Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
1.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38657972

ABSTRACT

Advances in fluorescence microscopy and tissue-clearing have revolutionised 3D imaging of fluorescently labelled tissues, organs and embryos. However, the complexity and high cost of existing software and computing solutions limit their widespread adoption, especially by researchers with limited resources. Here, we present Acto3D, an open-source software, designed to streamline the generation and analysis of high-resolution 3D images of targets labelled with multiple fluorescent probes. Acto3D provides an intuitive interface for easy 3D data import and visualisation. Although Acto3D offers straightforward 3D viewing, it performs all computations explicitly, giving users detailed control over the displayed images. Leveraging an integrated graphics processing unit, Acto3D deploys all pixel data to system memory, reducing visualisation latency. This approach facilitates accurate image reconstruction and efficient data processing in 3D, eliminating the need for expensive high-performance computers and dedicated graphics processing units. We have also introduced a method for efficiently extracting lumen structures in 3D. We have validated Acto3D by imaging mouse embryonic structures and by performing 3D reconstruction of pharyngeal arch arteries while preserving fluorescence information. Acto3D is a cost-effective and efficient platform for biological research.


Subject(s)
Imaging, Three-Dimensional , Software , Imaging, Three-Dimensional/methods , Animals , Mice , Microscopy, Fluorescence/methods , Optical Imaging/methods , Image Processing, Computer-Assisted/methods , Embryo, Mammalian/diagnostic imaging
2.
Phys Med Biol ; 69(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38631365

ABSTRACT

Objective.To report on a micro computed tomography (micro-CT) system capable of x-ray phase contrast imaging and of increasing spatial resolution at constant magnification.Approach.The micro-CT system implements the edge illumination (EI) method, which relies on two absorbing masks with periodically spaced transmitting apertures in the beam path; these split the beam into an array of beamlets and provide sensitivity to the beamlets' directionality, i.e. refraction. In EI, spatial resolution depends on the width of the beamlets rather than on the source/detector point spread function (PSF), meaning that resolution can be increased by decreasing the mask apertures, without changing the source/detector PSF or the magnification.Main results.We have designed a dedicated mask featuring multiple bands with differently sized apertures and used this to demonstrate that resolution is a tuneable parameter in our system, by showing that increasingly small apertures deliver increasingly detailed images. Phase contrast images of a bar pattern-based resolution phantom and a biological sample (a mouse embryo) were obtained at multiple resolutions.Significance.The new micro-CT system could find application in areas where phase contrast is already known to provide superior image quality, while the added tuneable resolution functionality could enable more sophisticated analyses in these applications, e.g. by scanning samples at multiple scales.


Subject(s)
Phantoms, Imaging , X-Ray Microtomography , X-Ray Microtomography/instrumentation , Mice , Animals , Embryo, Mammalian/diagnostic imaging , Image Processing, Computer-Assisted/methods
3.
STAR Protoc ; 4(4): 102603, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37742180

ABSTRACT

Tissue autofluorescence poses significant challenges for RNA and protein analysis using fluorescence-based techniques. Here, we present a protocol that combines oxidation-mediated autofluorescence reduction with detergent-based tissue permeabilization for whole-mount RNA-fluorescence in situ hybridization (FISH) on mouse embryonic limb buds. We describe the steps for embryo collection, fixation, photochemical bleaching, permeabilization, and RNA-FISH, followed by optical clearing of RNA-FISH and immunofluorescence samples for imaging. The protocol alleviates the need for digital image post-processing to remove autofluorescence and is applicable to other tissues, organs, and vertebrate embryos.


Subject(s)
Embryo, Mammalian , RNA , Animals , Mice , RNA/metabolism , In Situ Hybridization, Fluorescence/methods , Embryo, Mammalian/diagnostic imaging , Embryo, Mammalian/metabolism , Fluorescent Antibody Technique
5.
Ultrasound Med Biol ; 49(1): 356-367, 2023 01.
Article in English | MEDLINE | ID: mdl-36283941

