1
|
Moraiti S, Cheong VS, Dall’Ara E, Kadirkamanathan V, Bhattacharya P. A novel framework for elucidating the effect of mechanical loading on the geometry of ovariectomized mouse tibiae using principal component analysis. Front Bioeng Biotechnol 2024; 12:1469272. [PMID: 39502499 PMCID: PMC11534826 DOI: 10.3389/fbioe.2024.1469272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Murine models are used to test the effect of anti-osteoporosis treatments as they replicate some of the bone phenotypes observed in osteoporotic (OP) patients. The effect of disease and treatment is typically described as changes in bone geometry and microstructure over time. Conventional assessment of geometric changes relies on morphometric scalar parameters. However, being correlated with each other, these parameters do not describe separate fractions of variations and offer only a moderate insight into temporal changes. Methods The current study proposes a novel image-based framework that employs deformable image registration on in vivo longitudinal images of bones and Principal Component Analysis (PCA) for improved quantification of geometric effects of OP treatments. This PCA-based model and a novel post-processing of score changes provide orthogonal modes of shape variations temporally induced by a course of treatment (specifically in vivo mechanical loading). Results and Discussion Errors associated with the proposed framework are rigorously quantified and it is shown that the accuracy of deformable image registration in capturing the bone shapes (∼1 voxel = 10.4 μm) is of the same order of magnitude as the relevant state-of-the-art evaluation studies. Applying the framework to longitudinal image data from the midshaft section of ovariectomized mouse tibia, two mutually orthogonal mode shapes are reliably identified to be an effect of treatment. The mode shapes captured changes of the tibia geometry due to the treatment at the anterior crest (maximum of 0.103 mm) and across the tibia midshaft section and the posterior (0.030 mm) and medial (0.024 mm) aspects. These changes agree with those reported previously but are now described in a compact fashion, as a vector field of displacements on the bone surface. The proposed framework enables a more detailed investigation of the effect of disease and treatment on bones in preclinical studies and boosts the precision of such assessments.
Collapse
Affiliation(s)
- Stamatina Moraiti
- Department of Mechanical Engineering, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Vee San Cheong
- Department of Mechanical Engineering, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Future Health Technologies Programme, Singapore-ETH Centre, Create campus, Singapore, Singapore
| | - Enrico Dall’Ara
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Visakan Kadirkamanathan
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Pinaki Bhattacharya
- Department of Mechanical Engineering, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
2
|
Zwijnen AW, Watzema L, Ridwan Y, van Der Pluijm I, Smal I, Essers J. Self-adaptive deep learning-based segmentation for universal and functional clinical and preclinical CT image analysis. Comput Biol Med 2024; 179:108853. [PMID: 39013341 DOI: 10.1016/j.compbiomed.2024.108853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Methods to monitor cardiac functioning non-invasively can accelerate preclinical and clinical research into novel treatment options for heart failure. However, manual image analysis of cardiac substructures is resource-intensive and error-prone. While automated methods exist for clinical CT images, translating these to preclinical μCT data is challenging. We employed deep learning to automate the extraction of quantitative data from both CT and μCT images. METHODS We collected a public dataset of cardiac CT images of human patients, as well as acquired μCT images of wild-type and accelerated aging mice. The left ventricle, myocardium, and right ventricle were manually segmented in the μCT training set. After template-based heart detection, two separate segmentation neural networks were trained using the nnU-Net framework. RESULTS The mean Dice score of the CT segmentation results (0.925 ± 0.019, n = 40) was superior to those achieved by state-of-the-art algorithms. Automated and manual segmentations of the μCT training set were nearly identical. The estimated median Dice score (0.940) of the test set results was comparable to existing methods. The automated volume metrics were similar to manual expert observations. In aging mice, ejection fractions had significantly decreased, and myocardial volume increased by age 24 weeks. CONCLUSIONS With further optimization, automated data extraction expands the application of (μ)CT imaging, while reducing subjectivity and workload. The proposed method efficiently measures the left and right ventricular ejection fraction and myocardial mass. With uniform translation between image types, cardiac functioning in diastolic and systolic phases can be monitored in both animals and humans.
Collapse
Affiliation(s)
- Anne-Wietje Zwijnen
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Yanto Ridwan
- AMIE Core Facility, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ingrid van Der Pluijm
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ihor Smal
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Roberts BC, Cheong VS, Oliviero S, Arredondo Carrera HM, Wang N, Gartland A, Dall'Ara E. Combining PTH(1-34) and mechanical loading has increased benefit to tibia bone mechanics in ovariectomised mice. J Orthop Res 2024; 42:1254-1266. [PMID: 38151816 DOI: 10.1002/jor.25777] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 11/29/2023] [Accepted: 12/24/2023] [Indexed: 12/29/2023]
Abstract
Combined treatment with PTH(1-34) and mechanical loading confers increased structural benefits to bone than monotherapies. However, it remains unclear how this longitudinal adaptation affects the bone mechanics. This study quantified the individual and combined longitudinal effects of PTH(1-34) and mechanical loading on the bone stiffness and strength evaluated in vivo with validated micro-finite element (microFE) models. C57BL/6 mice were ovariectomised at 14-week-old and treated either with injections of PTH(1-34), compressive tibia loading or both interventions concurrently. Right tibiae were in vivo microCT-scanned every 2 weeks from 14 until 24-week-old. MicroCT images were rigidly registered to reference tibia and the cortical organ level (whole bone) and tissue level (midshaft) morphometric properties and bone mineral content were quantified. MicroCT images were converted into voxel-based homogeneous, linear elastic microFE models to estimate the bone stiffness and strength. This approach allowed us for the first time to quantify the longitudinal changes in mechanical properties induced by combined treatments in a model of accelerated bone resorption. Both changes of stiffness and strength were higher with co-treatment than with individual therapies, consistent with increased benefits with the tibia bone mineral content and cortical area, properties strongly associated with the tibia mechanics. The longitudinal data shows that the two bone anabolics, both individually and combined, had persistent benefit on estimated mechanical properties, and that benefits (increased stiffness and strength) remained after treatment was withdrawn.
Collapse
Affiliation(s)
- Bryant C Roberts
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
- Adelaide Microscopy, Division of Research and Innovation, The University of Adelaide, Adelaide, South Australia, Australia
| | - Vee San Cheong
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, UK
| | - Sara Oliviero
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | | | - Ning Wang
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK
| | - Alison Gartland
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK
| | - Enrico Dall'Ara
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
4
|
Fernandez ME, Martinez-Romero J, Aon MA, Bernier M, Price NL, de Cabo R. How is Big Data reshaping preclinical aging research? Lab Anim (NY) 2023; 52:289-314. [PMID: 38017182 DOI: 10.1038/s41684-023-01286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/10/2023] [Indexed: 11/30/2023]
Abstract
The exponential scientific and technological progress during the past 30 years has favored the comprehensive characterization of aging processes with their multivariate nature, leading to the advent of Big Data in preclinical aging research. Spanning from molecular omics to organism-level deep phenotyping, Big Data demands large computational resources for storage and analysis, as well as new analytical tools and conceptual frameworks to gain novel insights leading to discovery. Systems biology has emerged as a paradigm that utilizes Big Data to gain insightful information enabling a better understanding of living organisms, visualized as multilayered networks of interacting molecules, cells, tissues and organs at different spatiotemporal scales. In this framework, where aging, health and disease represent emergent states from an evolving dynamic complex system, context given by, for example, strain, sex and feeding times, becomes paramount for defining the biological trajectory of an organism. Using bioinformatics and artificial intelligence, the systems biology approach is leading to remarkable advances in our understanding of the underlying mechanism of aging biology and assisting in creative experimental study designs in animal models. Future in-depth knowledge acquisition will depend on the ability to fully integrate information from different spatiotemporal scales in organisms, which will probably require the adoption of theories and methods from the field of complex systems. Here we review state-of-the-art approaches in preclinical research, with a focus on rodent models, that are leading to conceptual and/or technical advances in leveraging Big Data to understand basic aging biology and its full translational potential.
Collapse
Affiliation(s)
- Maria Emilia Fernandez
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jorge Martinez-Romero
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Miguel A Aon
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
5
|
Tourkova IL, Larrouture QC, Onwuka KM, Liu S, Luo J, Schlesinger PH, Blair HC. Age-related decline in bone mineral transport and bone matrix proteins in osteoblasts from stromal stem cells. Am J Physiol Cell Physiol 2023; 325:C613-C622. [PMID: 37519232 PMCID: PMC10635645 DOI: 10.1152/ajpcell.00227.2023] [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: 05/23/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023]
Abstract
We studied osteoblast bone mineral transport and matrix proteins as a function of age. In isolated bone marrow cells from long bones of young (3 or 4 mo) and old (18 or 19 mo) mice, age correlated with reduced mRNA of mineral transport proteins: alkaline phosphatase (ALP), ankylosis (ANK), the Cl-/H+ exchanger ClC3, and matrix proteins collagen 1 (Col1) and osteocalcin (BGLAP). Some proteins, including the neutral phosphate transporter2 (NPT2), were not reduced. These are predominately osteoblast proteins, but in mixed cell populations. Remarkably, in osteoblasts differentiated from preparations of stromal stem cells (SSCs) made from bone marrow cells in young and old mice, differentiated in vitro on perforated polyethylene terephthalate membranes, mRNA confirmed decreased expression with age for most transport-related and bone matrix proteins. Additional mRNAs in osteoblasts in vitro included ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), unchanged, and ENPP2, reduced with age. Decrease with age in ALP activity and protein by Western blot was also significant. Transport protein findings correlated with micro-computed tomography of lumbar vertebra, showing that trabecular bone of old mice is osteopenic relative to young mice, consistent with other studies. Pathway analysis of osteoblasts differentiated in vitro showed that cells from old animals had reduced Erk1/2 phosphorylation and decreased suppressor of mothers against decapentaplegic 2 (Smad2) mRNA, consistent with TGFβ pathway, and reduced β-catenin mRNA, consistent with WNT pathway regulation. Our results show that decline in bone density with age reflects selective changes, resulting effectively in a phenotype modification. Reduction of matrix and mineral transport protein expression with age is regulated by multiple signaling pathways.NEW & NOTEWORTHY This work for the first time showed that specific enzymes in bone mineral transport, and matrix synthesis proteins, in the epithelial-like bone-forming cell layer are downregulated with aging. Results were compared using cells extracted from long bones of young and old mice, or in essentially uniform osteoblasts differentiated from stromal stem cells in vitro. The age effect showed memory in the stromal stem cells, a remarkable finding.
