1
|
McGlade EA, Mao J, Stephens KK, Rose CN, Lydon JP, Winuthayanon W. Loss of progesterone receptor in smooth muscle cells has no impact on oviductal contractions and preimplantation embryo development†. Biol Reprod 2025; 112:786-788. [PMID: 40057972 PMCID: PMC12078075 DOI: 10.1093/biolre/ioaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/04/2025] Open
Affiliation(s)
- Emily A McGlade
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, United States
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Jiude Mao
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Kalli K Stephens
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| | - Cayce N Rose
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, United States
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Wipawee Winuthayanon
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, United States
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
2
|
Scully DM, Xia T, Musina GR, McCown MA, Umezu K, Kircher BK, Behringer RR, Larina IV. Region-specific roles of oviductal motile cilia in oocyte/embryo transport and fertility†. Biol Reprod 2025; 112:651-662. [PMID: 39761349 PMCID: PMC11996758 DOI: 10.1093/biolre/ioaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 03/29/2025] Open
Abstract
The physiological and clinical importance of motile cilia in reproduction is well recognized; however, the specific role they play in transport through the oviduct and how ciliopathies lead to subfertility and infertility are still unclear. The contribution of cilia beating, fluid flow, and smooth muscle contraction to overall progressive transport within the oviduct remains under debate. Therefore, we investigated the role of cilia in the oviduct transport of preimplantation eggs/embryos using a combination of genetic and advanced imaging approaches. We show that the region of the oviduct where cumulus-oocyte complex circling occurs, around the time of fertilization, is correlated with asymmetrical mucosal fold arrangement and non-radially distributed ciliated epithelium. Our results suggest that motile cilia, as well as mucosal fold asymmetry, may contribute to the local flow fields that help steer luminal contents away from the epithelial walls. We also present, in vivo, volumetric evidence of delayed egg transport in a genetic mouse model with disrupted motile cilia function in the female reproductive system. Females with Dnah5 deleted in the oviduct epithelium are subfertile and demonstrate disrupted motile cilia activity within the oviduct mucosa. Fifty percent of Dnah5 mutant females have delayed egg transport where cumulus-oocyte complexes did not progress to the ampulla at the expected time point and remained within the ovarian bursa. The integration of advanced imaging with genetic dysfunction of motile cilia provides valuable insights into oviductal transport. Potentially, these data could be valuable for better understanding and management of tubal pathologies and human infertility.
Collapse
Affiliation(s)
- Deirdre M Scully
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Tian Xia
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Guzel R Musina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Michaela A McCown
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Kohei Umezu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Bonnie K Kircher
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irina V Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
Finnerty RM, Carulli DJ, Hedge A, Wang Y, Boadu F, Winuthayanon S, Jack Cheng J, Winuthayanon W. Multi-omics analyses and machine learning prediction of oviductal responses in the presence of gametes and embryos. eLife 2025; 13:RP100705. [PMID: 40009070 PMCID: PMC11864756 DOI: 10.7554/elife.100705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
The oviduct is the site of fertilization and preimplantation embryo development in mammals. Evidence suggests that gametes alter oviductal gene expression. To delineate the adaptive interactions between the oviduct and gamete/embryo, we performed a multi-omics characterization of oviductal tissues utilizing bulk RNA-sequencing (RNA-seq), single-cell RNA-sequencing (scRNA-seq), and proteomics collected from distal and proximal at various stages after mating in mice. We observed robust region-specific transcriptional signatures. Specifically, the presence of sperm induces genes involved in pro-inflammatory responses in the proximal region at 0.5 days post-coitus (dpc). Genes involved in inflammatory responses were produced specifically by secretory epithelial cells in the oviduct. At 1.5 and 2.5 dpc, genes involved in pyruvate and glycolysis were enriched in the proximal region, potentially providing metabolic support for developing embryos. Abundant proteins in the oviductal fluid were differentially observed between naturally fertilized and superovulated samples. RNA-seq data were used to identify transcription factors predicted to influence protein abundance in the proteomic data via a novel machine learning model based on transformers of integrating transcriptomics and proteomics data. The transformers identified influential transcription factors and correlated predictive protein expressions in alignment with the in vivo-derived data. Lastly, we found some differences between inflammatory responses in sperm-exposed mouse oviducts compared to hydrosalpinx Fallopian tubes from patients. In conclusion, our multi-omics characterization and subsequent in vivo confirmation of proteins/RNAs indicate that the oviduct is adaptive and responsive to the presence of sperm and embryos in a spatiotemporal manner.