ABSTRACT

Large-scale international efforts to generate and analyze loss-of-function mutations in each of the approximately 20,000 protein-encoding gene mutations are ongoing using the "knockout" mouse as a model organism. Because one-third of gene knockouts are expected to result in embryonic lethality, it is important to develop non-invasive in utero imaging methods to detect and monitor mutant phenotypes in mouse embryos. We describe the utility of 3-D high-frequency (40-MHz) ultrasound (HFU) for longitudinal in utero imaging of mouse embryos between embryonic days (E) 11.5 and E14.5, which represent critical stages of brain and organ development. Engrailed-1 knockout (En1-ko) mouse embryos and their normal control littermates were imaged with HFU in 3-D, enabling visualization of morphological phenotypes in the developing brains, limbs and heads of the En1-ko embryos. Recently developed deep learning approaches were used to automatically segment the embryonic brain ventricles and bodies from the 3-D HFU images, allowing quantitative volumetric analyses of the En1-ko brain phenotypes. Taken together, these results show great promise for the application of longitudinal 3-D HFU to analyze knockout mouse embryos in utero.


Subject(s)
Brain , Imaging, Three-Dimensional , Animals , Mice , Mice, Knockout , Ultrasonography , Imaging, Three-Dimensional/methods , Phenotype , Embryo, Mammalian/diagnostic imaging
6.
J Vis Exp ; (185)2022 07 12.
Article in English | MEDLINE | ID: mdl-35913197

ABSTRACT

The first steps of heart development imply drastic changes in cell behavior and differentiation. While analysis of fixed embryos allows studying in detail specific developmental stages in a still snapshot, live imaging captures dynamic morphogenetic events, such as cell migration, shape changes, and differentiation, by imaging the embryo as it develops. This complements fixed analysis and expands the understanding of how organs develop during embryogenesis. Despite its advantages, live imaging is rarely used in mouse models because of its technical challenges. Early mouse embryos are sensitive when cultured ex vivo and require efficient handling. To facilitate a broader use of live imaging in mouse developmental research, this paper presents a detailed protocol for two-photon live microscopy that allows long-term acquisition in mouse embryos. In addition to the protocol, tips are provided on embryo handling and culture optimization. This will help understand key events in early mouse organogenesis, enhancing the understanding of cardiovascular progenitor biology.


Subject(s)
Embryo, Mammalian , Embryonic Development , Animals , Embryo, Mammalian/diagnostic imaging , Heart/diagnostic imaging , Mice , Microscopy/methods , Organogenesis
7.
Reprod Biol Endocrinol ; 20(1): 53, 2022 Mar 19.
Article in English | MEDLINE | ID: mdl-35305653

ABSTRACT

BACKGROUND: Despite all research efforts during this era of novel time-lapse morphokinetic parameters, a morphological grading system is still routinely being used for embryo selection at the blastocyst stage. The blastocyst expansion grade, as evaluated during morphological assessment, is associated with clinical pregnancy. However, this assessment is performed without taking the dynamics of blastocoel expansion into account. Here, we studied the dynamics of blastocoel expansion by comparing longitudinal blastocoel surface measurements using time-lapse embryo culture. Our aim was to first assess if this is impacted by fertilization method and second, to study if an association exists between these measurement and ongoing pregnancy. METHODS: This was a retrospective cohort study including 225 couples undergoing 225 cycles of in vitro fertilization (IVF) treatment with time-lapse embryo culture. The fertilization method was either conventional IVF, intracytoplasmic sperm injection (ICSI) with ejaculated sperm or ICSI with sperm derived from testicular sperm extraction (TESE-ICSI). This resulted in 289 IVF embryos, 218 ICSI embryos and 259 TESE-ICSI embryos that reached at least the full blastocyst stage. Blastocoel surface measurements were performed on time-lapse images every hour, starting from full blastocyst formation (tB). Linear mixed model analysis was performed to study the association between blastocoel expansion, the calculated expansion rate (µm2/hour) and both fertilization method and ongoing pregnancy. RESULTS: The blastocoel of both ICSI embryos and TESE-ICSI embryos was significantly smaller than the blastocoel of IVF embryos (beta -1121.6 µm2; 95% CI: -1606.1 to -637.1, beta -646.8 µm2; 95% CI: -1118.7 to 174.8, respectively). Still, the blastocoel of transferred embryos resulting in an ongoing pregnancy was significantly larger (beta 795.4 µm2; 95% CI: 15.4 to 1575.4) and expanded significantly faster (beta 100.9 µm2/hour; 95% CI: 5.7 to 196.2) than the blastocoel of transferred embryos that did not, regardless of the fertilization method. CONCLUSION: Longitudinal blastocyst surface measurements and expansion rates are promising non-invasive quantitative markers that can aid embryo selection for transfer and cryopreservation. TRIAL REGISTRATION: Our study is a retrospective observational study, therefore trial registration is not applicable.


