalian models. This emerging technique decouples laser illumination and fluorescence detection to investigate cardiac micro-structure and function with a high spatial resolution while minimizing photodamage and maximizing penetration depth. To unravel the potential of volumetric imaging in cardiac development and repair, we sought to integrate our in-house LSM, Adipo-Clear, and virtual reality (VR) with neonatal mouse hearts. We demonstrate the use of Adipo-Clear to render mouse hearts transparent, the development of our in-house LSM to capture the myocardial architecture within the intact heart, and the integration of VR to explore, measure, and assess regions of interests in an interactive manner. Collectively, we have established an innovative and holistic strategy for image acquisition and interpretation, providing an entry point to assess myocardial micro-architecture throughout the entire mammalian heart for the understanding of cardiac morphogenesis.
Collapse
Affiliation(s)
- Oluwatofunmi Sodimu
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Milad Almasian
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Peiheng Gan
- Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sohail Hassan
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Xinyuan Zhang
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Ning Liu
- Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yichen Ding
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX 75080, USA
- Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Imaging and Surgical Innovation, The University of Texas at Dallas, Richardson, TX, 75080, USA
| |
Collapse
|
10
|
Du X, Chen Z, Li Q, Yang S, Jiang L, Yang Y, Li Y, Gu Z. Organoids revealed: morphological analysis of the profound next generation in-vitro model with artificial intelligence. Biodes Manuf 2023; 6:319-339. [PMID: 36713614 PMCID: PMC9867835 DOI: 10.1007/s42242-022-00226-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/06/2022] [Indexed: 01/21/2023]
Abstract
In modern terminology, "organoids" refer to cells that grow in a specific three-dimensional (3D) environment in vitro, sharing similar structures with their source organs or tissues. Observing the morphology or growth characteristics of organoids through a microscope is a commonly used method of organoid analysis. However, it is difficult, time-consuming, and inaccurate to screen and analyze organoids only manually, a problem which cannot be easily solved with traditional technology. Artificial intelligence (AI) technology has proven to be effective in many biological and medical research fields, especially in the analysis of single-cell or hematoxylin/eosin stained tissue slices. When used to analyze organoids, AI should also provide more efficient, quantitative, accurate, and fast solutions. In this review, we will first briefly outline the application areas of organoids and then discuss the shortcomings of traditional organoid measurement and analysis methods. Secondly, we will summarize the development from machine learning to deep learning and the advantages of the latter, and then describe how to utilize a convolutional neural network to solve the challenges in organoid observation and analysis. Finally, we will discuss the limitations of current AI used in organoid research, as well as opportunities and future research directions. Graphic abstract
Collapse
Affiliation(s)
- Xuan Du
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009 China
| | - Lincao Jiang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Yi Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Yanhui Li
- State Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, 210008 China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| |
Collapse
|
11
|
Visualization of regenerating and repairing hearts. Clin Sci (Lond) 2022; 136:787-798. [PMID: 35621122 PMCID: PMC9886236 DOI: 10.1042/cs20211116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023]
Abstract
With heart failure continuing to become more prevalent, investigating the mechanisms of heart injury and repair holds much incentive. In contrast with adult mammals, other organisms such as teleost fish, urodele amphibians, and even neonatal mammals are capable of robust cardiac regeneration to replenish lost or damaged myocardial tissue. Long-term high-resolution intravital imaging of the behaviors and interactions of different cardiac cell types in their native environment could yield unprecedented insights into heart regeneration and repair. However, this task remains challenging for the heart due to its rhythmic contraction and anatomical location. Here, we summarize recent advances in live imaging of heart regeneration and repair, discuss the advantages and limitations of current systems, and suggest future directions for novel imaging technology development.
Collapse
|
12
|
Roustaei M, In Baek K, Wang Z, Cavallero S, Satta S, Lai A, O'Donnell R, Vedula V, Ding Y, Marsden AL, Hsiai TK. Computational simulations of the 4D micro-circulatory network in zebrafish tail amputation and regeneration. J R Soc Interface 2022; 19:20210898. [PMID: 35167770 PMCID: PMC8848759 DOI: 10.1098/rsif.2021.0898] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/12/2022] [Indexed: 12/16/2022] Open
Abstract
Wall shear stress (WSS) contributes to the mechanotransduction underlying microvascular development and regeneration. Using computational fluid dynamics, we elucidated the interplay between WSS and vascular remodelling in a zebrafish model of tail amputation and regeneration. The transgenic Tg (fli1:eGFP; Gata1:ds-red) zebrafish line was used to track the three-dimensional fluorescently labelled vascular endothelium for post-image segmentation and reconstruction of the fluid domain. Particle image velocimetry was used to validate the blood flow. Following amputation to the dorsal aorta and posterior cardinal vein (PCV), vasoconstriction developed in the dorsal longitudinal anastomotic vessel (DLAV) along with increased WSS in the proximal segmental vessels (SVs) from amputation. Angiogenesis ensued at the tips of the amputated DLAV and PCV where WSS was minimal. At 2 days post amputation (dpa), vasodilation occurred in a pair of SVs proximal to amputation, followed by increased blood flow and WSS; however, in the SVs distal to amputation, WSS normalized to the baseline. At 3 dpa, the blood flow increased in the arterial SV proximal to amputation and through anastomosis with DLAV formed a loop with PCV. Thus, our in silico modelling revealed the interplay between WSS and microvascular adaptation to changes in WSS and blood flow to restore microcirculation following tail amputation.