Collapse
Affiliation(s)
- Ryan M Finnerty
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-ColumbiaColumbiaUnited States
| | - Daniel J Carulli
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| | - Akshata Hedge
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Yanli Wang
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Frimpong Boadu
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Sarayut Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| | - Jianlin Jack Cheng
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Wipawee Winuthayanon
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-ColumbiaColumbiaUnited States
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| |
Collapse
|
4
|
Faubert AC, Wang S. Clipping spline: interactive, dynamic 4D volume clipping and analysis based on thin plate spline. BIOMEDICAL OPTICS EXPRESS 2025; 16:499-519. [PMID: 39958850 PMCID: PMC11828437 DOI: 10.1364/boe.544231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 02/18/2025]
Abstract
Methods for seeing inside volumetric images are increasingly important with the rapid advancements in 3D and 4D (3D + time) biomedical imaging techniques. Here, we report a novel volume clipping method and its open-source implementation which enables unprecedented 4D visualization and analysis of embryonic mouse heart development with data from optical coherence tomography (OCT). Clipping a volume to extract information inside has long been a vital approach in biomedical image analysis; however, it is challenging to make a dynamic non-planar cutaway view that is simultaneously smooth, adjustable, efficient to compute, easy to control, and interactive in real time. We addressed this challenge by applying the thin plate spline (TPS) to create a new way of volume clipping, called the clipping spline. Specifically, the clipping spline produces a cutaway view by generating a binary mask based on the unique TPS surface that intersects with a set of 3D control points while having minimal curvature. We implemented this method in an open-source platform where the clipping spline can be interactively controlled for real-time, adjustable, and dynamic cutaway views into a volume. We also developed an algorithm that automatically connects and interpolates different sets of control points over time, providing 4D volume clipping. In addition to characterizing the clipping spline, we demonstrate its application by revealing a series of never-before-seen dynamics and processes of embryonic mouse heart development based on OCT data. We also show a TPS-based method for tracking the embryonic myocardium with control points over two timescales (heartbeat and development). Our results indicate that the clipping spline promises to be broadly used in volumetric biomedical image visualization and analysis, especially by the OCT community.
Collapse
Affiliation(s)
- Andre C. Faubert
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Shang Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
5
|
Finnerty RM, Carulli DJ, Hegde A, Wang Y, Baodu F, Winuthayanon S, Cheng J, Winuthayanon W. Multi-omics analyses and machine learning prediction of oviductal responses in the presence of gametes and embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598905. [PMID: 38915688 PMCID: PMC11195261 DOI: 10.1101/2024.06.13.598905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The oviduct is the site of fertilization and preimplantation embryo development in mammals. Evidence suggests that gametes alter oviductal gene expression. To delineate the adaptive interactions between the oviduct and gamete/embryo, we performed a multi-omics characterization of oviductal tissues utilizing bulk RNA-sequencing (RNA-seq), single-cell RNA-sequencing (scRNA-seq), and proteomics collected from distal and proximal at various stages after mating in mice. We observed robust region-specific transcriptional signatures. Specifically, the presence of sperm induces genes involved in pro-inflammatory responses in the proximal region at 0.5 days post-coitus (dpc). Genes involved in inflammatory responses were produced specifically by secretory epithelial cells in the oviduct. At 1.5 and 2.5 dpc, genes involved in pyruvate and glycolysis were enriched in the proximal region, potentially providing metabolic support for developing embryos. Abundant proteins in the oviductal fluid were differentially observed between naturally fertilized and superovulated samples. RNA-seq data were used to identify transcription factors predicted to influence protein abundance in the proteomic data via a novel machine learning model based on transformers of integrating transcriptomics and proteomics data. The transformers identified influential transcription factors and correlated predictive protein expressions in alignment with the in vivo-derived data. Lastly, we found some differences between inflammatory responses in sperm-exposed mouse oviducts compared to hydrosalpinx fallopian tubes from patients. In conclusion, our multi-omics characterization and subsequent in vivo confirmation of proteins/RNAs indicate that the oviduct is adaptive and responsive to the presence of sperm and embryos in a spatiotemporal manner.