Subject(s)
Blastocyst/physiology , Embryo, Mammalian/diagnostic imaging , Embryonic Development/physiology , Fertilization in Vitro/methods , Time-Lapse Imaging , Adult , Blastocyst/cytology , Cell Proliferation , Cell Shape , Cells, Cultured , Cleavage Stage, Ovum/cytology , Cleavage Stage, Ovum/physiology , Cohort Studies , Embryo Culture Techniques/methods , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Female , Fertilization/physiology , Humans , Longitudinal Studies , Male , Netherlands , Pregnancy/physiology , Retrospective Studies , Sperm Injections, Intracytoplasmic/methods , Surface Properties
8.
Environ Health Perspect ; 130(2): 27002, 2022 02.
Article in English | MEDLINE | ID: mdl-35108087

ABSTRACT

BACKGROUND: Severe environmental pollution and contaminants left in the environment due to the abuse of chemicals, such as methylmercury, are associated with an increasing number of embryonic disorders. Ultrasound imaging has been widely used to investigate embryonic development malformation and dysorganoplasia in both research and clinics. However, this technique is limited by its low contrast and lacking functional parameters such as the ability to measure blood oxygen saturation (SaO2) and hemoglobin content (HbT) in tissues, measures that could be early vital indicators for embryonic development abnormality. Herein, we proposed combining two highly complementary techniques into a photoacoustic-ultrasound (PA-US) dual-modality imaging approach to noninvasively detect early mouse embryo abnormalities caused by methylmercury chloride (MMC) in real time. OBJECTIVES: This study aimed to assess the use of PA-US dual-modality imaging for noninvasive detection of embryonic toxicity at different stages of growth following prenatal MMC exposure. Additionally, we compared the PA-US imagining results to traditional histological methods to determine whether this noninvasive method could detect early developmental defects in utero. METHODS: Different dosages of MMC were administrated to pregnant mice by gavage to establish models of different levels of embryonic malformation. Ultrasound, photoacoustic signal intensity (PSI), blood oxygen saturation (SaO2), and hemoglobin content (HbT) were quantified in all experimental groups. Furthermore, the embryos were sectioned and examined for pathological changes. RESULTS: Using PA-US imaging, we detected differences in PSI, SaO2, HbT, and heart volume at embryonic day (E)14.5 and E11.5 for low and high dosages of MMC, respectively. More important, our results showed that differences between control and treated embryos identified by in utero PA-US imaging were consistent with those identified in ex vivo embryos using histological methods. CONCLUSION: Our results suggest that noninvasive dual-modality PA-US is a promising strategy for detecting developmental toxicology in the uterus. Overall, this study presents a new approach for detecting embryonic toxicities, which could be crucial in clinics when diagnosing aberrant embryonic development. https://doi.org/10.1289/EHP8907.


Subject(s)
Methylmercury Compounds , Prenatal Exposure Delayed Effects , Animals , Embryo, Mammalian/diagnostic imaging , Female , Mammals , Methylmercury Compounds/toxicity , Mice , Pregnancy , Ultrasonography
9.
Mol Cell Endocrinol ; 540: 111508, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34800604

ABSTRACT

The uterine environment must provide sufficient endocrine conditions and nutrients for pregnancy maintenance and conceptus survival. The objective of this study was to determine the effects of preovulatory estradiol and conceptus presence on uterine transcripts and uterine luminal fluid (ULF) proteins. Beef cows/heifers were synchronized and artificially inseminated (d 0). Uteri were flushed (d 16); conceptuses and endometrial biopsies were collected. Total cellular RNA was extracted from endometrium for RNA sequencing and RT-PCR validation. There were two independent ULF pools made for each of the following groups: highE2/conceptus, highE2/noconceptus, lowE2/conceptus, and lowE2/noconceptus that were analyzed using the 2D LC-MS/MS based iTRAQ method. There were 64 differentially expressed genes (DEGs) and 77 differentially expressed proteins (DEPs) in common among the highE2/conceptus vs highE2/noconceptus and lowE2/conceptus vs lowE2/noconceptus groups. In summary, the interaction between preovulatory estradiol and the conceptus induces the expression of genes, proteins, and pathways necessary for pregnancy.