Collapse
Affiliation(s)
- Irina L Tourkova
- Research Service, VA Medical Center, Pittsburgh, Pennsylvania, United States
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Quitterie C Larrouture
- Research Service, VA Medical Center, Pittsburgh, Pennsylvania, United States
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Kelechi M Onwuka
- Research Service, VA Medical Center, Pittsburgh, Pennsylvania, United States
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Silvia Liu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jianhua Luo
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Paul H Schlesinger
- Department of Cell Biology & Physiology, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Harry C Blair
- Research Service, VA Medical Center, Pittsburgh, Pennsylvania, United States
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
6
|
Wang H, Lu J, Stevens T, Roberts A, Mandel J, Avula R, Ma B, Wu Y, Wang J, Land CV, Finkel T, Vockley JE, Airik M, Airik R, Muzumdar R, Gong Z, Torbenson MS, Prochownik EV. Premature aging and reduced cancer incidence associated with near-complete body-wide Myc inactivation. Cell Rep 2023; 42:112830. [PMID: 37481724 PMCID: PMC10591215 DOI: 10.1016/j.celrep.2023.112830] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/18/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
MYC proto-oncogene dysregulation alters metabolism, translation, and other functions in ways that support tumor induction and maintenance. Although Myc+/- mice are healthier and longer-lived than control mice, the long-term ramifications of more complete Myc loss remain unknown. We now describe the chronic consequences of body-wide Myc inactivation initiated postnatally. "MycKO" mice acquire numerous features of premature aging, including altered body composition and habitus, metabolic dysfunction, hepatic steatosis, and dysregulation of gene sets involved in functions that normally deteriorate with aging. Yet, MycKO mice have extended lifespans that correlate with a 3- to 4-fold lower lifetime cancer incidence. Aging tissues from normal mice and humans also downregulate Myc and gradually alter many of the same Myc target gene sets seen in MycKO mice. Normal aging and its associated cancer predisposition are thus highly linked via Myc.
Collapse
Affiliation(s)
- Huabo Wang
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Jie Lu
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Taylor Stevens
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Alexander Roberts
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Jordan Mandel
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Raghunandan Avula
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; The University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Bingwei Ma
- Tongji University School of Medicine, Shanghai, China
| | - Yijen Wu
- Department of Developmental Biology, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Jinglin Wang
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Central South University, Xiangya School of Medicine, Changsha, Hunan 410013, P.R. China
| | - Clinton Van't Land
- Division of Medical Genetics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Toren Finkel
- Division of Cardiology, The Department of Internal Medicine and the UPMC Aging Institute, Pittsburgh, PA 15224, USA
| | - Jerry E Vockley
- Division of Medical Genetics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Merlin Airik
- Division of Nephrology, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Rannar Airik
- Division of Nephrology, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Radhika Muzumdar
- Division of Endocrinology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Zhenwei Gong
- Division of Endocrinology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Michel S Torbenson
- Division of Laboratory Medicine and Pathology, The Mayo Clinic, Rochester, MN 55905, USA
| | - Edward V Prochownik
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Microbiology and Molecular Genetics, UPMC, Pittsburgh, PA 15261, USA; Hillman Cancer Center of UPMC, Pittsburgh, PA 15232, USA; Pittsburgh Liver Research Center, UPMC, Pittsburgh, PA 15261, USA.
| |
Collapse
|
7
|
Farhadi F, Rajagopal JR, Veziroglu EM, Abdollahi H, Shiri I, Nikpanah M, Morris MA, Zaidi H, Rahmim A, Saboury B. Multi-Scale Temporal Imaging: From Micro- and Meso- to Macro-scale-time Nuclear Medicine. PET Clin 2023; 18:135-148. [DOI: 10.1016/j.cpet.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
8
|
Boitet M, Eun H, Lee T, Kim J, Grailhe R. Non-invasive In Vivo Brain Astrogenesis and Astrogliosis Quantification Using a Far-red E2-Crimson Transgenic Reporter Mouse. Mol Neurobiol 2022; 59:6740-6753. [PMID: 36001234 DOI: 10.1007/s12035-022-02997-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
Abstract
Despite the adaptation of major clinical imaging modalities for small animals, optical bioluminescence imaging technology is the main approach readily reporting gene activity. Yet, in vivo bioluminescence monitoring requires the administration and diffusion of a substrate to the tissues of interest, resulting in experimental variability, high reagent cost, long acquisition time, and stress to the animal. In our study, we avoid such issues upon generating a new transgenic mouse (GFAP-E2crimson) expressing the far-red fluorescent protein E2-crimson under the control of the glial fibrillary acidic protein (GFAP) promoter. Using microscopy, we validated the selective expression of the reporter in the astrocyte cell population and by non-invasive in vivo fluorescence imaging its detection through the scalps and skulls of live animals. In addition, we performed a longitudinal study validating by in vivo imaging that the E2-crimson fluorescence signal is up-regulated, in pups during astrogenesis and in adult mice during astrogliosis upon kainic acid administration. Furthermore, upon crossing GFAP-E2crimson transgenic with 5XFAD Alzheimer's disease mice model, we were able to quantify the chronic inflammation triggered by amyloid deposit and aging over 18 months. As many diseases and conditions can trigger neuroinflammation, we believe that the GFAP-E2crimson reporter mice model delivers tremendous value for the non-invasive quantification of astrogliosis responses in living animals.
Collapse
Affiliation(s)
- Maylis Boitet
- Technology Development Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea
- Department of Biological Chemistry, IPK Campus, Korea University of Science and Technology, 217 Gajeong-ro Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Hyeju Eun
- Technology Development Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea
| | - Taekwan Lee
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jiho Kim
- Screening Discovery Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea
| | - Regis Grailhe
- Technology Development Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea.
- Department of Biological Chemistry, IPK Campus, Korea University of Science and Technology, 217 Gajeong-ro Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
9
|
Oliviero S, Cheong VS, Roberts BC, Orozco Diaz CA, Griffiths W, Bellantuono I, Dall’Ara E. Reproducibility of Densitometric and Biomechanical Assessment of the Mouse Tibia From In Vivo Micro-CT Images. Front Endocrinol (Lausanne) 2022; 13:915938. [PMID: 35846342 PMCID: PMC9282377 DOI: 10.3389/fendo.2022.915938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Interventions for bone diseases (e.g. osteoporosis) require testing in animal models before clinical translation and the mouse tibia is among the most common tested anatomical sites. In vivo micro-Computed Tomography (microCT) based measurements of the geometrical and densitometric properties are non-invasive and therefore constitute an important tool in preclinical studies. Moreover, validated micro-Finite Element (microFE) models can be used for predicting the bone mechanical properties non-invasively. However, considering that the image processing pipeline requires operator-dependant steps, the reproducibility of these measurements has to be assessed. The aim of this study was to evaluate the intra- and inter-operator reproducibility of several bone parameters measured from microCT images. Ten in vivo microCT images of the right tibia of five mice (at 18 and 22 weeks of age) were processed. One experienced operator (intra-operator analysis) and three different operators (inter-operator) aligned each image to a reference through a rigid registration and selected a volume of interest below the growth plate. From each image the following parameters were measured: total bone mineral content (BMC) and density (BMD), BMC in 40 subregions (ten longitudinal sections, four quadrants), microFE-based stiffness and failure load. Intra-operator reproducibility was acceptable for all parameters (precision error, PE < 3.71%), with lowest reproducibility for stiffness (3.06% at week 18, 3.71% at week 22). The inter-operator reproducibility was slightly lower (PE < 4.25%), although still acceptable for assessing the properties of most interventions. The lowest reproducibility was found for BMC in the lateral sector at the midshaft (PE = 4.25%). Densitometric parameters were more reproducible than most standard morphometric parameters calculated in the proximal trabecular bone. In conclusion, microCT and microFE models provide reproducible measurements for non-invasive assessment of the mouse tibia properties.
Collapse
Affiliation(s)
- Sara Oliviero
- Department of Oncology and Metabolism, Mellanby Centre for bone Research, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Industrial Engineering, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Medical Technology Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Vee San Cheong
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Bryant C. Roberts
- Department of Oncology and Metabolism, Mellanby Centre for bone Research, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Carlos Amnael Orozco Diaz
- Department of Oncology and Metabolism, Mellanby Centre for bone Research, University of Sheffield, Sheffield, United Kingdom
| | - William Griffiths
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Ilaria Bellantuono
- Department of Oncology and Metabolism, Mellanby Centre for bone Research, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
| | - Enrico Dall’Ara
- Department of Oncology and Metabolism, Mellanby Centre for bone Research, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
10
|
Li W, Liu YH, Estrada H, Rebling J, Reiss M, Galli S, Nombela-Arrieta C, Razansky D. Tracking Strain-Specific Morphogenesis and Angiogenesis of Murine Calvaria with Large-Scale Optoacoustic and Ultrasound Microscopy. J Bone Miner Res 2022; 37:1032-1043. [PMID: 35220594 PMCID: PMC9311448 DOI: 10.1002/jbmr.4533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/30/2022] [Accepted: 02/20/2022] [Indexed: 11/25/2022]
Abstract
Skull bone development is a dynamic and well-coordinated process playing a key role in maturation and maintenance of the bone marrow (BM), fracture healing, and progression of diseases such as osteoarthritis or osteoporosis. At present, dynamic transformation of the growing bone (osteogenesis) as well as its vascularization (angiogenesis) remain largely unexplored due to the lack of suitable in vivo imaging techniques capable of noninvasive visualization of the whole developing calvaria at capillary-level resolution. We present a longitudinal study on skull bone development using ultrasound-aided large-scale optoacoustic microscopy (U-LSOM). Skull bone morphogenesis and microvascular growth patterns were monitored in three common mouse strains (C57BL/6J, CD-1, and Athymic Nude-Foxn1nu) at the whole-calvaria scale over a 3-month period. Strain-specific differences in skull development were revealed by quantitative analysis of bone and vessel parameters, indicating the coupling between angiogenesis and osteogenesis during skull bone growth in a minimally invasive and label-free manner. The method further enabled identifying BM-specific sinusoidal vessels, and superficial skull vessels penetrating into BM compartments. Our approach furnishes a new high-throughput longitudinal in vivo imaging platform to study morphological and vascular skull alterations in health and disease, shedding light on the critical links between blood vessel formation, skull growth, and regeneration. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Weiye Li
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Yu-Hang Liu
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Héctor Estrada
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Johannes Rebling
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Michael Reiss
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Serena Galli
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Coppola A, Zorzetto G, Piacentino F, Bettoni V, Pastore I, Marra P, Perani L, Esposito A, De Cobelli F, Carcano G, Fontana F, Fiorina P, Venturini M. Imaging in experimental models of diabetes. Acta Diabetol 2022; 59:147-161. [PMID: 34779949 DOI: 10.1007/s00592-021-01826-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/30/2021] [Indexed: 12/01/2022]
Abstract
Translational medicine, experimental medicine and experimental animal models, in particular mice and rats, represent a multidisciplinary field that has made it possible to achieve, in the last decades, important scientific progress. In this review, we have summarized the most frequently used imaging animal models, such as ultrasound (US), micro-CT, MRI and the optical imaging methods, and their main implications in diagnostic and therapeutic fields, with a particular focus on diabetes mellitus, a multifactorial disease extremely widespread among the general population.