Collapse
Affiliation(s)
- Mehrdad Roustaei
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kyung In Baek
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zhaoqiang Wang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Susana Cavallero
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sandro Satta
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Angela Lai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ryan O'Donnell
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Vijay Vedula
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Yichen Ding
- Department of Bioengineering, University of Texas Dallas, Dallas, TX, USA
| | | | - Tzung K. Hsiai
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
13
|
Engelbrecht L, Ollewagen T, de Swardt D. Advances in fluorescence microscopy can reveal important new aspects of tissue regeneration. Biochimie 2022; 196:194-202. [DOI: 10.1016/j.biochi.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
14
|
Roustaei M, In Baek K, Wang Z, Cavallero S, Satta S, Lai A, O'Donnell R, Vedula V, Ding Y, Marsden AL, Hsiai TK. Computational simulations of the 4D micro-circulatory network in zebrafish tail amputation and regeneration. J R Soc Interface 2022. [PMID: 35167770 DOI: 10.1101/2021.02.10.430654v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
Wall shear stress (WSS) contributes to the mechanotransduction underlying microvascular development and regeneration. Using computational fluid dynamics, we elucidated the interplay between WSS and vascular remodelling in a zebrafish model of tail amputation and regeneration. The transgenic Tg (fli1:eGFP; Gata1:ds-red) zebrafish line was used to track the three-dimensional fluorescently labelled vascular endothelium for post-image segmentation and reconstruction of the fluid domain. Particle image velocimetry was used to validate the blood flow. Following amputation to the dorsal aorta and posterior cardinal vein (PCV), vasoconstriction developed in the dorsal longitudinal anastomotic vessel (DLAV) along with increased WSS in the proximal segmental vessels (SVs) from amputation. Angiogenesis ensued at the tips of the amputated DLAV and PCV where WSS was minimal. At 2 days post amputation (dpa), vasodilation occurred in a pair of SVs proximal to amputation, followed by increased blood flow and WSS; however, in the SVs distal to amputation, WSS normalized to the baseline. At 3 dpa, the blood flow increased in the arterial SV proximal to amputation and through anastomosis with DLAV formed a loop with PCV. Thus, our in silico modelling revealed the interplay between WSS and microvascular adaptation to changes in WSS and blood flow to restore microcirculation following tail amputation.
Collapse
Affiliation(s)
- Mehrdad Roustaei
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kyung In Baek
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zhaoqiang Wang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Susana Cavallero
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sandro Satta
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Angela Lai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ryan O'Donnell
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Vijay Vedula
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Yichen Ding
- Department of Bioengineering, University of Texas Dallas, Dallas, TX, USA
| | | | - Tzung K Hsiai
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Division of Cardiology, Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
15
|
Zopf LM, Heimel P, Geyer SH, Kavirayani A, Reier S, Fröhlich V, Stiglbauer-Tscholakoff A, Chen Z, Nics L, Zinnanti J, Drexler W, Mitterhauser M, Helbich T, Weninger WJ, Slezak P, Obenauf A, Bühler K, Walter A. Cross-Modality Imaging of Murine Tumor Vasculature-a Feasibility Study. Mol Imaging Biol 2021. [PMID: 34101107 DOI: 10.1007/s11307-021-01615-y/figures/6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Tumor vasculature and angiogenesis play a crucial role in tumor progression. Their visualization is therefore of utmost importance to the community. In this proof-of-principle study, we have established a novel cross-modality imaging (CMI) pipeline to characterize exactly the same murine tumors across scales and penetration depths, using orthotopic models of melanoma cancer. This allowed the acquisition of a comprehensive set of vascular parameters for a single tumor. The workflow visualizes capillaries at different length scales, puts them into the context of the overall tumor vessel network and allows quantification and comparison of vessel densities and morphologies by different modalities. The workflow adds information about hypoxia and blood flow rates. The CMI approach includes well-established technologies such as magnetic resonance imaging (MRI), positron emission tomography (PET), computed tomography (CT), and ultrasound (US), and modalities that are recent entrants into preclinical discovery such as optical coherence tomography (OCT) and high-resolution episcopic microscopy (HREM). This novel CMI platform establishes the feasibility of combining these technologies using an extensive image processing pipeline. Despite the challenges pertaining to the integration of microscopic and macroscopic data across spatial resolutions, we also established an open-source pipeline for the semi-automated co-registration of the diverse multiscale datasets, which enables truly correlative vascular imaging. Although focused on tumor vasculature, our CMI platform can be used to tackle a multitude of research questions in cancer biology.
Collapse
Affiliation(s)
- Lydia M Zopf
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | - Patrick Heimel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Austrian BioImaging/CMI, Vienna, Austria
- Core Facility Hard Tissue and Biomaterial Research, Karl Donath Laboratory, University Clinic of Dentistry, Medical University Vienna, Vienna, Austria
| | - Stefan H Geyer
- Division of Anatomy, MIC, Medical University of Vienna, Austrian BioImaging/CMI, Vienna, Austria
| | - Anoop Kavirayani
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | - Susanne Reier
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | - Vanessa Fröhlich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Vienna, Austria
| | - Alexander Stiglbauer-Tscholakoff
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Vienna, Austria
| | - Zhe Chen
- Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Medical University of Vienna, Vienna, Austria
| | - Jelena Zinnanti
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | | | - Markus Mitterhauser
- Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Thomas Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Vienna, Austria
| | - Wolfgang J Weninger
- Division of Anatomy, MIC, Medical University of Vienna, Austrian BioImaging/CMI, Vienna, Austria
| | - Paul Slezak
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Austrian BioImaging/CMI, Vienna, Austria
| | - Anna Obenauf
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Katja Bühler
- VRVis Zentrum für Virtual Reality und Visualisierung Forschungs-GmbH, Austrian BioImaging/CMI, Vienna, Austria
| | - Andreas Walter
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria.
| |
Collapse
|
16
|
Ha Y, Ochoa LF, Solomon O, Shi S, Villarreal PP, Li S, Buscho S, Vargas G, Zhang W. Light-Sheet Microscopy of the Optic Nerve Reveals Axonal Degeneration and Microglial Activation in NMDA-Induced Retinal Injury. EC OPHTHALMOLOGY 2021; 12:23-31. [PMID: 36108311 PMCID: PMC9450914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
PURPOSE Optic nerve degeneration is a feature of neurodegenerative eye diseases and causes irreversible vision loss. Therefore, understanding the degenerating patterns of the optic nerve is critical to find the potential therapeutic target for optic neuropathy. However, the traditional method of optic nerve degeneration has the limitations of losing spatiotemporal tissue information. Light sheet fluorescence microscopy (LSFM) is a fluorescence microscopy technique that allows capturing 3D images rapidly with a high spatial optical resolution. In this study, we evaluated the availability of LSFM on the optic nerve with NMDA injected Thy1-CFP mice. METHODS NMDA injected to both eyes of Thy1-CFP mice. After 7 days from the injection, the retina and optic nerve were collected and immunostained with anti-Iba1 antibody. NMDA excitotoxicity induced RGC, and its axon loss and microglial activation in the retina were observed using confocal microscopy. The immunostained optic nerve was completed the optical clearing process with TDE and mounted for LSFM imaging. RESULTS We found that retinal flatmounts confirmed significant loss of CFP-expressing RGC and axon degradation and loss in Thy1-CFP mice at 7 days after NMDA injection. Together with these data verifying that NMDA induces RGC and its axon loss, we confirmed that NMDA excitotoxicity induced microglia activation and leukostasis, such as increased microglia number, transform its morphology to ameboid or round, and increase in attached leukocytes in vessels. Using LSFM, we observed that CFP expressing nerve fiber was well organized and arranged parallel in vehicle treated optic nerve, whileas NMDA injected optic nerve showed axon swelling and fragmentation and loss of axon density from the anterior to the posterior regions. Furthermore, LSFM enabled the observation of microglia phenotype transformation in the entire optic nerve. Unlike microglia in vehicle injected optic nerve, microglia in NMDA injected optic nerve displayed larger soma and short process with high Iba1 expression through the entire optic nerve from the anterior to posterior. CONCLUSIONS In summary, we examined the applicability of the modified optic clearing protocol for the optic nerve and verified it enabled to acquiring of the 3D images of the optic nerve successfully revealing the complex spatial relationships between the axons, microglia and vasculature throughout the entire organ with single acquisitions. With these optimized techniques, we successfully obtained the high-resolution 3D images of NMDA-induced optic neuropathy, including the clues for optic nerve degeneration such as axon swelling, axonal fragmentation, and microglia activation. Overall, we believe that our current study could help understand the pathology of the optic nerve in neurodegenerative diseases, and it will be the basis for translational research.