Subject(s)
Cattle , Embryo, Mammalian/physiology , Perception/physiology , Pregnancy, Animal , Uterus/metabolism , Animals , Cattle/genetics , Cattle/physiology , Embryo, Mammalian/diagnostic imaging , Embryonic Development/physiology , Endometrium/metabolism , Estradiol/pharmacology , Female , Follicular Phase/drug effects , Follicular Phase/physiology , Gene Expression Regulation , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Pregnancy, Animal/genetics , Pregnancy, Animal/psychology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ultrasonography, Prenatal/veterinary , Uterus/diagnostic imaging
10.
STAR Protoc ; 2(4): 100980, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34927090

ABSTRACT

Cardiac function and morphology by mouse fetal echocardiography can be assessed by scanning the uterus extracted from the abdominal cavity (trans-uterine ultrasound) or the womb (trans-abdominal ultrasound). Advantages of trans-abdominal ultrasound include (1) non-invasive longitudinal analysis at different stages, reducing animal use; and (2) maintenance of natural environment, diminishing perturbations on functional parameters, which are more frequent in trans-uterine conditions. Here we describe both approaches, explaining how to identify congenital cardiac defects and defining the correlation between echocardiography findings and histological analysis. For complete details on the use and execution of this protocol, please refer to (Menendez-Montes et al., 2016) and (Menendez-Montes et al., 2021).


Subject(s)
Echocardiography/methods , Embryo, Mammalian/diagnostic imaging , Fetal Heart/diagnostic imaging , Heart Defects, Congenital/diagnostic imaging , Image Interpretation, Computer-Assisted/methods , Animals , Female , Male , Mice , Pregnancy , Ultrasonography, Prenatal/methods
11.
Reprod Biomed Online ; 43(5): 843-852, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34521598

ABSTRACT

RESEARCH QUESTION: Can artificial intelligence (AI) improve the prediction of live births based on embryo images? DESIGN: The AI system was created by using the Attention Branch Network associated with deep learning to predict the probability of live birth from 141,444 images recorded by time-lapse imaging of 470 transferred embryos, of which 91 resulted in live birth and 379 resulted in non-live birth that included implantation failure, biochemical pregnancy and clinical miscarriage. The possibility that the calculated confidence scores of each embryo and the focused areas visualized in each embryo image can help predict subsequent live birth was examined. RESULTS: The AI system for the first time successfully visualized embryo features in focused areas that had potential to distinguish between live and non-live births. No visual feature of embryos were visualized that were associated with live or non-live births, although there were many images in which high-focused areas existed around the zona pellucida. When a cut-off level for the confidence score was set at 0.341, the live birth rate was significantly greater for embryos with a score higher than the cut-off level than for those with a score lower than the cut-off level (P < 0.001). In addition, the live birth rate of embryos with good morphological quality and confidence scores higher than 0.341 was 41.1%. CONCLUSIONS: The authors have created an AI system with a confidence score that is useful for non-invasive selection of embryos that could result in live birth. Further study is necessary to improve selection accuracy.


Subject(s)
Artificial Intelligence , Embryo, Mammalian/diagnostic imaging , Fertilization in Vitro , Live Birth , Time-Lapse Imaging , Adult , Cohort Studies , Embryo Transfer , Embryo, Mammalian/physiology , Female , Humans , Pregnancy , Retrospective Studies
12.
Cell Rep ; 36(2): 109382, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260920

ABSTRACT

Developmental biologists have always relied on imaging to shed light on dynamic cellular events. However, processes such as mammalian fertilization and embryogenesis are generally inaccessible for direct imaging. In consequence, how the oviduct (fallopian tube) facilitates the transport of gametes and preimplantation embryos continues to be unanswered. Here we present a combination of intravital window and optical coherence tomography for dynamic, volumetric, in vivo imaging of oocytes and embryos as they are transported through the mouse oviduct. We observed location-dependent circling, oscillating, and long-distance bi-directional movements of oocytes and embryos that suggest regulatory mechanisms driving transport and question established views in the field. This in vivo imaging approach can be combined with a variety of genetic and pharmacological manipulations for live functional analysis, bringing the potential to investigate reproductive physiology in its native state.