Collapse
Affiliation(s)
- Andrea Coppola
- Diagnostic and Interventional Radiology Unit, ASST Settelaghi, Varese, Italy.
| | | | - Filippo Piacentino
- Diagnostic and Interventional Radiology Unit, ASST Settelaghi, Varese, Italy
- Insubria University, Varese, Italy
| | - Valeria Bettoni
- Diagnostic and Interventional Radiology Unit, ASST Settelaghi, Varese, Italy
| | - Ida Pastore
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Paolo Marra
- Department of Diagnostic Radiology, Giovanni XXIII Hospital, Milano-Bicocca University, Bergamo, Italy
| | - Laura Perani
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Esposito
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
- Radiology Unit, San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milan, Italy
| | - Francesco De Cobelli
- Radiology Unit, San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milan, Italy
| | - Giulio Carcano
- Insubria University, Varese, Italy
- General, Emergency, and Transplant Surgery Unit, ASST Settelaghi, Varese, Italy
| | - Federico Fontana
- Diagnostic and Interventional Radiology Unit, ASST Settelaghi, Varese, Italy
- Insubria University, Varese, Italy
| | - Paolo Fiorina
- International Center for T1D, Centro di Ricerca Pediatrica Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche "L. Sacco", Università di Milano, Milan, Italy
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Endocrinology Division, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Massimo Venturini
- Diagnostic and Interventional Radiology Unit, ASST Settelaghi, Varese, Italy
- Insubria University, Varese, Italy
| |
Collapse
|
12
|
Oliviero S, Millard E, Chen Z, Rayson A, Roberts B, Ismail H, Bellantuono I, Dall’Ara E. Accuracy of in vivo microCT imaging in assessing the microstructural properties of the mouse tibia subchondral bone. Front Endocrinol (Lausanne) 2022; 13:1016321. [PMID: 36714558 PMCID: PMC9874301 DOI: 10.3389/fendo.2022.1016321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common musculoskeletal diseases. OA is characterized by degeneration of the articular cartilage as well as the underlying subchondral bone. Post-traumatic osteoarthritis (PTOA) is a subset of OA caused by mechanical trauma. Mouse models, such as destabilization of the medial meniscus (DMM), are useful to study PTOA. Ex vivo micro-Computed Tomography (microCT) imaging is the predominant technique used to scan the mouse knee in OA studies. Nevertheless, in vivo microCT enables the longitudinal assessment of bone microstructure, reducing measurement variability and number of animals required. The effect of image resolution in measuring subchondral bone parameters was previously evaluated only for a limited number of parameters. The aim of this study was to evaluate the ability of in vivo microCT imaging in measuring the microstructural properties of the mouse tibia trabecular and cortical subchondral bone, with respect to ex vivo high resolution imaging, in a DMM model of PTOA. Sixteen male C57BL/6J mice received DMM surgery or sham operation at 14 weeks of age (N=8 per group). The right knee of each mouse was microCT scanned in vivo (10.4μm voxel size) and ex vivo (4.35μm voxel size) at the age of 26 weeks. Each image was aligned to a reference image using rigid registration. The subchondral cortical bone plate thickness was measured at the lateral and medial condyles. Standard morphometric parameters were measured in the subchondral trabecular bone. In vivo microCT imaging led to significant underestimation of bone volume fraction (-14%), bone surface density (-3%) and trabecular number (-16%), whereas trabecular thickness (+3%) and separation (+5%) were significantly overestimated. Nevertheless, most trabecular parameters measured in vivo were well correlated with ex vivo measurements (R2 = 0.69-0.81). Degree of anisotropy, structure model index and connectivity density were measured in vivo with lower accuracy. Excellent accuracy was found for cortical thickness measurements. In conclusion, this study identified what bone morphological parameters can be reliably measured by in vivo microCT imaging of the subchondral bone in the mouse tibia. It highlights that this approach can be used to study longitudinal effects of diseases and treatments on the subchondral cortical bone and on most subchondral trabecular bone parameters, but systematic over- or under-estimations should be considered when interpreting the results.
Collapse
Affiliation(s)
- S. Oliviero
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- Department of Industrial Engineering, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - E. Millard
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Z. Chen
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - A. Rayson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - B.C. Roberts
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - H.M.S. Ismail
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
| | - I. Bellantuono
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
| | - E. Dall’Ara
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
- *Correspondence: E. Dall’Ara,
| |
Collapse
|
13
|
Foessl I, Bassett JHD, Bjørnerem Å, Busse B, Calado Â, Chavassieux P, Christou M, Douni E, Fiedler IAK, Fonseca JE, Hassler E, Högler W, Kague E, Karasik D, Khashayar P, Langdahl BL, Leitch VD, Lopes P, Markozannes G, McGuigan FEA, Medina-Gomez C, Ntzani E, Oei L, Ohlsson C, Szulc P, Tobias JH, Trajanoska K, Tuzun Ş, Valjevac A, van Rietbergen B, Williams GR, Zekic T, Rivadeneira F, Obermayer-Pietsch B. Bone Phenotyping Approaches in Human, Mice and Zebrafish - Expert Overview of the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal traits TranslatiOnal NEtwork"). Front Endocrinol (Lausanne) 2021; 12:720728. [PMID: 34925226 PMCID: PMC8672201 DOI: 10.3389/fendo.2021.720728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
A synoptic overview of scientific methods applied in bone and associated research fields across species has yet to be published. Experts from the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal Traits translational Network") Working Group 2 present an overview of the routine techniques as well as clinical and research approaches employed to characterize bone phenotypes in humans and selected animal models (mice and zebrafish) of health and disease. The goal is consolidation of knowledge and a map for future research. This expert paper provides a comprehensive overview of state-of-the-art technologies to investigate bone properties in humans and animals - including their strengths and weaknesses. New research methodologies are outlined and future strategies are discussed to combine phenotypic with rapidly developing -omics data in order to advance musculoskeletal research and move towards "personalised medicine".
Collapse
Affiliation(s)
- Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| | - J. H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Åshild Bjørnerem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Norwegian Research Centre for Women’s Health, Oslo University Hospital, Oslo, Norway
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Ângelo Calado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | | | - Maria Christou
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Eleni Douni
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Imke A. K. Fiedler
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Eva Hassler
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University Graz, Graz, Austria
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Erika Kague
- The School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, United Kingdom
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Patricia Khashayar
- Center for Microsystems Technology, Imec and Ghent University, Ghent, Belgium
| | - Bente L. Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Victoria D. Leitch
- Innovative Manufacturing Cooperative Research Centre, Royal Melbourne Institute of Technology, School of Engineering, Carlton, VIC, Australia
| | - Philippe Lopes
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Georgios Markozannes
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | | | | | - Evangelia Ntzani
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
- Department of Health Services, Policy and Practice, Center for Research Synthesis in Health, School of Public Health, Brown University, Providence, RI, United States
| | - Ling Oei
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pawel Szulc
- INSERM UMR 1033, University of Lyon, Lyon, France
| | - Jonathan H. Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, Bristol, University of Bristol, Bristol, United Kingdom
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Şansın Tuzun
- Physical Medicine & Rehabilitation Department, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Amina Valjevac
- Department of Human Physiology, School of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Bert van Rietbergen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Tatjana Zekic
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | | | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| |
Collapse
|
14
|
Zhang C, Wang D, Wang J, Wang L, Qiu W, Kume T, Dowell R, Yi R. Escape of hair follicle stem cells causes stem cell exhaustion during aging. NATURE AGING 2021; 1:889-903. [PMID: 37118327 PMCID: PMC11323283 DOI: 10.1038/s43587-021-00103-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 07/25/2021] [Indexed: 04/30/2023]
Abstract
Stem cell (SC) exhaustion is a hallmark of aging. However, the process of SC depletion during aging has not been observed in live animals, and the underlying mechanism contributing to tissue deterioration remains obscure. We find that, in aged mice, epithelial cells escape from the hair follicle (HF) SC compartment to the dermis, contributing to HF miniaturization. Single-cell RNA-seq and assay for transposase-accessible chromatin using sequencing (ATAC-seq) reveal reduced expression of cell adhesion and extracellular matrix genes in aged HF-SCs, many of which are regulated by Foxc1 and Nfatc1. Deletion of Foxc1 and Nfatc1 recapitulates HF miniaturization and causes hair loss. Live imaging captures individual epithelial cells migrating away from the SC compartment and HF disintegration. This study illuminates a hitherto unknown activity of epithelial cells escaping from their niche as a mechanism underlying SC reduction and tissue degeneration. Identification of homeless epithelial cells in aged tissues provides a new perspective for understanding aging-associated diseases.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Pathology, Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dongmei Wang
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Pathology, Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jingjing Wang
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Pathology, Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Li Wang
- 10x Genomics, Pleasanton, CA, USA
| | - Wenli Qiu
- Lung Biology Center, Department of Medicine, UCSF, San Francisco, CA, USA
| | - Tsutomu Kume
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Robin Dowell
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Rui Yi
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA.