Collapse
Affiliation(s)
- Yonju Ha
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lorenzo F Ochoa
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Olivia Solomon
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
- Human Pathophysiology and Translational Medicine Graduate Program, University of Texas Medical Branch, Galveston, Texas, USA
| | - Shuizhen Shi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Paula P Villarreal
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Shengguo Li
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Seth Buscho
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Gracie Vargas
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
17
|
Wang Z, Ding Y, Satta S, Roustaei M, Fei P, Hsiai TK. A hybrid of light-field and light-sheet imaging to study myocardial function and intracardiac blood flow during zebrafish development. PLoS Comput Biol 2021; 17:e1009175. [PMID: 34228702 PMCID: PMC8284633 DOI: 10.1371/journal.pcbi.1009175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 07/16/2021] [Accepted: 06/11/2021] [Indexed: 01/07/2023] Open
Abstract
Biomechanical forces intimately contribute to cardiac morphogenesis. However, volumetric imaging to investigate the cardiac mechanics with high temporal and spatial resolution remains an imaging challenge. We hereby integrated light-field microscopy (LFM) with light-sheet fluorescence microscopy (LSFM), coupled with a retrospective gating method, to simultaneously access myocardial contraction and intracardiac blood flow at 200 volumes per second. While LSFM allows for the reconstruction of the myocardial function, LFM enables instantaneous acquisition of the intracardiac blood cells traversing across the valves. We further adopted deformable image registration to quantify the ventricular wall displacement and particle tracking velocimetry to monitor intracardiac blood flow. The integration of LFM and LSFM enabled the time-dependent tracking of the individual blood cells and the differential rates of segmental wall displacement during a cardiac cycle. Taken together, we demonstrated a hybrid system, coupled with our image analysis pipeline, to simultaneously capture the myocardial wall motion with intracardiac blood flow during cardiac development. During the conception of the heart, cardiac muscular contraction and blood flow generate biomechanical forces to influence the functional and structural development. To elucidate the underlying biomechanical mechanisms, we have embraced the zebrafish system for the ease of genetic and pharmacological manipulations and its rapidity for organ development. However, acquiring the dynamic processes (space + time domain) in the small beating zebrafish heart remains a challenge. In the presence of a rapid heartbeat, microscopy is confined by temporal resolution to image the cardiac contraction and blood flow. In this context, we demonstrated an integrated light-sheet and light-field imaging system to visualize cardiac contraction along with the flowing blood cells inside the cardiac chambers. Assuming the periodicity of the cardiac cycle, we synchronized the image data in post-processing for 3-D reconstruction. We further quantified the velocity of the various regions of cardiac muscular contraction, and tracked the individual blood cells during the cardiac cycles. The time-dependent velocity maps allow for uncovering differential segments of cardiac contraction and relaxation, and for revealing the patterns of blood flow. Thus, our integrated light-sheet and light-field imaging system provides an experimental basis to further investigate cardiac function and development.
Collapse
Affiliation(s)
- Zhaoqiang Wang
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Yichen Ding
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
| | - Sandro Satta
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
| | - Mehrdad Roustaei
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Peng Fei
- School of Optical and Electronic Information-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (PF); (TKH)
| | - Tzung K. Hsiai
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
- Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, California, United States of America
- * E-mail: (PF); (TKH)
| |
Collapse
|
18
|
Zopf LM, Heimel P, Geyer SH, Kavirayani A, Reier S, Fröhlich V, Stiglbauer-Tscholakoff A, Chen Z, Nics L, Zinnanti J, Drexler W, Mitterhauser M, Helbich T, Weninger WJ, Slezak P, Obenauf A, Bühler K, Walter A. Cross-Modality Imaging of Murine Tumor Vasculature-a Feasibility Study. Mol Imaging Biol 2021; 23:874-893. [PMID: 34101107 PMCID: PMC8578087 DOI: 10.1007/s11307-021-01615-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 11/29/2022]
Abstract
Tumor vasculature and angiogenesis play a crucial role in tumor progression. Their visualization is therefore of utmost importance to the community. In this proof-of-principle study, we have established a novel cross-modality imaging (CMI) pipeline to characterize exactly the same murine tumors across scales and penetration depths, using orthotopic models of melanoma cancer. This allowed the acquisition of a comprehensive set of vascular parameters for a single tumor. The workflow visualizes capillaries at different length scales, puts them into the context of the overall tumor vessel network and allows quantification and comparison of vessel densities and morphologies by different modalities. The workflow adds information about hypoxia and blood flow rates. The CMI approach includes well-established technologies such as magnetic resonance imaging (MRI), positron emission tomography (PET), computed tomography (CT), and ultrasound (US), and modalities that are recent entrants into preclinical discovery such as optical coherence tomography (OCT) and high-resolution episcopic microscopy (HREM). This novel CMI platform establishes the feasibility of combining these technologies using an extensive image processing pipeline. Despite the challenges pertaining to the integration of microscopic and macroscopic data across spatial resolutions, we also established an open-source pipeline for the semi-automated co-registration of the diverse multiscale datasets, which enables truly correlative vascular imaging. Although focused on tumor vasculature, our CMI platform can be used to tackle a multitude of research questions in cancer biology.