Subject(s)
Embryo, Mammalian/diagnostic imaging , Imaging, Three-Dimensional , Oocytes/cytology , Oviducts/diagnostic imaging , Animals , Blastocyst , Cumulus Cells/cytology , Female , Mice , Models, Biological , Movement
13.
Methods Mol Biol ; 2319: 93-104, 2021.
Article in English | MEDLINE | ID: mdl-34331247

ABSTRACT

Lightsheet microscopy is a form of fluorescence microscopy that can be used to visualize specimen with high resolution, a large depth-of-field, and minimal photodamage and photobleaching as compared to traditional confocal microscopy. As this technology becomes much more readily available, it will be useful in revealing new findings in the cardiovascular development field that may be hidden or difficult to image. In this manuscript, we describe an approach for mounting and culturing postimplantation mouse embryos to visualize blood vessel development with a lightsheet microscope.


Subject(s)
Angiography/methods , Blood Vessels/diagnostic imaging , Culture Techniques/methods , Embryo, Mammalian/diagnostic imaging , Embryonic Development , Microscopy, Fluorescence/methods , Neovascularization, Physiologic , Animals , Blood Vessels/growth & development , Blood Vessels/metabolism , Culture Media/chemistry , Dissection/methods , Embryo, Mammalian/blood supply , Mice , Mice, Transgenic , Microscopy, Confocal
14.
Methods Mol Biol ; 2319: 119-136, 2021.
Article in English | MEDLINE | ID: mdl-34331250

ABSTRACT

We describe a novel, efficient method to identify cis-acting DNA sequences that drive cell-specific gene expression during development. We utilize transfer of Bacterial Artificial Chromosome (BAC) genomic DNAs, modified to contain a reporter gene, into fertilized mouse embryos and placing the injected embryos into pseudopregnant recipient females. The embryos are allowed to develop in utero for defined times after which they are collected for analysis. Using DNAs containing the LacZ reporter gene facilitates the analysis of gene activity through microscopy of intact embryos and subsequent sectioning of the stained embryos. With this technique cis-element activity can be identified and evaluated through further mutational analysis of the injected BAC DNA. This allows the identification of important gene regulatory domains that specify stage-specific gene expression in the developing embryo.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Embryo, Mammalian/diagnostic imaging , Regulatory Sequences, Nucleic Acid/genetics , Animals , Embryo, Mammalian/metabolism , Female , Genes, Reporter/genetics , Mice , Mice, Transgenic , Microinjections/methods , Recombination, Genetic , beta-Galactosidase/genetics
15.
Nat Biomed Eng ; 5(6): 571-585, 2021 06.
Article in English | MEDLINE | ID: mdl-34112997

ABSTRACT

In machine learning for image-based medical diagnostics, supervised convolutional neural networks are typically trained with large and expertly annotated datasets obtained using high-resolution imaging systems. Moreover, the network's performance can degrade substantially when applied to a dataset with a different distribution. Here, we show that adversarial learning can be used to develop high-performing networks trained on unannotated medical images of varying image quality. Specifically, we used low-quality images acquired using inexpensive portable optical systems to train networks for the evaluation of human embryos, the quantification of human sperm morphology and the diagnosis of malarial infections in the blood, and show that the networks performed well across different data distributions. We also show that adversarial learning can be used with unlabelled data from unseen domain-shifted datasets to adapt pretrained supervised networks to new distributions, even when data from the original distribution are not available. Adaptive adversarial networks may expand the use of validated neural-network models for the evaluation of data collected from multiple imaging systems of varying quality without compromising the knowledge stored in the network.