- Department of Pathology, Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
15
|
Oliviero S, Roberts M, Owen R, Reilly GC, Bellantuono I, Dall'Ara E. Non-invasive prediction of the mouse tibia mechanical properties from microCT images: comparison between different finite element models. Biomech Model Mechanobiol 2021; 20:941-955. [PMID: 33523337 PMCID: PMC8154847 DOI: 10.1007/s10237-021-01422-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/07/2021] [Indexed: 01/01/2023]
Abstract
New treatments for bone diseases require testing in animal models before clinical translation, and the mouse tibia is among the most common models. In vivo micro-Computed Tomography (microCT)-based micro-Finite Element (microFE) models can be used for predicting the bone strength non-invasively, after proper validation against experimental data. Different modelling techniques can be used to estimate the bone properties, and the accuracy associated with each is unclear. The aim of this study was to evaluate the ability of different microCT-based microFE models to predict the mechanical properties of the mouse tibia under compressive load. Twenty tibiae were microCT scanned at 10.4 µm voxel size and subsequently compressed at 0.03 mm/s until failure. Stiffness and failure load were measured from the load-displacement curves. Different microFE models were generated from each microCT image, with hexahedral or tetrahedral mesh, and homogeneous or heterogeneous material properties. Prediction accuracy was comparable among models. The best correlations between experimental and predicted mechanical properties, as well as lower errors, were obtained for hexahedral models with homogeneous material properties. Experimental stiffness and predicted stiffness were reasonably well correlated (R2 = 0.53-0.65, average error of 13-17%). A lower correlation was found for failure load (R2 = 0.21-0.48, average error of 9-15%). Experimental and predicted mechanical properties normalized by the total bone mass were strongly correlated (R2 = 0.75-0.80 for stiffness, R2 = 0.55-0.81 for failure load). In conclusion, hexahedral models with homogeneous material properties based on in vivo microCT images were shown to best predict the mechanical properties of the mouse tibia.
Collapse
Affiliation(s)
- S Oliviero
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield, UK
| | - M Roberts
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - R Owen
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
- Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham Biodiscovery Institute, University Park, UK
| | - G C Reilly
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
| | - I Bellantuono
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield, UK
- Healthy Lifespan Institute, The Medical School, University of Sheffield, Sheffield, UK
| | - E Dall'Ara
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield, UK.
- Healthy Lifespan Institute, The Medical School, University of Sheffield, Sheffield, UK.
| |
Collapse
|
16
|
Petr MA, Alfaras I, Krawcyzk M, Bair WN, Mitchell SJ, Morrell CH, Studenski SA, Price NL, Fishbein KW, Spencer RG, Scheibye-Knudsen M, Lakatta EG, Ferrucci L, Aon MA, Bernier M, de Cabo R. A cross-sectional study of functional and metabolic changes during aging through the lifespan in male mice. eLife 2021; 10:e62952. [PMID: 33876723 PMCID: PMC8099423 DOI: 10.7554/elife.62952] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Aging is associated with distinct phenotypical, physiological, and functional changes, leading to disease and death. The progression of aging-related traits varies widely among individuals, influenced by their environment, lifestyle, and genetics. In this study, we conducted physiologic and functional tests cross-sectionally throughout the entire lifespan of male C57BL/6N mice. In parallel, metabolomics analyses in serum, brain, liver, heart, and skeletal muscle were also performed to identify signatures associated with frailty and age-dependent functional decline. Our findings indicate that declines in gait speed as a function of age and frailty are associated with a dramatic increase in the energetic cost of physical activity and decreases in working capacity. Aging and functional decline prompt organs to rewire their metabolism and substrate selection and toward redox-related pathways, mainly in liver and heart. Collectively, the data provide a framework to further understand and characterize processes of aging at the individual organism and organ levels.
Collapse
Affiliation(s)
- Michael A Petr
- Center for Healthy Aging, ICMM, University of CopenhagenCopenhagenDenmark
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
| | - Irene Alfaras
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
| | - Melissa Krawcyzk
- Laboratory of Cardiovascular Science, National Institute on Aging, NIHBaltimoreUnited States
| | - Woei-Nan Bair
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
- Department of Physical Therapy, University of SciencesPhiladelphiaUnited States
| | - Sarah J Mitchell
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
| | - Christopher H Morrell
- Laboratory of Cardiovascular Science, National Institute on Aging, NIHBaltimoreUnited States
| | - Stephanie A Studenski
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
- Division of Geriatric Medicine, Department of Medicine, University of PittsburghPittsburghUnited States
| | - Nathan L Price
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Yale University School of MedicineNew HavenUnited States
- Vascular Biology and Therapeutics Program, Yale University School of MedicineNew HavenUnited States
- Department of Comparative Medicine, Yale University School of MedicineNew HavenUnited States
- Department of Pathology, Yale University School of MedicineNew HavenUnited States
| | - Kenneth W Fishbein
- Laboratory of Clinical Investigation, National Institute on Aging, NIHBaltimoreUnited States
| | - Richard G Spencer
- Laboratory of Clinical Investigation, National Institute on Aging, NIHBaltimoreUnited States
| | | | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, NIHBaltimoreUnited States
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
| | - Miguel A Aon
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
- Laboratory of Cardiovascular Science, National Institute on Aging, NIHBaltimoreUnited States
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIHBaltimoreUnited States
| |
Collapse
|
17
|
Zanjani-Pour S, Giorgi M, Dall'Ara E. Development of Subject Specific Finite Element Models of the Mouse Knee Joint for Preclinical Applications. Front Bioeng Biotechnol 2020; 8:558815. [PMID: 33178671 PMCID: PMC7593650 DOI: 10.3389/fbioe.2020.558815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/03/2020] [Indexed: 11/20/2022] Open
Abstract
Osteoarthritis is the most common musculoskeletal disabling disease worldwide. Preclinical studies on mice are commonly performed to test new interventions. Finite element (FE) models can be used to study joint mechanics, but usually simplified geometries are used. The aim of this project was to create a realistic subject specific FE model of the mouse knee joint for the assessment of joint mechanical properties. Four different FE models of a C57Bl/6 female mouse knee joint were created based on micro-computed tomography images of specimens stained with phosphotungstic acid in order to include different features: individual cartilage layers with meniscus, individual cartilage layers without meniscus, homogeneous cartilage layers with two different thickness values, and homogeneous cartilage with same thickness for both condyles. They were all analyzed under compressive displacement and the cartilage contact pressure was compared at 0.3 N reaction force. Peak contact pressure in the femur cartilage was 25% lower in the model with subject specific cartilage compared to the simpler model with homogeneous cartilage. A much more homogeneous pressure distribution across the joint was observed in the model with meniscus, with cartilage peak pressure 5–34% lower in the two condyles compared to that with individual cartilage layers. In conclusion, modeling the meniscus and individual cartilage was found to affect the pressure distribution in the mouse knee joint under compressive load and should be included in realistic models for assessing the effect of interventions preclinically.
Collapse
Affiliation(s)
- Sahand Zanjani-Pour
- Department of Oncology and Metabolism, Mellanby Center for Bone Research, University of Sheffield, Sheffield, United Kingdom.,Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Mario Giorgi
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom.,Certara Quantitative System Pharmacology, Certara UK Ltd., Simcyp Division, Sheffield, United Kingdom
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, Mellanby Center for Bone Research, University of Sheffield, Sheffield, United Kingdom.,Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
18
|
Oliviero S, Owen R, Reilly GC, Bellantuono I, Dall'Ara E. Optimization of the failure criterion in micro-Finite Element models of the mouse tibia for the non-invasive prediction of its failure load in preclinical applications. J Mech Behav Biomed Mater 2020; 113:104190. [PMID: 33191174 DOI: 10.1016/j.jmbbm.2020.104190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/23/2020] [Accepted: 10/27/2020] [Indexed: 01/21/2023]
Abstract
New treatments against osteoporosis require testing in animal models and the mouse tibia is among the most common studied anatomical sites. In vivo micro-Computed Tomography (microCT) based micro-Finite Element (microFE) models can be used for predicting the bone strength non-invasively, after proper validation against experiments. The aim of this study was to evaluate the ability of different microCT-based bone parameters and microFE models to predict tibial structural mechanical properties in compression. Twenty tibiae were scanned at 10.4 μm voxel size and subsequently tested in uniaxial compression at 0.03 mm/s until failure. Stiffness and failure load were measured from the load-displacement curves. Standard morphometric parameters were measured from the microCT images. The spatial distribution of bone mineral content (BMC) was evaluated by dividing the tibia into 40 regions. MicroFE models were generated by converting each microCT image into a voxel-based mesh with homogeneous isotropic material properties. Failure load was estimated by using different failure criteria, and the optimized parameters were selected by minimising the errors with respect to experimental measurements. Experimental and predicted stiffness were moderately correlated (R2 = 0.65, error = 14% ± 8%). Normalized failure load was best predicted by microFE models (R2 = 0.81, error = 9% ± 6%). Failure load was not correlated to the morphometric parameters and weakly correlated with some geometrical parameters (R2 < 0.37). In conclusion, microFE models can improve the current estimation of the mouse tibia structural properties and in this study an optimal failure criterion has been defined. Since it is a non-invasive method, this approach can be applied longitudinally for evaluating temporal changes in the bone strength.
Collapse
Affiliation(s)
- S Oliviero
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, UK; INSIGNEO Institute for in Silico Medicine, University of Sheffield, UK
| | - R Owen
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, UK; Department of Materials Science and Engineering, University of Sheffield, UK; Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham Biodiscovery Institute, University Park, UK
| | - G C Reilly
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, UK; Department of Materials Science and Engineering, University of Sheffield, UK
| | - I Bellantuono
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, UK; INSIGNEO Institute for in Silico Medicine, University of Sheffield, UK; Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, UK
| | - E Dall'Ara
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, UK; INSIGNEO Institute for in Silico Medicine, University of Sheffield, UK.
| |
Collapse
|
19
|
Llambrich S, Wouters J, Himmelreich U, Dierssen M, Sharpe J, Gsell W, Martínez-Abadías N, Vande Velde G. ViceCT and whiceCT for simultaneous high-resolution visualization of craniofacial, brain and ventricular anatomy from micro-computed tomography. Sci Rep 2020; 10:18772. [PMID: 33128010 PMCID: PMC7599226 DOI: 10.1038/s41598-020-75720-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Up to 40% of congenital diseases present disturbances of brain and craniofacial development resulting in simultaneous alterations of both systems. Currently, the best available method to preclinically visualize the brain and the bones simultaneously is to co-register micro-magnetic resonance (µMR) and micro-computed tomography (µCT) scans of the same specimen. However, this requires expertise and access to both imaging techniques, dedicated software and post-processing knowhow. To provide a more affordable, reliable and accessible alternative, recent research has focused on optimizing a contrast-enhanced µCT protocol using iodine as contrast agent that delivers brain and bone images from a single scan. However, the available methods still cannot provide the complete visualization of both the brain and whole craniofacial complex. In this study, we have established an optimized protocol to diffuse the contrast into the brain that allows visualizing the brain parenchyma and the complete craniofacial structure in a single ex vivo µCT scan (whiceCT). In addition, we have developed a new technique that allows visualizing the brain ventricles using a bilateral stereotactic injection of iodine-based contrast (viceCT). Finally, we have tested both techniques in a mouse model of Down syndrome, as it is a neurodevelopmental disorder with craniofacial, brain and ventricle defects. The combined use of viceCT and whiceCT provides a complete visualization of the brain and bones with intact craniofacial structure of an adult mouse ex vivo using a single imaging modality.