Collapse
Affiliation(s)
- Lydia M Zopf
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | - Patrick Heimel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Austrian BioImaging/CMI, Vienna, Austria.,Core Facility Hard Tissue and Biomaterial Research, Karl Donath Laboratory, University Clinic of Dentistry, Medical University Vienna, Vienna, Austria
| | - Stefan H Geyer
- Division of Anatomy, MIC, Medical University of Vienna, Austrian BioImaging/CMI, Vienna, Austria
| | - Anoop Kavirayani
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | - Susanne Reier
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | - Vanessa Fröhlich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Vienna, Austria
| | - Alexander Stiglbauer-Tscholakoff
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Vienna, Austria
| | - Zhe Chen
- Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Medical University of Vienna, Vienna, Austria
| | - Jelena Zinnanti
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria
| | | | - Markus Mitterhauser
- Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Thomas Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Vienna, Austria
| | - Wolfgang J Weninger
- Division of Anatomy, MIC, Medical University of Vienna, Austrian BioImaging/CMI, Vienna, Austria
| | - Paul Slezak
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Austrian BioImaging/CMI, Vienna, Austria
| | - Anna Obenauf
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Katja Bühler
- VRVis Zentrum für Virtual Reality und Visualisierung Forschungs-GmbH, Austrian BioImaging/CMI, Vienna, Austria
| | - Andreas Walter
- Austrian BioImaging/CMI, Vienna BioCenter Core Facilities GmbH (VBCF), Vienna, Austria.
| |
Collapse
|
19
|
Kolesová H, Olejníčková V, Kvasilová A, Gregorovičová M, Sedmera D. Tissue clearing and imaging methods for cardiovascular development. iScience 2021; 24:102387. [PMID: 33981974 PMCID: PMC8086021 DOI: 10.1016/j.isci.2021.102387] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tissue imaging in 3D using visible light is limited and various clearing techniques were developed to increase imaging depth, but none provides universal solution for all tissues at all developmental stages. In this review, we focus on different tissue clearing methods for 3D imaging of heart and vasculature, based on chemical composition (solvent-based, simple immersion, hyperhydration, and hydrogel embedding techniques). We discuss in detail compatibility of various tissue clearing techniques with visualization methods: fluorescence preservation, immunohistochemistry, nuclear staining, and fluorescent dyes vascular perfusion. We also discuss myocardium visualization using autofluorescence, tissue shrinking, and expansion. Then we overview imaging methods used to study cardiovascular system and live imaging. We discuss heart and vessels segmentation methods and image analysis. The review covers the whole process of cardiovascular system 3D imaging, starting from tissue clearing and its compatibility with various visualization methods to the types of imaging methods and resulting image analysis.
Collapse
Affiliation(s)
- Hana Kolesová
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Science, Prague, Czech Republic
| | - Veronika Olejníčková
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Science, Prague, Czech Republic
| | - Alena Kvasilová
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martina Gregorovičová
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Science, Prague, Czech Republic
| | - David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Science, Prague, Czech Republic
| |
Collapse
|
20
|
Zhu P, Qiu Q, Harris PC, Xu X, Lin X. mtor Haploinsufficiency Ameliorates Renal Cysts and Cilia Abnormality in Adult Zebrafish tmem67 Mutants. J Am Soc Nephrol 2021; 32:822-836. [PMID: 33574160 PMCID: PMC8017545 DOI: 10.1681/asn.2020070991] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/21/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Although zebrafish embryos have been used to study ciliogenesis and model polycystic kidney disease (PKD), adult zebrafish remain unexplored. METHODS Transcription activator-like effector nucleases (TALEN) technology was used to generate mutant for tmem67, the homolog of the mammalian causative gene for Meckel syndrome type 3 (MKS3). Classic 2D and optical-clearing 3D imaging of an isolated adult zebrafish kidney were used to examine cystic and ciliary phenotypes. A hypomorphic mtor strain or rapamycin was used to inhibit mTOR activity. RESULTS Adult tmem67 zebrafish developed progressive mesonephric cysts that share conserved features of mammalian cystogenesis, including a switch of cyst origin with age and an increase in proliferation of cyst-lining epithelial cells. The mutants had shorter and fewer distal single cilia and greater numbers of multiciliated cells (MCCs). Absence of a single cilium preceded cystogenesis, and expansion of MCCs occurred after pronephric cyst formation and was inversely correlated with the severity of renal cysts in young adult zebrafish, suggesting a primary defect and an adaptive action, respectively. Finally, the mutants exhibited hyperactive mTOR signaling. mTOR inhibition ameliorated renal cysts in both the embryonic and adult zebrafish models; however, it only rescued ciliary abnormalities in the adult mutants. CONCLUSIONS Adult zebrafish tmem67 mutants offer a new vertebrate model for renal cystic diseases, in which cilia morphology can be analyzed at a single-nephron resolution and mTOR inhibition proves to be a candidate therapeutic strategy.
Collapse
Affiliation(s)
- Ping Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Qi Qiu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Peter C. Harris
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota,Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
21
|
Roostalu U, Thisted L, Skytte JL, Salinas CG, Pedersen PJ, Hecksher-Sørensen J, Rolin B, Hansen HH, MacKrell JG, Christie RM, Vrang N, Jelsing J, Zois NE. Effect of captopril on post-infarction remodelling visualized by light sheet microscopy and echocardiography. Sci Rep 2021; 11:5241. [PMID: 33664407 PMCID: PMC7933438 DOI: 10.1038/s41598-021-84812-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/22/2021] [Indexed: 02/08/2023] Open
Abstract
Angiotensin converting enzyme inhibitors, among them captopril, improve survival following myocardial infarction (MI). The mechanisms of captopril action remain inadequately understood due to its diverse effects on multiple signalling pathways at different time periods following MI. Here we aimed to establish the role of captopril in late-stage post-MI remodelling. Left anterior descending artery (LAD) ligation or sham surgery was carried out in male C57BL/6J mice. Seven days post-surgery LAD ligated mice were allocated to daily vehicle or captopril treatment continued over four weeks. To provide comprehensive characterization of the changes in mouse heart following MI a 3D light sheet imaging method was established together with automated image analysis workflow. The combination of echocardiography and light sheet imaging enabled to assess cardiac function and the underlying morphological changes. We show that delayed captopril treatment does not affect infarct size but prevents left ventricle dilation and hypertrophy, resulting in improved ejection fraction. Quantification of lectin perfused blood vessels showed improved vascular density in the infarct border zone in captopril treated mice in comparison to vehicle dosed control mice. These results validate the applicability of combined echocardiographic and light sheet assessment of drug mode of action in preclinical cardiovascular research.