Subject(s)
Image Interpretation, Computer-Assisted/statistics & numerical data , Malaria, Falciparum/diagnostic imaging , Neural Networks, Computer , Spermatozoa/ultrastructure , Supervised Machine Learning , Datasets as Topic , Embryo, Mammalian/diagnostic imaging , Embryo, Mammalian/ultrastructure , Female , Histocytochemistry/methods , Humans , Malaria, Falciparum/parasitology , Male , Microscopy/methods , Plasmodium falciparum/ultrastructure , Time-Lapse Imaging/methods , Time-Lapse Imaging/statistics & numerical data
16.
NMR Biomed ; 34(8): e4536, 2021 08.
Article in English | MEDLINE | ID: mdl-33955062

ABSTRACT

NMR offers the potential to holistically screen hundreds of metabolites and has already proved to be a powerful technique able to provide a global picture of metabolic changes in a wide range of biological systems underlying complex and multifactorial matrixes. This review covers the literature until May 2020 centered on the early prediction of the viability of in vitro developed embryos using several analytical techniques, including NMR. Nowadays, the predominant non-invasive technique for selecting viable embryos is based on morphology, where variables associated with the rate of cleavage and blastocyst formation are evaluated by the embryologist following standardized criteria that are somewhat subjective. This morphological approach is therefore inadequate for the prediction of embryo quality, and several studies have focused on developing new non-invasive methods using molecular approaches based particularly on metabolomics. This review outlines the potential of NMR as one of these non-invasive in vitro methods based on the analysis of spent embryo culture media.


Subject(s)
Culture Media/pharmacology , Embryo Implantation , Embryo, Mammalian/diagnostic imaging , Magnetic Resonance Spectroscopy , Fertilization in Vitro , Humans , Metabolomics , Software
17.
Sci Rep ; 11(1): 5722, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707458

ABSTRACT

Chondrogenesis and angiogenesis drive endochondral ossification. Using the atmospheric scanning electron microscopy (ASEM) without decalcification and dehydration, we directly imaged angiogenesis-driven ossification at different developmental stages shortly after aldehyde fixation, using aqueous radical scavenger glucose solution to preserve water-rich structures. An embryonic day 15.5 mouse femur was fixed and stained with phosphotungstic acid (PTA), and blood vessel penetration into the hypertrophic chondrocyte zone was visualised. We observed a novel envelope between the perichondrium and proliferating chondrocytes, which was lined with spindle-shaped cells that could be borderline chondrocytes. At postnatal day (P)1, trabecular and cortical bone mineralisation was imaged without staining. Additional PTA staining visualised surrounding soft tissues; filamentous connections between osteoblast-like cells and osteocytes in cortical bone were interpreted as the osteocytic lacunar-canalicular system. By P10, resorption pits had formed on the tibial trabecular bone surface. The applicability of ASEM for pathological analysis was addressed using knockout mice of Keap1, an oxidative-stress sensor. In Keap1-/- femurs, we observed impaired calcification and angiogenesis of epiphyseal cartilage, suggesting impaired bone development. Overall, the quick ASEM method we developed revealed mineralisation and new structures in wet bone tissue at EM resolution and can be used to study mineralisation-associated phenomena of any hydrated tissue.


Subject(s)
Atmosphere , Bone and Bones/pathology , Bone and Bones/ultrastructure , Cartilage/ultrastructure , Kelch-Like ECH-Associated Protein 1/deficiency , Microscopy, Electron, Scanning , Osteogenesis , Osteomalacia/pathology , Animals , Bone and Bones/diagnostic imaging , Calcification, Physiologic , Cartilage/diagnostic imaging , Cartilage/pathology , Chondrogenesis , Cortical Bone/diagnostic imaging , Cortical Bone/ultrastructure , Embryo, Mammalian/diagnostic imaging , Femur/diagnostic imaging , Femur/ultrastructure , Imaging, Three-Dimensional , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred C57BL , Osteocytes/metabolism , Phenotype , Tibia/diagnostic imaging , Tibia/ultrastructure
18.
Opt Express ; 29(2): 2049-2064, 2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33726406