Collapse
Affiliation(s)
- Sergi Llambrich
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Herestraat 49 O&N1 box 505, 3000, Leuven, Belgium.,Molecular Small Animal Imaging Centre (MoSAIC), KU Leuven, Leuven, Belgium
| | - Jens Wouters
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Herestraat 49 O&N1 box 505, 3000, Leuven, Belgium.,Molecular Small Animal Imaging Centre (MoSAIC), KU Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Herestraat 49 O&N1 box 505, 3000, Leuven, Belgium.,Molecular Small Animal Imaging Centre (MoSAIC), KU Leuven, Leuven, Belgium
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG, The Barcelona Institute of Science and Technology, 08003, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), 08003, Barcelona, Spain
| | - James Sharpe
- EMBL Barcelona, European Molecular Biology Laboratory, Barcelona, Spain Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Willy Gsell
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Herestraat 49 O&N1 box 505, 3000, Leuven, Belgium.,Molecular Small Animal Imaging Centre (MoSAIC), KU Leuven, Leuven, Belgium
| | - Neus Martínez-Abadías
- GREAB-Research Group in Biological Anthropology. Department of Evolutionary Biology, Ecology and Environmental Sciences, BEECA. Universitat de Barcelona, Barcelona, Spain
| | - Greetje Vande Velde
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Herestraat 49 O&N1 box 505, 3000, Leuven, Belgium. .,Molecular Small Animal Imaging Centre (MoSAIC), KU Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
Scheuren AC, Kuhn GA, Müller R. Effects of long-term in vivo micro-CT imaging on hallmarks of osteopenia and frailty in aging mice. PLoS One 2020; 15:e0239534. [PMID: 32966306 PMCID: PMC7511008 DOI: 10.1371/journal.pone.0239534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/08/2020] [Indexed: 11/26/2022] Open
Abstract
In vivo micro-CT has already been used to monitor microstructural changes of bone in mice of different ages and in models of age-related diseases such as osteoporosis. However, as aging is accompanied by frailty and subsequent increased sensitivity to external stimuli such as handling and anesthesia, the extent to which longitudinal imaging can be applied in aging studies remains unclear. Consequently, the potential of monitoring individual mice during the entire aging process-from healthy to frail status-has not yet been exploited. In this study, we assessed the effects of long-term in vivo micro-CT imaging-consisting of 11 imaging sessions over 20 weeks-on hallmarks of aging both on a local (i.e., static and dynamic bone morphometry) and systemic (i.e., frailty index (FI) and body weight) level at various stages of the aging process. Furthermore, using a premature aging model (PolgA(D257A/D257A)), we assessed whether these effects differ between genotypes. The 6th caudal vertebrae of 4 groups of mice (PolgA(D257A/D257A) and PolgA(+/+)) were monitored by in vivo micro-CT every 2 weeks. One group was subjected to 11 scans between weeks 20 and 40 of age, whereas the other groups were subjected to 5 scans between weeks 26-34, 32-40 and 40-46, respectively. The long-term monitoring approach showed small but significant changes in the static bone morphometric parameters compared to the other groups. However, no interaction effect between groups and genotype was found, suggesting that PolgA mutation does not render bone more or less susceptible to long-term micro-CT imaging. The differences between groups observed in the static morphometric parameters were less pronounced in the dynamic morphometric parameters. Moreover, the body weight and FI were not affected by more frequent imaging sessions. Finally, we observed that longitudinal designs including baseline measurements at young adult age are more powerful at detecting effects of in vivo micro-CT imaging on hallmarks of aging than cross-sectional comparisons between multiple groups of aged mice subjected to fewer imaging sessions.
Collapse
Affiliation(s)
| | - Gisela A. Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Osborne B, Bakula D, Ben Ezra M, Dresen C, Hartmann E, Kristensen SM, Mkrtchyan GV, Nielsen MH, Petr MA, Scheibye-Knudsen M. New methodologies in ageing research. Ageing Res Rev 2020; 62:101094. [PMID: 32512174 DOI: 10.1016/j.arr.2020.101094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/14/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
Ageing is arguably the most complex phenotype that occurs in humans. To understand and treat ageing as well as associated diseases, highly specialised technologies are emerging that reveal critical insight into the underlying mechanisms and provide new hope for previously untreated diseases. Herein, we describe the latest developments in cutting edge technologies applied across the field of ageing research. We cover emerging model organisms, high-throughput methodologies and machine-driven approaches. In all, this review will give you a glimpse of what will be pushing the field onwards and upwards.
Collapse
Affiliation(s)
- Brenna Osborne
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Ben Ezra
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Dresen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Esben Hartmann
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Stella M Kristensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Garik V Mkrtchyan
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Malte H Nielsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael A Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
22
|
Taylor EN, Huang N, Wisco J, Wang Y, Morgan KG, Hamilton JA. The brains of aged mice are characterized by altered tissue diffusion properties and cerebral microbleeds. J Transl Med 2020; 18:277. [PMID: 32641073 PMCID: PMC7346388 DOI: 10.1186/s12967-020-02441-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022] Open
Abstract
Background Brain aging is a major risk factor in the progression of cognitive diseases including Alzheimer’s disease (AD) and vascular dementia. We investigated a mouse model of brain aging up to 24 months old (mo). Methods A high field (11.7T) MRI protocol was developed to characterize specific features of brain aging including the presence of cerebral microbleeds (CMBs), morphology of grey and white matter, and tissue diffusion properties. Mice were selected from age categories of either young (3 mo), middle-aged (18 mo), or old (24 mo) and fed normal chow over the duration of the study. Mice were imaged in vivo with multimodal MRI, including conventional T2-weighted (T2W) and T2*-weighted (T2*W) imaging, followed by ex vivo diffusion-weighted imaging (DWI) and T2*W MR-microscopy to enhance the detection of microstructural features. Results Structural changes observed in the mouse brain with aging included reduced cortical grey matter volume and enlargement of the brain ventricles. A remarkable age-related change in the brains was the development of CMBs found starting at 18 mo and increasing in total volume at 24 mo, primarily in the thalamus. CMBs presence was confirmed with high resolution ex vivo MRI and histology. DWI detected further brain tissue changes in the aged mice including reduced fractional anisotropy, increased radial diffusion, increased mean diffusion, and changes in the white matter fibers visualized by color-coded tractography, including around a large cortical CMB. Conclusions The mouse is a valuable model of age-related vascular contributions to cognitive impairment and dementia (VCID). In composite, these methods and results reveal brain aging in older mice as a multifactorial process including CMBs and tissue diffusion alterations that can be well characterized by high field MRI.
Collapse
Affiliation(s)
- Erik N Taylor
- Department of Radiology, University of New Mexico, Albuquerque, NM, USA. .,Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA. .,Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Nasi Huang
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA.,Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Jonathan Wisco
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Yandan Wang
- Department of Health Sciences, Boston University, Boston, MA, USA
| | | | - James A Hamilton
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA. .,Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
23
|
Roberts BC, Arredondo Carrera HM, Zanjani-Pour S, Boudiffa M, Wang N, Gartland A, Dall'Ara E. PTH(1-34) treatment and/or mechanical loading have different osteogenic effects on the trabecular and cortical bone in the ovariectomized C57BL/6 mouse. Sci Rep 2020; 10:8889. [PMID: 32483372 PMCID: PMC7264307 DOI: 10.1038/s41598-020-65921-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/12/2020] [Indexed: 12/18/2022] Open
Abstract
In preclinical mouse models, a synergistic anabolic response to PTH(1–34) and tibia loading was shown. Whether combined treatment improves bone properties with oestrogen deficiency, a cardinal feature of osteoporosis, remains unknown. This study quantified the individual and combined longitudinal effects of PTH(1–34) and loading on the bone morphometric and densitometric properties in ovariectomised mice. C57BL/6 mice were ovariectomised at 14-weeks-old and treated either with injections of PTH(1–34); compressive loading of the right tibia; both interventions concurrently; or both interventions on alternating weeks. Right tibiae were microCT-scanned from 14 until 24-weeks-old. Trabecular metaphyseal and cortical midshaft morphometric properties, and bone mineral content (BMC) in 40 different regions of the tibia were measured. Mice treated only with loading showed the highest trabecular bone volume fraction at week 22. Cortical thickness was higher with co-treatment than in the mice treated with PTH alone. In the mid-diaphysis, increases in BMC were significantly higher with loading than PTH. In ovariectomised mice, the osteogenic benefits of co-treatment on the trabecular bone were lower than loading alone. However, combined interventions had increased, albeit regionally-dependent, benefits to cortical bone. Increased benefits were largest in the mid-diaphysis and postero-laterally, regions subjected to higher strains under compressive loads.