Collapse
Affiliation(s)
- Urmas Roostalu
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark.
| | | | | | | | | | | | - Bidda Rolin
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
- Novo Nordisk, 2760, Maaloev, Denmark
| | | | - James G MacKrell
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Robert M Christie
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Niels Vrang
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
| | - Jacob Jelsing
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
| | | |
Collapse
|
22
|
Wang W, Zhang Y, Hui H, Tong W, Wei Z, Li Z, Zhang S, Yang X, Tian J, Chen Y. The effect of endothelial progenitor cell transplantation on neointimal hyperplasia and reendothelialisation after balloon catheter injury in rat carotid arteries. Stem Cell Res Ther 2021; 12:99. [PMID: 33536065 PMCID: PMC7860581 DOI: 10.1186/s13287-021-02135-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/01/2021] [Indexed: 12/20/2022] Open
Abstract
Background Reendothelialisation is the natural pathway that inhibits neointimal hyperplasia and in-stent restenosis. Circulating endothelial progenitor cells (EPCs) derived from bone marrow (BM) might contribute to endothelial repair. However, the temporal and spatial distributions of reendothelialisation and neointimal hyperplasia after EPC transplantation in injured arteries are currently unclear. Methods A carotid balloon injury (BI) model was established in Sprague-Dawley rats, and PKH26-labelled BM-derived EPCs were transplanted after BI. The carotid arteries were harvested on the first, fourth, seventh, and 14th day post-injury and analysed via light-sheet fluorescence microscopy and pathological staining (n = 3). EPC and human umbilical vein endothelial cell culture supernatants were collected, and blood samples were collected before and after transplantation. The paracrine effects of VEGF, IGF-1, and TGF-β1 in cell culture supernatants and serum were analysed by enzyme-linked immunosorbent assay (n = 4). Results Transplanted EPCs labelled with PKH26 were attached to the injured luminal surface the first day after BI. In the sham operation group, the transplanted EPCs did not adhere to the luminal surface. From the fourth day after BI, the mean fluorescence intensity of PKH26 decreased significantly. However, reendothelialisation and inhibition of neointimal hyperplasia were significantly promoted by transplanted EPCs. The degree of reendothelialisation of the EPC7d and EPC14d groups was higher than that of the BI7d and BI14d groups, and the difference in neointimal hyperplasia was observed between the EPC14d and BI14d groups. The number of endothelial cells on the luminal surface of the EPC14d group was higher than that of the BI14d group. The number of infiltrated macrophages in the injured artery decreased in the EPC transplanted groups. Conclusions Transplanted EPCs had chemotactic enrichment and attached to the injured arterial luminal surface. Although decreasing significantly after the fourth day at the site of injury after transplantation, transplanted EPCs could still promote reendothelialisation and inhibit neointimal hyperplasia. The underlying mechanism is through paracrine cytokines and not differentiation into mature endothelial cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02135-w.
Collapse
Affiliation(s)
- Wei Wang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yingqian Zhang
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Tong
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zechen Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhongxuan Li
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Suhui Zhang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xin Yang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100083, China.
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
23
|
Chang CC, Chu A, Meyer S, Ding Y, Sun MM, Abiri P, Baek KI, Gudapati V, Ding X, Guihard P, Bostrom KI, Li S, Gordon LK, Zheng JJ, Hsiai TK. Three-dimensional Imaging Coupled with Topological Quantification Uncovers Retinal Vascular Plexuses Undergoing Obliteration. Theranostics 2021; 11:1162-1175. [PMID: 33391527 PMCID: PMC7738897 DOI: 10.7150/thno.53073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction: Murine models provide microvascular insights into the 3-D network disarray seen in retinopathy and cardiovascular diseases. Light-sheet fluorescence microscopy (LSFM) has emerged to capture retinal vasculature in 3-D, allowing for assessment of the progression of retinopathy and the potential to screen new therapeutic targets in mice. We hereby coupled LSFM, also known as selective plane illumination microscopy, with topological quantification, to characterize the retinal vascular plexuses undergoing preferential obliteration. Method and Result: In postnatal mice, we revealed the 3-D retinal microvascular network in which the vertical sprouts bridge the primary (inner) and secondary (outer) plexuses, whereas, in an oxygen-induced retinopathy (OIR) mouse model, we demonstrated preferential obliteration of the secondary plexus and bridging vessels with a relatively unscathed primary plexus. Using clustering coefficients and Euler numbers, we computed the local versus global vascular connectivity. While local connectivity was preserved (p > 0.05, n = 5 vs. normoxia), the global vascular connectivity in hyperoxia-exposed retinas was significantly reduced (p < 0.05, n = 5 vs. normoxia). Applying principal component analysis (PCA) for auto-segmentation of the vertical sprouts, we corroborated the obliteration of the vertical sprouts bridging the secondary plexuses, as evidenced by impaired vascular branching and connectivity, and reduction in vessel volumes and lengths (p < 0.05, n = 5 vs. normoxia). Conclusion: Coupling 3-D LSFM with topological quantification uncovered the retinal vasculature undergoing hyperoxia-induced obliteration from the secondary (outer) plexus to the vertical sprouts. The use of clustering coefficients, Euler's number, and PCA provided new network insights into OIR-associated vascular obliteration, with translational significance for investigating therapeutic interventions to prevent visual impairment.