ABSTRACT

X-ray phase contrast imaging is a powerful analysis technique for materials science and biomedicine. Here, we report on laboratory grating-based X-ray interferometry employing a microfocus X-ray source and a high Talbot order (35th) asymmetric geometry to achieve high angular sensitivity and high spatial resolution X-ray phase contrast imaging in a compact system (total length <1 m). The detection of very small refractive angles (∼50 nrad) at an interferometer design energy of 19 keV was enabled by combining small period X-ray gratings (1.0, 1.5 and 3.0 µm) and a single-photon counting X-ray detector (75 µm pixel size). The performance of the X-ray interferometer was fully characterized in terms of angular sensitivity and spatial resolution. Finally, the potential of laboratory X-ray phase contrast for biomedical imaging is demonstrated by obtaining high resolution X-ray phase tomographies of a mouse embryo embedded in solid paraffin and a formalin-fixed full-thickness sample of human left ventricle in water with a spatial resolution of 21.5 µm.


Subject(s)
Embryo, Mammalian/diagnostic imaging , Heart Ventricles/diagnostic imaging , Interferometry/instrumentation , Microscopy, Phase-Contrast/instrumentation , Tomography, X-Ray Computed/methods , Animals , Equipment Design , Humans , Image Processing, Computer-Assisted/methods , Mice , Paraffin Embedding
19.
J Vis Exp ; (168)2021 02 28.
Article in English | MEDLINE | ID: mdl-33720141

ABSTRACT

Somitogenesis is a hallmark of vertebrate embryonic development. For years, researchers have been studying this process in a variety of organisms using a wide range of techniques encompassing ex vivo and in vitro approaches. However, most studies still rely on the analysis of two-dimensional (2D) imaging data, which limits proper evaluation of a developmental process like axial extension and somitogenesis involving highly dynamic interactions in a complex 3D space. Here we describe techniques that allow mouse live imaging acquisition, dataset processing, visualization and analysis in 3D and 4D to study the cells (e.g., neuromesodermal progenitors) involved in these developmental processes. We also provide a step-by-step protocol for optical projection tomography and whole-mount immunofluorescence microscopy in mouse embryos (from sample preparation to image acquisition) and show a pipeline that we developed to process and visualize 3D image data. We extend the use of some of these techniques and highlight specific features of different available software (e.g., Fiji/ImageJ, Drishti, Amira and Imaris) that can be used to improve our current understanding of axial extension and somite formation (e.g., 3D reconstructions). Altogether, the techniques here described emphasize the importance of 3D data visualization and analysis in developmental biology, and might help other researchers to better address 3D and 4D image data in the context of vertebrate axial extension and segmentation. Finally, the work also employs novel tools to facilitate teaching vertebrate embryonic development.


Subject(s)
Body Patterning , Imaging, Three-Dimensional/methods , Vertebrates/anatomy & histology , Vertebrates/embryology , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/diagnostic imaging , Embryonic Development , Fluorescent Antibody Technique , Mice, Knockout , Snail Family Transcription Factors/deficiency , Snail Family Transcription Factors/metabolism , Software , Time Factors , Tissue Fixation , Tomography, Optical
20.
Commun Biol ; 4(1): 190, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33580156

ABSTRACT

The development of the mammalian gut was first described more than a century ago. Since then, it has been believed that a series of highly orchestrated developmental processes occur before the intestine achieves its final formation. The key steps include the formation of the umbilicus, the so-called "physiological herniation" of the midgut into the umbilical cord, an intestinal "rotation", and the "return of the gut" into the abdominal cavity. However, this sequence of events is predominantly based on histological sections of dissected embryos, a 2D technique with methodological limitations. For a better understanding of spatial relationships in the embryo, we utilized microcomputed tomography (µCT), a nondestructive 3D imaging method. Here, we show the detailed processes and mechanisms of intestinal development in rat embryos, including the development of the umbilicus, the formation of loops inside the umbilical coelom, and the subsequent shift of these loops into the abdominal cavity. Our 3D datasets of developing intestines will substantially advance the understanding of normal mammalian midgut embryology and offer new possibilities to reveal unknown mechanisms in the pathogenesis of congenital disorders.


Subject(s)
Embryo, Mammalian/diagnostic imaging , Intestines/diagnostic imaging , X-Ray Microtomography , Animals , Female , Gestational Age , Imaging, Three-Dimensional , Intestines/embryology , Morphogenesis , Predictive Value of Tests , Pregnancy , Radiographic Image Interpretation, Computer-Assisted , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...