Collapse
Affiliation(s)
- Bryant C Roberts
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom. .,Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom.
| | - Hector M Arredondo Carrera
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.,MRC Arthritis Research UK, Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, United Kingdom
| | - Sahand Zanjani-Pour
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.,Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Maya Boudiffa
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.,MRC Arthritis Research UK, Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, United Kingdom
| | - Ning Wang
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.,MRC Arthritis Research UK, Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, United Kingdom
| | - Alison Gartland
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.,MRC Arthritis Research UK, Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, United Kingdom
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.,Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom.,MRC Arthritis Research UK, Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
24
|
Pepe V, Oliviero S, Cristofolini L, Dall'Ara E. Regional Nanoindentation Properties in Different Locations on the Mouse Tibia From C57BL/6 and Balb/C Female Mice. Front Bioeng Biotechnol 2020; 8:478. [PMID: 32500069 PMCID: PMC7243342 DOI: 10.3389/fbioe.2020.00478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/24/2020] [Indexed: 01/03/2023] Open
Abstract
The local spatial heterogeneity of the material properties of the cortical and trabecular bone extracted from the mouse tibia is not well-known. Nevertheless, its characterization is fundamental to be able to study comprehensively the effect of interventions and to generate computational models to predict the bone strength preclinically. The goal of this study was to evaluate the nanoindentation properties of bone tissue extracted from two different mouse strains across the tibia length and in different sectors. Left tibiae were collected from four female mice, two C57BL/6, and two Balb/C mice. Nanoindentations with maximum 6 mN load were performed on different microstructures, regions along the axis of the tibiae, and sectors (379 in total). Reduced modulus (Er) and hardness (H) were computed for each indentation. Trabecular bone of Balb/C mice was 21% stiffer than that of C57BL/6 mice (20.8 ± 4.1 GPa vs. 16.5 ± 7.1 GPa). Moreover, the proximal regions of the bones were 13-36% less stiff than the mid-shaft and distal regions of the same bones. No significant differences were found for the different sectors for E r and H for Balb/C mice. The bone in the medial sector was found to be 8-14% harder and stiffer than the bone in the anterior or posterior sectors for C57BL/6 mice. In conclusion, this study showed that the nanoindentation properties of the mouse tibia are heterogeneous across the tibia length and the trabecular bone properties are different between Balb/C and C57BL/6 mice. These results will help the research community to identify regions where to characterize the mechanical properties of the bone during preclinical optimisation of treatments for skeletal diseases.
Collapse
Affiliation(s)
- Valentina Pepe
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, United Kingdom.,INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom.,Department of Industrial Engineering, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Sara Oliviero
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, United Kingdom.,INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Luca Cristofolini
- Department of Industrial Engineering, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, United Kingdom.,INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
25
|
Oliviero S, Giorgi M, Laud PJ, Dall’Ara E. Effect of repeated in vivo microCT imaging on the properties of the mouse tibia. PLoS One 2019; 14:e0225127. [PMID: 31751367 PMCID: PMC6874075 DOI: 10.1371/journal.pone.0225127] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/29/2019] [Indexed: 01/01/2023] Open
Abstract
In longitudinal studies, in vivo micro-Computed Tomography (microCT) imaging is used to investigate bone changes over time due to interventions in mice. However, ionising radiation can provoke significant variations in bone morphometric parameters. In a previous study, we evaluated the effect of reducing the integration time on the properties of the mouse tibia measured from microCT images. A scanning procedure (100 ms integration time, 256 mGy nominal radiation dose) was selected as the best compromise between image quality and radiation dose induced on the animal. In this work, the effect of repeated in vivo scans has been evaluated using the selected procedure. The right tibia of twelve female C57BL/6 (six wild type, WT, six ovariectomised, OVX) and twelve BALB/c (six WT, six OVX) mice was scanned every two weeks, starting at week 14 of age. At week 24, mice were sacrificed and both tibiae were scanned. Standard trabecular and cortical morphometric parameters were calculated. The spatial distribution of densitometric parameters (e.g. bone mineral content) was obtained by dividing each tibia in 40 partitions. Stiffness and strength in compression were estimated using homogeneous linear elastic microCT-based micro-Finite Element models. Differences between right (irradiated) and left (non-irradiated control) tibiae were evaluated for each parameter. The irradiated tibiae had higher Tb.Th (+3.3%) and Tb.Sp (+11.6%), and lower Tb.N (-14.2%) compared to non-irradiated tibiae, consistently across both strains and intervention groups. A reduction in Tb.BV/TV (-14.9%) was also observed in the C57BL/6 strain. In the OVX group, a small reduction was also observed in Tt.Ar (-5.0%). In conclusion, repeated microCT scans (at 256 mGy, 5 scans, every two weeks) had limited effects on the mouse tibia, compared to the expected changes induced by bone treatments. Therefore, the selected scanning protocol is acceptable for measuring the effect of bone interventions in vivo.
Collapse
Affiliation(s)
- Sara Oliviero
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| | - Mario Giorgi
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Certara QSP, Certara UK Ltd., Simcyp Division, Sheffield, United Kingdom
| | - Peter J. Laud
- Statistical Services Unit, University of Sheffield, Sheffield, United Kingdom
| | - Enrico Dall’Ara
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
26
|
Roberts BC, Giorgi M, Oliviero S, Wang N, Boudiffa M, Dall'Ara E. The longitudinal effects of ovariectomy on the morphometric, densitometric and mechanical properties in the murine tibia: A comparison between two mouse strains. Bone 2019; 127:260-270. [PMID: 31254730 DOI: 10.1016/j.bone.2019.06.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022]
Abstract
Oestrogen deficiency-related bone loss in the ovariectomized (OVX) mouse is a common model for osteoporosis. However, a comprehensive in vivo assessment of intervention-related changes in multiple bone properties, and in multiple mouse strains, is required in order to identify an appropriate model for future evaluation of novel anti-osteoporotic therapies. The aim of this study was to evaluate the effect of OVX on the morphometric and densitometric properties measured in the microCT images and the mechanical properties estimated with finite element models of the tibia in two mouse strains, C57BL/6 and BALB/c. 14-weeks-old female C57BL/6 and BALB/c mice were divided into two groups per strain: (1) ovariectomized, (2) non-operated control. The right tibia was scanned at baseline (14 weeks) and then every two weeks thereafter, until 24-weeks-old, using in vivo microCT. Changes in trabecular and cortical bone morphometry, spatiotemporal changes in densitometric properties and in mechanical properties (from micro-finite element (μFE) analysis) were computed. Differences between OVX and non-operated controls were evaluated by ANCOVA, adjusted for 14-weeks baseline. In morphometry, trabecular bone mass was significantly reduced in both C57BL/6 and BALB/c from four weeks following surgery. Though the OVX-effect was transient in BALB/c as bone mass reached skeletal homeostasis. OVX inhibited the age-related thickening of cortical bone only in C57BL/6. In both strains, increments in bone mineral content were significantly lower with OVX only in the proximal tibia, with intervention-related differences increasing with time. OVX had no effect on μFE estimates of stiffness nor failure load in either strain. The results of this study show strain-, time- and region-(trabecular or cortical) dependent changes in morphometric and densitometric properties. These findings highlight the importance of choosing an appropriate mouse model and time points for research of treatments against accelerated bone resorption.
Collapse
Affiliation(s)
- Bryant C Roberts
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Mario Giorgi
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Certara QSP, Certara UK Ltd., Simcyp Division, Sheffield, UK
| | - Sara Oliviero
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Ning Wang
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, UK
| | - Maya Boudiffa
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, UK
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), University of Sheffield, Sheffield, UK.
| |
Collapse
|
27
|
Becker K, Schwarz F, Rauch NJ, Khalaph S, Mihatovic I, Drescher D. Can implants move in bone? A longitudinal in vivo micro-CT analysis of implants under constant forces in rat vertebrae. Clin Oral Implants Res 2019; 30:1179-1189. [PMID: 31494964 DOI: 10.1111/clr.13531] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/24/2019] [Accepted: 08/22/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Whereas stationary stability of implants has been postulated for decades, recent studies suggested a phenomenon termed implant migration. This describes a change in position of implants as a reaction to applied forces. The present study aims at employing image registration of in vivo micro-CT scans from different time points and to assess (a) if migration of continuously loaded implants is possible and (b) migration correlates with the force magnitude. MATERIAL AND METHODS Two customized machined implants were placed in the dorsal portion of caudal vertebrae in n = 61 rats and exposed to standardized forces (0.5 N, 1.0 N, and 1.5 N) applied through a flat nickel-titanium contraction spring, or no forces (control). Micro-CT scans were performed at 0, 1, 2, 4, 6, and 8 weeks after surgery. The baseline image was registered with the forthcoming scans. Implant migration was measured as the Euclidean distance between implant tips. Bone remodeling was assessed between the baseline and the forthcoming scans. RESULTS The findings confirmed a positional change of the implants at 2 and 8 weeks of healing, and a linear association between applied force and velocity of movement (anterior implant: χ2 = 12.12, df = 3, and p = .007 and posterior implant: χ2 = 20.35, df = 3, and p < .001). Bone apposition was observed around the implants and accompanied by formation of load-bearing trabeculae and a general cortical thickening close and also distant to the implants. CONCLUSION The present analysis confirmed that implants can migrate in bone. The applied forces seemed to stimulate bone thickening, which could explain why implants migrate without affecting stability.