Collapse
Affiliation(s)
- Chih-Chiang Chang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA
| | - Alison Chu
- Division of Neonatology and Developmental Biology, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Scott Meyer
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Yichen Ding
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Michel M. Sun
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Parinaz Abiri
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kyung In Baek
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Varun Gudapati
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Xili Ding
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA
| | - Pierre Guihard
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kristina I. Bostrom
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Greater Los Angeles VA Healthcare System, Los Angeles, CA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA
| | - Lynn K. Gordon
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jie J. Zheng
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Tzung K. Hsiai
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Greater Los Angeles VA Healthcare System, Los Angeles, CA
- Medical Engineering, California Institute of Technology, Pasadena, CA
| |
Collapse
|
24
|
Ding Y, Gudapati V, Lin R, Fei Y, Sevag Packard RR, Song S, Chang CC, Baek KI, Wang Z, Roustaei M, Kuang D, Jay Kuo CC, Hsiai TK. Saak Transform-Based Machine Learning for Light-Sheet Imaging of Cardiac Trabeculation. IEEE Trans Biomed Eng 2021; 68:225-235. [PMID: 32365015 PMCID: PMC7606319 DOI: 10.1109/tbme.2020.2991754] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Recent advances in light-sheet fluorescence microscopy (LSFM) enable 3-dimensional (3-D) imaging of cardiac architecture and mechanics in toto. However, segmentation of the cardiac trabecular network to quantify cardiac injury remains a challenge. METHODS We hereby employed "subspace approximation with augmented kernels (Saak) transform" for accurate and efficient quantification of the light-sheet image stacks following chemotherapy-treatment. We established a machine learning framework with augmented kernels based on the Karhunen-Loeve Transform (KLT) to preserve linearity and reversibility of rectification. RESULTS The Saak transform-based machine learning enhances computational efficiency and obviates iterative optimization of cost function needed for neural networks, minimizing the number of training datasets for segmentation in our scenario. The integration of forward and inverse Saak transforms can also serve as a light-weight module to filter adversarial perturbations and reconstruct estimated images, salvaging robustness of existing classification methods. The accuracy and robustness of the Saak transform are evident following the tests of dice similarity coefficients and various adversary perturbation algorithms, respectively. The addition of edge detection further allows for quantifying the surface area to volume ratio (SVR) of the myocardium in response to chemotherapy-induced cardiac remodeling. CONCLUSION The combination of Saak transform, random forest, and edge detection augments segmentation efficiency by 20-fold as compared to manual processing. SIGNIFICANCE This new methodology establishes a robust framework for post light-sheet imaging processing, and creating a data-driven machine learning for automated quantification of cardiac ultra-structure.
Collapse
Affiliation(s)
- Yichen Ding
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Varun Gudapati
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Ruiyuan Lin
- Ming-Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089 USA
| | - Yanan Fei
- Ming-Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089 USA
| | - René R Sevag Packard
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Sibo Song
- Ming-Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089 USA
| | - Chih-Chiang Chang
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Kyung In Baek
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Zhaoqiang Wang
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Mehrdad Roustaei
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Dengfeng Kuang
- Tianjin Key Laboratory of Optoelectronic Sensor and Sensing Network Technology, and Institute of Modern Optics, Nankai University, Tianjin 300350, China
| | - C.-C. Jay Kuo
- Ming-Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089 USA
| | - Tzung K. Hsiai
- Henry Samueli School of Engineering and David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
25
|
Jurtz VI, Skovbjerg G, Salinas CG, Roostalu U, Pedersen L, Hecksher-Sørensen J, Rolin B, Nyberg M, van de Bunt M, Ingvorsen C. Deep learning reveals 3D atherosclerotic plaque distribution and composition. Sci Rep 2020; 10:21523. [PMID: 33299076 PMCID: PMC7726562 DOI: 10.1038/s41598-020-78632-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Complications of atherosclerosis are the leading cause of morbidity and mortality worldwide. Various genetically modified mouse models are used to investigate disease trajectory with classical histology, currently the preferred methodology to elucidate plaque composition. Here, we show the strength of light-sheet fluorescence microscopy combined with deep learning image analysis for characterising and quantifying plaque burden and composition in whole aorta specimens. 3D imaging is a non-destructive method that requires minimal ex vivo handling and can be up-scaled to large sample sizes. Combined with deep learning, atherosclerotic plaque in mice can be identified without any ex vivo staining due to the autofluorescent nature of the tissue. The aorta and its branches can subsequently be segmented to determine how anatomical position affects plaque composition and progression. Here, we find the highest plaque accumulation in the aortic arch and brachiocephalic artery. Simultaneously, aortas can be stained for markers of interest (for example the pan immune cell marker CD45) and quantified. In ApoE-/- mice we observe that levels of CD45 reach a plateau after which increases in plaque volume no longer correlate to immune cell infiltration. All underlying code is made publicly available to ease adaption of the method.
Collapse
MESH Headings
- Animals
- Aorta/pathology
- Aortic Diseases
- Apolipoproteins E/analysis
- Atherosclerosis/complications
- Atherosclerosis/pathology
- Deep Learning
- Disease Models, Animal
- Female
- Image Processing, Computer-Assisted/methods
- Imaging, Three-Dimensional/methods
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Fluorescence/methods
- Plaque, Atherosclerotic/diagnostic imaging
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Receptors, LDL/analysis
Collapse
Affiliation(s)
| | - Grethe Skovbjerg
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- Gubra, 2970, Hoersholm, Denmark
| | | | | | - Louise Pedersen
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- University of Copenhagen, 1017, Copenhagen, Denmark
| | | | - Bidda Rolin
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- Gubra, 2970, Hoersholm, Denmark
| | - Michael Nyberg
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
| | | | | |
Collapse
|
26
|
Dejea H, Bonnin A, Cook AC, Garcia-Canadilla P. Cardiac multi-scale investigation of the right and left ventricle ex vivo: a review. Cardiovasc Diagn Ther 2020; 10:1701-1717. [PMID: 33224784 DOI: 10.21037/cdt-20-269] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The heart is a complex multi-scale system composed of components integrated at the subcellular, cellular, tissue and organ levels. The myocytes, the contractile elements of the heart, form a complex three-dimensional (3D) network which enables propagation of the electrical signal that triggers the contraction to efficiently pump blood towards the whole body. Cardiovascular diseases (CVDs), a major cause of mortality in developed countries, often lead to cardiovascular remodeling affecting cardiac structure and function at all scales, from myocytes and their surrounding collagen matrix to the 3D organization of the whole heart. As yet, there is no consensus as to how the myocytes are arranged and packed within their connective tissue matrix, nor how best to image them at multiple scales. Cardiovascular imaging is routinely used to investigate cardiac structure and function as well as for the evaluation of cardiac remodeling in CVDs. For a complete understanding of the relationship between structural remodeling and cardiac dysfunction in CVDs, multi-scale imaging approaches are necessary to achieve a detailed description of ventricular architecture along with cardiac function. In this context, ventricular architecture has been extensively studied using a wide variety of imaging techniques: ultrasound (US), optical coherence tomography (OCT), microscopy (confocal, episcopic, light sheet, polarized light), magnetic resonance imaging (MRI), micro-computed tomography (micro-CT) and, more recently, synchrotron X-ray phase contrast imaging (SR X-PCI). Each of these techniques have their own set of strengths and weaknesses, relating to sample size, preparation, resolution, 2D/3D capabilities, use of contrast agents and possibility of performing together with in vivo studies. Therefore, the combination of different imaging techniques to investigate the same sample, thus taking advantage of the strengths of each method, could help us to extract the maximum information about ventricular architecture and function. In this review, we provide an overview of available and emerging cardiovascular imaging techniques for assessing myocardial architecture ex vivo and discuss their utility in being able to quantify cardiac remodeling, in CVDs, from myocyte to whole organ.