Collapse
Affiliation(s)
- Ryan M. Finnerty
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| | - Daniel J. Carulli
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| | - Akshata Hegde
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Yanli Wang
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Frimpong Baodu
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Sarayut Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| | - Jianlin Cheng
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Wipawee Winuthayanon
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| |
Collapse
|
6
|
Rice P, Aitken M, Shir H, Cordova R, Montague J, Tran D, Utzinger U, Peng L, Mouneimne G, Barton J. A domed window chamber for multi-modality optical imaging. Biotechniques 2024; 76:535-545. [PMID: 39743842 DOI: 10.1080/07366205.2024.2445452] [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: 09/24/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Current dorsal skin flap window chambers with flat glass windows are compatible with optical coherence tomography (OCT) and multiphoton microscopy (MPM) imaging. However, light sheet fluorescence microscopy (LSFM) performs best with a cylindrical or spherical sample located between its two 90° objectives and when all sample materials have the same index of refraction (n). A modified window chamber with a domed viewing window made from fluorinated ethylene propylene (FEP), with n similar to water and tissue, was designed. In vitro imaging of collagen gels and microsphere phantoms with and without the dome showed small decreases in signal strength and image resolution due to the dome. Using a custom mouse platform for stabilization and anesthesia support, in vivo multimodality imaging with OCT, MPM, and LSFI was demonstrated.
Collapse
Affiliation(s)
- Photini Rice
- Biomedical Engineering, The University of Arizona, Tucson, AZ, USA
| | - Makenna Aitken
- Biomedical Engineering, The University of Arizona, Tucson, AZ, USA
| | - Hasina Shir
- Biomedical Engineering, The University of Arizona, Tucson, AZ, USA
| | - Ricky Cordova
- Biomedical Engineering, The University of Arizona, Tucson, AZ, USA
| | - Jenna Montague
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Dustin Tran
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Urs Utzinger
- Biomedical Engineering, The University of Arizona, Tucson, AZ, USA
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Leilei Peng
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Ghassan Mouneimne
- Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Jennifer Barton
- Biomedical Engineering, The University of Arizona, Tucson, AZ, USA
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
7
|
Suh J, Liu Y, Smith J, Watanabe M, Rollins AM, Jenkins MW. A Simple and Fast Optical Clearing Method for Whole-Mount Fluorescence In Situ Hybridization (FISH) Imaging. JOURNAL OF BIOPHOTONICS 2024:e202400258. [PMID: 39389582 DOI: 10.1002/jbio.202400258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024]
Abstract
We report a single-step optical clearing method that is compatible with RNA fluorescence in situ hybridization (FISH) imaging. We previously demonstrated microscopy imaging with immunohistochemistry and genetic reporters using a technique called lipid-preserving refractive index matching for prolonged imaging depth (LIMPID). Our protocol reliably produces high-resolution three-dimensional (3D) images with minimal aberrations using high magnification objectives, captures large field-of-view images of whole-mount tissues, and supports co-labeling with antibody and FISH probes. We also custom-designed FISH probes for quail embryos, demonstrating the ease of fabricating probes for use with less common animal models. Furthermore, we show high-quality 3D images using a conventional fluorescence microscope, without using more advanced depth sectioning instruments such as confocal or light-sheet microscopy. For broader adoption, we simplified and optimized 3D-LIMPID-FISH to minimize the barrier to entry, and we provide a detailed protocol to aid users with navigating the thick and thin of 3D microscopy.
Collapse
Affiliation(s)
- Junwoo Suh
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yehe Liu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jordan Smith
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michiko Watanabe
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael W Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Stephens KK, Finnerty RM, Grant DG, Winuthayanon S, Martin-DeLeon PA, Winuthayanon W. Proteomic analysis and in vivo visualization of extracellular vesicles from mouse oviducts during pre-implantation embryo development. FASEB J 2024; 38:e70035. [PMID: 39239798 PMCID: PMC11384279 DOI: 10.1096/fj.202400041rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Pre-implantation embryonic development occurs in the oviduct during the first few days of pregnancy. The presence of oviductal extracellular vesicles (oEVs, also called oviductosomes) is crucial for pre-implantation embryonic development in vivo as oEVs often contain molecular transmitters such as proteins. Therefore, evaluating oEV cargo during early pregnancy could provide insights into factors required for proper early embryonic development that are missing in the current in vitro embryo culture setting. In this study, we isolated oEVs from the oviductal fluid at estrus and different stages of early embryonic development. The 2306-3066 proteins in oEVs identified at the different time points revealed 58-60 common EV markers identified in exosome databases. Oviductal extracellular vesicle proteins from pregnant samples significantly differed from those in non-pregnant samples. In addition, superovulation changes the protein contents in oEVs compared to natural ovulation at estrus. Importantly, we have identified that embryo-protectant proteins such as high-mobility protein group B1 and serine (or cysteine) peptidase inhibitor were only enriched in the presence of embryos. We also visualized the physical interaction of EVs and the zona pellucida of 4- to 8-cell stage embryos using transmission electron microscopy as well as in vivo live imaging of epithelial cell-derived GFP-tagged CD9 mouse model. All protein data in this study are readily available to the scientific community in a searchable format at https://genes.winuthayanon.com/winuthayanon/oviduct_ev_proteins/. In conclusion, we identified oEVs proteins that could be tested to determine whether they can improve embryonic developmental outcomes in vivo and in vitro setting.