Collapse
Affiliation(s)
- Kathrin Becker
- Department of Orthodontics, Universitätsklinikum Düsseldorf, Düsseldorf, Germany.,Department of Oral Surgery and Implantology, Carolinum, Goethe University, Frankfurt, Germany
| | - Frank Schwarz
- Department of Oral Surgery and Implantology, Carolinum, Goethe University, Frankfurt, Germany
| | - Nicole Jasmin Rauch
- Department of Orthodontics, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Silava Khalaph
- Department of Orthodontics, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Ilja Mihatovic
- Department of Oral Surgery, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Dieter Drescher
- Department of Orthodontics, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
28
|
Zhang Y, Dall’Ara E, Viceconti M, Kadirkamanathan V. A new method to monitor bone geometry changes at different spatial scales in the longitudinal in vivo μCT studies of mice bones. PLoS One 2019; 14:e0219404. [PMID: 31329619 PMCID: PMC6645529 DOI: 10.1371/journal.pone.0219404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/21/2019] [Indexed: 12/17/2022] Open
Abstract
Longitudinal studies of bone adaptation in mice using in vivo micro-computed tomography (μCT) have been commonly used for pre-clinical evaluation of physical and pharmacological interventions. The main advantage of this approach is to use each mouse as its own control, reducing considerably the sample size required by statistical power analysis. To date, multi-scale estimation of bone adaptations become essential since the bone activity that takes place at different scales may be associated with different bone mechanisms. Measures of bone adaptations at different time scales have been attempted in a previous study. This paper extends quantification of bone activity at different spatial scales with a proposition of a novel framework. The method involves applying level-set method (LSM) to track the geometric changes from the longitudinal in vivo μCT scans of mice tibia. Bone low- and high-spatial frequency patterns are then estimated using multi-resolution analysis. The accuracy of the framework is quantified by applying it to two times separated scanned images with synthetically manipulated global and/or local activity. The Root Mean Square Deviation (RMSD) was approximately 1.5 voxels or 0.7 voxels for the global low-spatial frequency or local high-spatial frequency changes, respectively. The framework is further applied to the study of bone changes in longitudinal datasets of wild-type mice tibiae over time and space. The results demonstrate the ability for the spatio-temporal quantification and visualisation of bone activity at different spatial scales in longitudinal studies thus providing further insight into bone adaptation mechanisms.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Automatic Control and Systems Engineering, The University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
| | - Enrico Dall’Ara
- INSIGNEO Institute for in Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
- Department of Oncology & Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Marco Viceconti
- Department of Industrial Engineering, Alma Mater Studiorum, University of Bologna, Bologna Area, Italy
- Medical Technology Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Visakan Kadirkamanathan
- Department of Automatic Control and Systems Engineering, The University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
29
|
Janus J, Kanber B, Mahbuba W, Beynon C, Ramnarine KV, Lambert DG, Samani NJ, Stringer EJ, Kelly ME. A preclinical ultrasound method for the assessment of vascular disease progression in murine models. ULTRASOUND : JOURNAL OF THE BRITISH MEDICAL ULTRASOUND SOCIETY 2019; 27:85-93. [PMID: 31037092 DOI: 10.1177/1742271x18793919] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/09/2018] [Indexed: 11/16/2022]
Abstract
Introduction The efficacy of preclinical ultrasound at providing a quantitative assessment of mouse models of vascular disease is relatively unknown. In this study, preclinical ultrasound was used in combination with a semi-automatic image processing method to track arterial distension alterations in mouse models of abdominal aortic aneurysm and atherosclerosis. Methods Longitudinal B-mode ultrasound images of the abdominal aorta were acquired using a preclinical ultrasound scanner. Arterial distension was assessed using a semi-automatic image processing algorithm to track vessel wall motion over the cardiac cycle. A standard, manual analysis method was applied for comparison. Results Mean arterial distension was significantly lower in abdominal aortic aneurysm mice between day 0 and day 7 post-onset of disease (p < 0.01) and between day 0 and day 14 (p < 0.001), while no difference was observed in sham control mice. Manual analysis detected a significant decrease (p < 0.05) between day 0 and day 14 only. Atherosclerotic mice showed alterations in arterial distension relating to genetic modification and diet. Arterial distension was significantly lower (p < 0.05) in Ldlr-/- (++/--) mice fed high-fat western diet when compared with both wild type (++/++) mice and Ldlr-/- (++/--) mice fed chow diet. The manual method did not detect a significant difference between these groups. Conclusions Arterial distension can be used as an early marker for the detection of arterial disease in murine models. The semi-automatic analysis method provided increased sensitivity to differences between experimental groups when compared to the manual analysis method.
Collapse
Affiliation(s)
- Justyna Janus
- Preclinical Imaging Facility, Core Biotechnology Services, University of Leicester, Leicester, UK
| | - Baris Kanber
- Translational Imaging Group, Department of Medical Physics and Bioengineering, University College London, London, UK
| | | | - Charlotte Beynon
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Kumar V Ramnarine
- Department of Medical Physics, University Hospitals of Leicester Trust, Leicester, UK
| | - David G Lambert
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Emma J Stringer
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Michael E Kelly
- Preclinical Imaging Facility, Core Biotechnology Services, University of Leicester, Leicester, UK
| |
Collapse
|
30
|
Makinde HM, Just TB, Cuda CM, Bertolino N, Procissi D, Schwulst SJ. Monocyte depletion attenuates the development of posttraumatic hydrocephalus and preserves white matter integrity after traumatic brain injury. PLoS One 2018; 13:e0202722. [PMID: 30383765 PMCID: PMC6211627 DOI: 10.1371/journal.pone.0202722] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/17/2018] [Indexed: 01/06/2023] Open
Abstract
Monocytes are amongst the first cells recruited into the brain after traumatic brain injury (TBI). We have shown monocyte depletion 24 hours prior to TBI reduces brain edema, decreases neutrophil infiltration and improves behavioral outcomes. Additionally, both lesion and ventricle size correlate with poor neurologic outcome after TBI. Therefore, we aimed to determine the association between monocyte infiltration, lesion size, and ventricle volume. We hypothesized that monocyte depletion would attenuate lesion size, decrease ventricle enlargement, and preserve white matter in mice after TBI. C57BL/6 mice underwent pan monocyte depletion via intravenous injection of liposome-encapsulated clodronate. Control mice were injected with liposome-encapsulated PBS. TBI was induced via an open-head, controlled cortical impact. Mice were imaged using magnetic resonance imaging (MRI) at 1, 7, and 14 days post-injury to evaluate progression of lesion and to detect morphological changes associated with injury (3D T1-weighted MRI) including regional alterations in white matter patterns (multi-direction diffusion MRI). Lesion size and ventricle volume were measured using semi-automatic segmentation and active contour methods with the software program ITK-SNAP. Data was analyzed with the statistical software program PRISM. No significant effect of monocyte depletion on lesion size was detected using MRI following TBI (p = 0.4). However, progressive ventricle enlargement following TBI was observed to be attenuated in the monocyte-depleted cohort (5.3 ± 0.9mm3) as compared to the sham-depleted cohort (13.2 ± 3.1mm3; p = 0.02). Global white matter integrity and regional patterns were evaluated and quantified for each mouse after extracting fractional anisotropy maps from the multi-direction diffusion-MRI data using Siemens Syngo DTI analysis package. Fractional anisotropy (FA) values were preserved in the monocyte-depleted cohort (123.0 ± 4.4mm3) as compared to sham-depleted mice (94.9 ± 4.6mm3; p = 0.025) by 14 days post-TBI. All TBI mice exhibited FA values lower than those from a representative naïve control group with intact white matter tracts and FA~200 mm3). The MRI derived assessment of injury progression suggests that monocyte depletion at the time of injury may be a novel therapeutic strategy in the treatment of TBI. Furthermore, non-invasive longitudinal imaging allows for the evaluation of both TBI progression as well as therapeutic response over the course of injury.
Collapse
Affiliation(s)
- Hadijat M. Makinde
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University, Chicago, Illinois, United States of America
| | - Talia B. Just
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University, Chicago, Illinois, United States of America
| | - Carla M. Cuda
- Department of Medicine, Division of Rheumatology, Northwestern University, Chicago, Illinois, United States of America
| | - Nicola Bertolino
- Department of Radiology, Northwestern University, Chicago, Illinois, United States of America
| | - Daniele Procissi
- Department of Radiology, Northwestern University, Chicago, Illinois, United States of America
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States of America
| | - Steven J. Schwulst
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
31
|
Owen R, Reilly GC. In vitro Models of Bone Remodelling and Associated Disorders. Front Bioeng Biotechnol 2018; 6:134. [PMID: 30364287 PMCID: PMC6193121 DOI: 10.3389/fbioe.2018.00134] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/07/2018] [Indexed: 01/02/2023] Open
Abstract
Disruption of bone remodelling by diseases such as osteoporosis results in an imbalance between bone formation by osteoblasts and resorption by osteoclasts. Research into these metabolic bone disorders is primarily performed in vivo; however, in the last decade there has been increased interest in generating in vitro models that can reduce or replace our reliance on animal testing. With recent advances in biomaterials and tissue engineering the feasibility of laboratory-based alternatives is growing; however, to date there are no established in vitro models of bone remodelling. In vivo, remodelling is performed by organised packets of osteoblasts and osteoclasts called bone multicellular units (BMUs). The key determinant of whether osteoclasts form and remodelling occurs is the ratio between RANKL, a cytokine which stimulates osteoclastogenesis, and OPG, its inhibitor. This review initially details the different circumstances, conditions, and factors which have been found to modulate the RANKL:OPG ratio, and fundamental factors to be considered if a robust in vitro model is to be developed. Following this, an examination of what has been achieved thus far in replicating remodelling in vitro using three-dimensional co-cultures is performed, before overviewing how such systems are already being utilised in the study of associated diseases, such as metastatic cancer and dental disorders. Finally, a discussion of the most important considerations to be incorporated going forward is presented. This details the need for the use of cells capable of endogenously producing the required cytokines, application of mechanical stimulation, and the presence of appropriate hormones in order to produce a robust model of bone remodelling.
Collapse
Affiliation(s)
| | - Gwendolen C. Reilly
- Department of Materials Science and Engineering, University of Sheffield, Insigneo Institute for in silico Medicine, Sheffield, United Kingdom
| |
Collapse
|
32
|
Patient-Derived Xenograft Models for Endometrial Cancer Research. Int J Mol Sci 2018; 19:ijms19082431. [PMID: 30126113 PMCID: PMC6121639 DOI: 10.3390/ijms19082431] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/03/2018] [Accepted: 08/13/2018] [Indexed: 12/21/2022] Open
Abstract
Endometrial cancer (EC) is the most common malignancy of the genital tract among women in developed countries. Recently, a molecular classification of EC has been performed providing a system that, in conjunction with histological observations, reliably improves EC classification and enhances patient management. Patient-derived xenograft models (PDX) represent nowadays a promising tool for translational research, since they closely resemble patient tumour features and retain molecular and histological features. In EC, PDX models have already been used, mainly as an individualized approach to evaluate the efficacy of novel therapies and to identify treatment-response biomarkers; however, their uses in more global or holistic approaches are still missing. As a collaborative effort within the ENITEC network, here we describe one of the most extensive EC PDX cohorts developed from primary tumour and metastasis covering all EC subtypes. Our models are histologically and molecularly characterized and represent an excellent reservoir of EC tumour samples for translational research. This review compiles the information on current methods of EC PDX generation and their utility and provides new perspectives for the exploitation of these valuable tools in order to increase the success ratio for translating results to clinical practice.
Collapse
|
33
|
From bed to bench: How in silico medicine can help ageing research. Mech Ageing Dev 2018; 177:103-108. [PMID: 30005915 DOI: 10.1016/j.mad.2018.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/19/2018] [Accepted: 07/09/2018] [Indexed: 02/06/2023]
Abstract
Driven by the raising ethical concerns surrounding animal experimentation, there is a growing interest for non-animal methods, in vitro or in silico technologies that can be used to reduce, refine, and replace animal experimentation. In addition, animal experimentation is being critically revised in regard to its ability to predict clinical outcomes. In this manuscript we describe an initial exploration where a set of in vivo imaging based subject-specific technologies originally developed to predict the risk of femoral strength and hip fracture in osteoporotic patients, were adapted to assess the efficacy of bone drugs pre-clinically on mice. The CT2S technology we developed generates subject-specific models based on Computed Tomography that can separate fractured and non-fractured patients with an accuracy of 82%. When used in mouse experiments the use of in vivo imaging and modelling was found to improve the reproducibility of Bone Mineral Content measurements to a point where up to 63% less mice would be required to achieve the same statistical power of a conventional cross-sectional study. We also speculate about a possible approach where animal-specific and patient-specific models could be used to better translate the observation made on animal models into predictions of response in humans.