Collapse
Affiliation(s)
- Hector Dejea
- Paul Scherrer Institut, Villigen PSI, Villigen, Switzerland.,Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Anne Bonnin
- Paul Scherrer Institut, Villigen PSI, Villigen, Switzerland
| | - Andrew C Cook
- Institute of Cardiovascular Science, University College London, London, UK
| | - Patricia Garcia-Canadilla
- Institute of Cardiovascular Science, University College London, London, UK.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
27
|
Hassan S, Barrett CJ, Crossman DJ. Imaging tools for assessment of myocardial fibrosis in humans: the need for greater detail. Biophys Rev 2020; 12:969-987. [PMID: 32705483 PMCID: PMC7429810 DOI: 10.1007/s12551-020-00738-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
Myocardial fibrosis is recognized as a key pathological process in the development of cardiac disease and a target for future therapeutics. Despite this recognition, the assessment of fibrosis is not a part of routine clinical practice. This is primarily due to the difficulties in obtaining an accurate assessment of fibrosis non-invasively. Moreover, there is a clear discrepancy between the understandings of myocardial fibrosis clinically where fibrosis is predominately studied with comparatively low-resolution medical imaging technologies like MRI compared with the basic science laboratories where fibrosis can be visualized invasively with high resolution using molecularly specific fluorescence microscopes at the microscopic and nanoscopic scales. In this article, we will first review current medical imaging technologies for assessing fibrosis including echo and MRI. We will then highlight the need for greater microscopic and nanoscopic analysis of human tissue and how this can be addressed through greater utilization of human tissue available through endomyocardial biopsies and cardiac surgeries. We will then describe the relatively new field of molecular imaging that promises to translate research findings to the clinical practice by non-invasively monitoring the molecular signature of fibrosis in patients.
Collapse
Affiliation(s)
- Summer Hassan
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Auckland City Hospital, Auckland District Health Board, Auckland, New Zealand
| | - Carolyn J Barrett
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - David J Crossman
- Department of Physiology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
28
|
Yang Y, Li G, Chen L. High resolution three-dimensional imaging of the ocular surface and intact eyeball using tissue clearing and light sheet microscopy. Ocul Surf 2020; 18:526-532. [PMID: 32417103 DOI: 10.1016/j.jtos.2020.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 01/23/2023]
Abstract
PURPOSE High resolution visualization of the ocular surface and intact eyeball is critical and essential for our understanding and treatment of eye diseases. This study is to achieve this goal using advanced tissue clearing and three-dimensional (3D) imaging technologies. METHODS Wild type and fluorescently labeled transgenic mice of Prox-1-GFP (green fluorescent protein) or Thy1-YFP (yellow fluorescent protein) were used in the study. Eyeballs were harvested from normal or a disease model of corneal inflammation. Samples were infused with hydrogel monomers and heated for polymerization. Lipids were removed by electrophoresis. The transparent tissue-hydrogel hybrids of the anterior segments or intact eyeballs with immunolabeling or endogenous fluorescence were imaged by an advanced light sheet fluorescent microscope. High resolution 3D images and videos were captured for a wide array of structures and cell types. RESULTS Optical transparency was achieved from intact eyeballs of both normal and diseased conditions. A variety of important structures and cell types, such as blood and lymphatic vessels, Schlemm's canal, nerves and endothelial cells, were detected with their natural morphology, location and organizational network. CONCLUSIONS This study provides the first comprehensive and 3D high resolution imaging of the intact eyeball using tissue clearing and advanced light sheet microscopy. Given that the eye is the window of the body, we anticipate this advanced technology will facilitate diverse applications in biomedical research inside and outside the eye.
Collapse
Affiliation(s)
- Yujia Yang
- Vision Science Graduate Program, Center for Eye Disease and Development, and School of Optometry, University of California, Berkeley, CA, 94720, USA; Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Guangyu Li
- Vision Science Graduate Program, Center for Eye Disease and Development, and School of Optometry, University of California, Berkeley, CA, 94720, USA
| | - Lu Chen
- Vision Science Graduate Program, Center for Eye Disease and Development, and School of Optometry, University of California, Berkeley, CA, 94720, USA; The Proctor Foundation for Research in Ophthalmology, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
29
|
Feng J, Cavallero S, Hsiai T, Li R. Impact of air pollution on intestinal redox lipidome and microbiome. Free Radic Biol Med 2020; 151:99-110. [PMID: 31904545 DOI: 10.1016/j.freeradbiomed.2019.12.044] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/29/2019] [Accepted: 12/30/2019] [Indexed: 12/20/2022]
Abstract
Air pollution is a rising public health issue worldwide. Cumulative epidemiological and experimental studies have shown that exposure to air pollution such as particulate matter (PM) is linked with increased hospital admissions and all-cause mortality. While previous studies on air pollution mostly focused on the respiratory and cardiovascular effects, emerging evidence supports a significant impact of air pollution on the gastrointestinal (GI) system. The gut is exposed to PM as most of the inhaled particles are removed from the lungs to the GI tract via mucociliary clearance. Ingestion of contaminated food and water is another common source of GI tract exposure to pollutants. Recent studies have associated air pollution with intestinal diseases, including appendicitis, colorectal cancer, and inflammatory bowel disease. In addition to the liver and adipose tissue, intestine is an important organ system for lipid metabolism, and the intestinal redox lipids might be tightly associated with the intestinal and systematic inflammation. The gut microbiota modulates lipid metabolism and contributes to the initiation and development of intestinal disease including inflammatory bowel disease. Recent data support microbiome implication in air pollution-mediated intestinal and systematic effects. In this review, the associations between air pollution and intestinal diseases, and the alterations of intestinal lipidome and gut microbiome by air pollution are highlighted. The potential mechanistic aspects underlying air pollution-mediated intestinal pathology will also be discussed.