Collapse
Affiliation(s)
- Kalli K. Stephens
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, MO, 65201, USA
| | - Ryan M. Finnerty
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
- Translational Biosciences Program, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - DeAna G. Grant
- Electron Microscopy Core Facility, University of Missouri, Columbia, MO, 65211, USA
| | - Sarayut Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, MO, 65201, USA
| | | | - Wipawee Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, MO, 65201, USA
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
- Translational Biosciences Program, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
9
|
Morawiec S, Ajduk A, Stremplewski P, Kennedy BF, Szkulmowski M. Full-field optical coherence microscopy enables high-resolution label-free imaging of the dynamics of live mouse oocytes and early embryos. Commun Biol 2024; 7:1057. [PMID: 39191989 PMCID: PMC11349948 DOI: 10.1038/s42003-024-06745-x] [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: 04/10/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
High quality label-free imaging of oocytes and early embryos is essential for accurate assessment of their developmental potential, a key element of assisted reproduction procedures. To achieve this goal, we propose full-field optical coherence microscopy (FF-OCM), constructed as a compact module fully integrated with a commercial wide-field fluorescence microscope. Our system achieves optical sectioning in wide-field, high in-plane resolution of 0.5 µm, and high sensitivity to backscattered light. To demonstrate its imaging capabilities, we study live mouse oocytes and embryos at all important stages of meiotic maturation and early embryogenesis. Our system enables visualization of intracellular structures, which are not visible in common bright-field microscopy, i.e., internal structure of nuclear apparatus, cytoskeletal filaments, cellular cortex, cytoplasmic protrusions, or zona pellucida features. Additionally, we visualize and quantify intracellular dynamics like cytoplasmic stirring motion, nuclear envelope fluctuations and nucleolus mobility. Altogether, we demonstrate that FF-OCM is a powerful tool for research in developmental biology that also holds great potential for non-invasive time-lapse monitoring of oocyte and embryo quality in assisted reproduction.
Collapse
Affiliation(s)
- Seweryn Morawiec
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Torun, Torun, Poland.
| | - Anna Ajduk
- Department of Embryology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Patrycjusz Stremplewski
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Brendan F Kennedy
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Torun, Torun, Poland
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA, Australia
| | - Maciej Szkulmowski
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Torun, Torun, Poland
| |
Collapse
|
10
|
Chow DJX, Tan TCY, Upadhya A, Lim M, Dholakia K, Dunning KR. Viewing early life without labels: optical approaches for imaging the early embryo†. Biol Reprod 2024; 110:1157-1174. [PMID: 38647415 PMCID: PMC11180623 DOI: 10.1093/biolre/ioae062] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
Embryo quality is an important determinant of successful implantation and a resultant live birth. Current clinical approaches for evaluating embryo quality rely on subjective morphology assessments or an invasive biopsy for genetic testing. However, both approaches can be inherently inaccurate and crucially, fail to improve the live birth rate following the transfer of in vitro produced embryos. Optical imaging offers a potential non-invasive and accurate avenue for assessing embryo viability. Recent advances in various label-free optical imaging approaches have garnered increased interest in the field of reproductive biology due to their ability to rapidly capture images at high resolution, delivering both morphological and molecular information. This burgeoning field holds immense potential for further development, with profound implications for clinical translation. Here, our review aims to: (1) describe the principles of various imaging systems, distinguishing between approaches that capture morphological and molecular information, (2) highlight the recent application of these technologies in the field of reproductive biology, and (3) assess their respective merits and limitations concerning the capacity to evaluate embryo quality. Additionally, the review summarizes challenges in the translation of optical imaging systems into routine clinical practice, providing recommendations for their future development. Finally, we identify suitable imaging approaches for interrogating the mechanisms underpinning successful embryo development.