Collapse
|
34
|
Oliviero S, Giorgi M, Dall'Ara E. Validation of finite element models of the mouse tibia using digital volume correlation. J Mech Behav Biomed Mater 2018; 86:172-184. [PMID: 29986291 DOI: 10.1016/j.jmbbm.2018.06.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/29/2018] [Accepted: 06/15/2018] [Indexed: 01/30/2023]
Abstract
The mouse tibia is a common site to investigate bone adaptation. Micro-Finite Element (microFE) models based on micro-Computed Tomography (microCT) images can estimate bone mechanical properties non-invasively but their outputs need to be validated with experiments. Digital Volume Correlation (DVC) can provide experimental measurements of displacements over the whole bone volume. In this study we applied DVC to validate the local predictions of microFE models of the mouse tibia in compression. Six mouse tibiae were stepwise compressed within a microCT system. MicroCT images were acquired in four configurations with applied compression of 0.5 N (preload), 6.5 N, 13.0 N and 19.5 N. Failure load was measured after the last scan. A global DVC algorithm was applied to the microCT images in order to obtain the displacement field over the bone volume. Homogeneous, isotropic linear hexahedral microFE models were generated from the images collected in the preload configuration with boundary conditions interpolated from the DVC displacements at the extremities of the tibia. Experimental displacements from DVC and numerical predictions were compared at corresponding locations in the middle of the bone. Stiffness and strength were also estimated from each model and compared with the experimental measurements. The magnitude of the displacement vectors predicted by microFE models was highly correlated with experimental measurements (R2 >0.82). Higher but still reasonable errors were found for the Cartesian components. The models tended to overestimate local displacements in the longitudinal direction (R2 = 0.69-0.90, slope of the regression line=0.50-0.97). Errors in the prediction of structural mechanical properties were 14% ± 11% for stiffness and 9% ± 9% for strength. In conclusion, the DVC approach has been applied to the validation of microFE models of the mouse tibia. The predictions of the models for both structural and local properties have been found reasonable for most preclinical applications.
Collapse
Affiliation(s)
- S Oliviero
- Department of Oncology and Metabolism and INSIGNEO Institute for in Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, S13JD Sheffield, UK.
| | - M Giorgi
- Department of Oncology and Metabolism and INSIGNEO Institute for in Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, S13JD Sheffield, UK.
| | - E Dall'Ara
- Department of Oncology and Metabolism and INSIGNEO Institute for in Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, S13JD Sheffield, UK.
| |
Collapse
|
35
|
Gordon S, Dolgopyat I, Kahn I, Riklin Raviv T. Multidimensional co-segmentation of longitudinal brain MRI ensembles in the presence of a neurodegenerative process. Neuroimage 2018; 178:346-369. [PMID: 29723637 DOI: 10.1016/j.neuroimage.2018.04.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/14/2018] [Accepted: 04/18/2018] [Indexed: 11/25/2022] Open
Abstract
MRI Segmentation of a pathological brain poses a significant challenge, as the available anatomical priors that provide top-down information to aid segmentation are inadequate in the presence of abnormalities. This problem is further complicated for longitudinal data capturing impaired brain development or neurodegenerative conditions, since the dynamic of brain atrophies has to be considered as well. For these cases, the absence of compatible annotated training examples renders the commonly used multi-atlas or machine-learning approaches impractical. We present a novel segmentation approach that accounts for the lack of labeled data via multi-region multi-subject co-segmentation (MMCoSeg) of longitudinal MRI sequences. The underlying, unknown anatomy is learned throughout an iterative process, in which the segmentation of a region is supported both by the segmentation of the neighboring regions, which share common boundaries, and by the segmentation of corresponding regions, in the other jointly segmented images. A 4D multi-region atlas that models the spatio-temporal deformations and can be adapted to different subjects undergoing similar degeneration processes is reconstructed concurrently. An inducible mouse model of p25 accumulation (the CK-p25 mouse) that displays key pathological hallmarks of Alzheimer disease (AD) is used as a gold-standard to test the proposed algorithm by providing a conditional control of rapid neurodegeneration. Applying the MMCoSeg to a cohort of CK-p25 mice and littermate controls yields promising segmentation results that demonstrate high compatibility with expertise manual annotations. An extensive comparative analysis with respect to current well-established, atlas-based segmentation methods highlights the advantage of the proposed approach, which provides accurate segmentation of longitudinal brain MRIs in pathological conditions, where only very few annotated examples are available.
Collapse
Affiliation(s)
- Shiri Gordon
- Electrical and Computer Engineering Department and the Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Irit Dolgopyat
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Itamar Kahn
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Tammy Riklin Raviv
- Electrical and Computer Engineering Department and the Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
36
|
Propagation-based phase-contrast synchrotron imaging of aortic dissection in mice: from individual elastic lamella to 3D analysis. Sci Rep 2018; 8:2223. [PMID: 29396472 PMCID: PMC5797148 DOI: 10.1038/s41598-018-20673-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/23/2018] [Indexed: 11/09/2022] Open
Abstract
In order to show the advantage and potential of propagation-based phase-contrast synchrotron imaging in vascular pathology research, we analyzed aortic medial ruptures in BAPN/AngII-infused mice, a mouse model for aortic dissection. Ascending and thoraco-abdominal samples from n = 3 control animals and n = 10 BAPN/AngII-infused mice (after 3, 7 and 14 days of infusion, total of 24 samples) were scanned. A steep increase in the number of ruptures was already noted after 3 days of BAPN/AngII-infusion. The largest ruptures were found at the latest time points. 133 ruptures affected only the first lamella while 135 ruptures affected multiple layers. Medial ruptures through all lamellar layers, leading to false channel formation and intramural hematoma, occurred only in the thoraco-abdominal aorta and interlamellar hematoma formation in the ascending aorta could be directly related to ruptures of the innermost lamellae. The advantages of this technique are (i) ultra-high resolution that allows to visualize the individual elastic lamellae in the aorta; (ii) quantitative and qualitative analysis of medial ruptures; (iii) 3D analysis of the complete aorta; (iv) high contrast for qualitative information extraction, reducing the need for histology coupes; (v) earlier detection of (micro-) ruptures.
Collapse
|
37
|
Effect of integration time on the morphometric, densitometric and mechanical properties of the mouse tibia. J Biomech 2017; 65:203-211. [DOI: 10.1016/j.jbiomech.2017.10.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/22/2017] [Accepted: 10/27/2017] [Indexed: 12/14/2022]
|
38
|
Lu Y, Boudiffa M, Dall'Ara E, Liu Y, Bellantuono I, Viceconti M. Longitudinal effects of Parathyroid Hormone treatment on morphological, densitometric and mechanical properties of mouse tibia. J Mech Behav Biomed Mater 2017; 75:244-251. [PMID: 28756285 DOI: 10.1016/j.jmbbm.2017.07.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/15/2017] [Accepted: 07/24/2017] [Indexed: 01/19/2023]
Abstract
The use of Parathyroid Hormone (PTH) as bone anabolic is limited due to cost-benefit assessments. Preclinical studies evaluating the effects of PTH on bone have reported variable and often contradictory results. Here, we have applied a new approach using a combination of in-vivo longitudinal µCT, image processing techniques and finite element models to monitor early local changes in the whole tibia (divided in 40 compartments) and mechanical properties of female C57BL/6J mice treated with PTH 1-34, compared to controls. Compared with standard 3D bone morphometric analysis, our new approach allowed detection of much smaller and localised changes in bone mineral content (BMC) at very early time points (1 week vs 3 weeks with standard methods) and showed that changes do not occur uniformly over time and across the anatomical space. Indeed, in the PTH treated mice, significant changes in BMC were observed in the medial and posterior sectors of the proximal tibia, a week after treatment, and in the medial sector of the tibia midshaft region a week later (p < 0.05). By the third week, two thirds of the regions showed significantly higher values of BMC (p < 0.05). The effect of PTH on bone regional volume is similar to that on BMC, but there is almost no effect of PTH on bone tissue mineral density. The differences in estimated mechanical properties became significant after three weeks of treatment (p < 0.05). These results provide the first evidence of an early and localised PTH effect on murine bone, and show that our novel partitioning approach, compared to the standard evaluation protocol, allows a more precise quantification of bone changes following treatment, which would facilitate preclinical testing of novel mono- and/or combination therapies throughout the bone.
Collapse
Affiliation(s)
- Yongtao Lu
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China; Department of Mechanical Engineering, The University of Sheffield, Sheffield, UK; Insigneo Institute for in Silico Medicine, The University of Sheffield, Sheffield, UK
| | - Maya Boudiffa
- MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Enrico Dall'Ara
- Insigneo Institute for in Silico Medicine, The University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK.
| | - Yue Liu
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Ilaria Bellantuono
- Insigneo Institute for in Silico Medicine, The University of Sheffield, Sheffield, UK; MRC Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Marco Viceconti
- Department of Mechanical Engineering, The University of Sheffield, Sheffield, UK; Insigneo Institute for in Silico Medicine, The University of Sheffield, Sheffield, UK
| |
Collapse
|
39
|
Wu YL, Lo CW. Diverse application of MRI for mouse phenotyping. Birth Defects Res 2017; 109:758-770. [PMID: 28544650 DOI: 10.1002/bdr2.1051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/15/2017] [Indexed: 02/01/2023]
Abstract
Small animal models, particularly mouse models, of human diseases are becoming an indispensable tool for biomedical research. Studies in animal models have provided important insights into the etiology of diseases and accelerated the development of therapeutic strategies. Detailed phenotypic characterization is essential, both for the development of such animal models and mechanistic studies into disease pathogenesis and testing the efficacy of experimental therapeutics. MRI is a versatile and noninvasive imaging modality with excellent penetration depth, tissue coverage, and soft tissue contrast. MRI, being a multi-modal imaging modality, together with proven imaging protocols and availability of good contrast agents, is ideally suited for phenotyping mutant mouse models. Here we describe the applications of MRI for phenotyping structural birth defects involving the brain, heart, and kidney in mice. The versatility of MRI and its ease of use are well suited to meet the rapidly increasing demands for mouse phenotyping in the coming age of functional genomics. Birth Defects Research 109:758-770, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yijen L Wu
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|