Collapse
Affiliation(s)
- Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, China
| | - Susana Cavallero
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Tzung Hsiai
- Department of Medicine, University of California, Los Angeles, CA, USA; Department of Bioengineering, University of California, Los Angeles, CA, USA; West Los Angeles Healthcare System, USA; Medical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rongsong Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, China.
| |
Collapse
|
30
|
Becher T, Riascos-Bernal DF, Kramer DJ, Almonte VM, Chi J, Tong T, Oliveira-Paula GH, Koleilat I, Chen W, Cohen P, Sibinga NES. Three-Dimensional Imaging Provides Detailed Atherosclerotic Plaque Morphology and Reveals Angiogenesis After Carotid Artery Ligation. Circ Res 2020; 126:619-632. [PMID: 31914850 DOI: 10.1161/circresaha.119.315804] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE Remodeling of the vessel wall and the formation of vascular networks are dynamic processes that occur during mammalian embryonic development and in adulthood. Plaque development and excessive neointima formation are hallmarks of atherosclerosis and vascular injury. As our understanding of these complex processes evolves, there is a need to develop new imaging techniques to study underlying mechanisms. OBJECTIVE We used tissue clearing and light-sheet microscopy for 3-dimensional (3D) profiling of the vascular response to carotid artery ligation and induction of atherosclerosis in mouse models. METHODS AND RESULTS Adipo-Clear and immunolabeling in combination with light-sheet microscopy were applied to image carotid arteries and brachiocephalic arteries, allowing for 3D reconstruction of vessel architecture. Entire 3D neointima formations with different geometries were observed within the carotid artery and scored by volumetric analysis. Additionally, we identified a CD31-positive adventitial plexus after ligation of the carotid artery that evolved and matured over time. We also used this method to characterize plaque extent and composition in the brachiocephalic arteries of ApoE-deficient mice on high-fat diet. The plaques exhibited inter-animal differences in terms of plaque volume, geometry, and ratio of acellular core to plaque volume. A 3D reconstruction of the endothelium overlying the plaque was also generated. CONCLUSIONS We present a novel approach to characterize vascular remodeling in adult mice using Adipo-Clear in combination with light-sheet microscopy. Our method reconstructs 3D neointima formation after arterial injury and allows for volumetric analysis of remodeling, in addition to revealing angiogenesis and maturation of a plexus surrounding the carotid artery. This method generates complete 3D reconstructions of atherosclerotic plaques and uncovers their volume, geometry, acellular component, surface, and spatial position within the brachiocephalic arteries. Our approach may be used in a number of mouse models of cardiovascular disease to assess vessel geometry and volume. Visual Overview: An online visual overview is available for this article.
Collapse
Affiliation(s)
- Tobias Becher
- From the Laboratory of Molecular Metabolism (T.B., D.J.K., J.C., P.C.), The Rockefeller University, NY.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (T.B.).,First Department of Medicine (Division of Cardiology), University Medical Center Mannheim, Germany (T.B.)
| | - Dario F Riascos-Bernal
- (Cardiology Division) Department of Medicine, Department of Developmental and Molecular Biology, Wilf Family Cardiovascular Research Institute (D.F.R.-B., V.M.A., G.H.O.-P., N.E.S.S.), Albert Einstein College of Medicine, Bronx, NY
| | - Daniel J Kramer
- From the Laboratory of Molecular Metabolism (T.B., D.J.K., J.C., P.C.), The Rockefeller University, NY
| | - Vanessa M Almonte
- (Cardiology Division) Department of Medicine, Department of Developmental and Molecular Biology, Wilf Family Cardiovascular Research Institute (D.F.R.-B., V.M.A., G.H.O.-P., N.E.S.S.), Albert Einstein College of Medicine, Bronx, NY
| | - Jingy Chi
- From the Laboratory of Molecular Metabolism (T.B., D.J.K., J.C., P.C.), The Rockefeller University, NY
| | - Tao Tong
- Bio-Imaging Resource Center (T.T.), The Rockefeller University, NY
| | - Gustavo H Oliveira-Paula
- (Cardiology Division) Department of Medicine, Department of Developmental and Molecular Biology, Wilf Family Cardiovascular Research Institute (D.F.R.-B., V.M.A., G.H.O.-P., N.E.S.S.), Albert Einstein College of Medicine, Bronx, NY
| | - Issam Koleilat
- Department of Cardiothoracic and Vascular Surgery (Division of Vascular Surgery), Montefiore Medical Center, Bronx, NY (I.K.)
| | - Wei Chen
- Department of Medicine (Nephrology Division) (W.C.), Albert Einstein College of Medicine, Bronx, NY.,Department of Medicine, University of Rochester School of Medicine and Dentistry, NY (W.C.)
| | - Paul Cohen
- From the Laboratory of Molecular Metabolism (T.B., D.J.K., J.C., P.C.), The Rockefeller University, NY
| | - Nicholas E S Sibinga
- (Cardiology Division) Department of Medicine, Department of Developmental and Molecular Biology, Wilf Family Cardiovascular Research Institute (D.F.R.-B., V.M.A., G.H.O.-P., N.E.S.S.), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
31
|
Chen J, Ding Y, Chen M, Gau J, Jen N, Nahal C, Tu S, Chen C, Zhou S, Chang CC, Lyu J, Xu X, Hsiai TK, Packard RRS. Displacement analysis of myocardial mechanical deformation (DIAMOND) reveals segmental susceptibility to doxorubicin-induced injury and regeneration. JCI Insight 2019; 4:125362. [PMID: 30996130 PMCID: PMC6538350 DOI: 10.1172/jci.insight.125362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/27/2019] [Indexed: 12/27/2022] Open
Abstract
Zebrafish are increasingly utilized to model cardiomyopathies and regeneration. Current methods evaluating cardiac function have known limitations, fail to reliably detect focal mechanics, and are not readily feasible in zebrafish. We developed a semiautomated, open-source method - displacement analysis of myocardial mechanical deformation (DIAMOND) - for quantitative assessment of 4D segmental cardiac function. We imaged transgenic embryonic zebrafish in vivo using a light-sheet fluorescence microscopy system with 4D cardiac motion synchronization. Our method permits the derivation of a transformation matrix to quantify the time-dependent 3D displacement of segmental myocardial mass centroids. Through treatment with doxorubicin, and by chemically and genetically manipulating the myocardial injury-activated Notch signaling pathway, we used DIAMOND to demonstrate that basal ventricular segments adjacent to the atrioventricular canal display the highest 3D displacement and are also the most susceptible to doxorubicin-induced injury. Thus, DIAMOND provides biomechanical insights into in vivo segmental cardiac function scalable to high-throughput research applications.
Collapse
Affiliation(s)
- Junjie Chen
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Yichen Ding
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Michael Chen
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Jonathan Gau
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Nelson Jen
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Chadi Nahal
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Sally Tu
- Department of Neuroscience, College of Letters and Science, University of California, Los Angeles, California, USA
| | - Cynthia Chen
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Steve Zhou
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Chih-Chiang Chang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Jintian Lyu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Tzung K. Hsiai
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | | |
Collapse
|