Collapse
Affiliation(s)
- Darren J X Chow
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
| | - Tiffany C Y Tan
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
| | - Avinash Upadhya
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Megan Lim
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Kishan Dholakia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
- Scottish Universities Physics Alliance, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom
| | - Kylie R Dunning
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
11
|
Kircher BK, McCown MA, Scully DM, Behringer RR, Larina IV. Structural analysis of the female reptile reproductive system by micro-computed tomography and optical coherence tomography†. Biol Reprod 2024; 110:1077-1085. [PMID: 38641547 PMCID: PMC11180613 DOI: 10.1093/biolre/ioae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 04/21/2024] Open
Abstract
Volumetric data provide unprecedented structural insight to the reproductive tract and add vital anatomical context to the relationships between organs. The morphology of the female reproductive tract in non-avian reptiles varies between species, corresponding to a broad range of reproductive modes and providing valuable insight to comparative investigations of reproductive anatomy. However, reproductive studies in reptilian models, such as the brown anole studied here, have historically relied on histological methods to understand the anatomy. While these methods are highly effective for characterizing the cell types present in each organ, histological methods lose the 3D relationships between images and leave the architecture of the organ system poorly understood. We present the first comprehensive volumetric analyses of the female brown anole reproductive tract using two non-invasive, non-destructive imaging modalities: micro-computed tomography (microCT) and optical coherence tomography (OCT). Both are specialized imaging technologies that facilitate high-throughput imaging and preserve three-dimensional information. This study represents the first time that microCT has been used to study all reproductive organs in this species and the very first time that OCT has been applied to this species. We show how the non-destructive volumetric imaging provided by each modality reveals anatomical context including orientation and relationships between reproductive organs of the anole lizard. In addition to broad patterns of morphology, both imaging modalities provide the high resolution necessary to capture details and key anatomical features of each organ. We demonstrate that classic histological features can be appreciated within whole-organ architecture in volumetric imaging using microCT and OCT, providing the complementary information necessary to understand the relationships between tissues and organs in the reproductive system. This side-by-side imaging analysis using microCT and OCT allows us to evaluate the specific advantages and limitations of these two methods for the female reptile reproductive system.
Collapse
Affiliation(s)
- Bonnie K Kircher
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Michaela A McCown
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Deirdre M Scully
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Irina V Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
Folts L, Martinez AS, McKey J. Tissue clearing and imaging approaches for in toto analysis of the reproductive system†. Biol Reprod 2024; 110:1041-1054. [PMID: 38159104 PMCID: PMC11180619 DOI: 10.1093/biolre/ioad182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024] Open
Abstract
New microscopy techniques in combination with tissue clearing protocols and emerging analytical approaches have presented researchers with the tools to understand dynamic biological processes in a three-dimensional context. This paves the road for the exploration of new research questions in reproductive biology, for which previous techniques have provided only approximate resolution. These new methodologies now allow for contextualized analysis of far-larger volumes than was previously possible. Tissue optical clearing and three-dimensional imaging techniques posit the bridging of molecular mechanisms, macroscopic morphogenic development, and maintenance of reproductive function into one cohesive and comprehensive understanding of the biology of the reproductive system. In this review, we present a survey of the various tissue clearing techniques and imaging systems, as they have been applied to the developing and adult reproductive system. We provide an overview of tools available for analysis of experimental data, giving particular attention to the emergence of artificial intelligence-assisted methods and their applicability to image analysis. We conclude with an evaluation of how novel image analysis approaches that have been applied to other organ systems could be incorporated into future experimental evaluation of reproductive biology.
Collapse
Affiliation(s)
- Lillian Folts
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO, USA
| | - Anthony S Martinez
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO, USA
| | - Jennifer McKey
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO, USA
| |
Collapse
|
13
|
Yang L, Yan C, Tao S, He Y, Zhao J, Wang Y, Wu Y, Liu N, Qin Y. In Vivo Imaging of Rabbit Follicles through Combining Ultrasound Bio-Microscopy and Intravital Window. Animals (Basel) 2024; 14:1727. [PMID: 38929346 PMCID: PMC11200761 DOI: 10.3390/ani14121727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Continuous ovarian imaging has been proven to be a method for monitoring the development of follicles in vivo. The aim of this study was to evaluate the efficacy of combining ultrasound bio-microscopy (UBM) with an intravital window for follicle imaging in rabbits and to monitor the ovarian dynamic processes. New Zealand White female rabbits (n = 10) received ovarian translocation to a subcutaneous position. The ovarian tissue was sutured onto the abdominal muscles and covered with an intravital window for the continuous monitoring of the follicles using UBM. Results show that physiological changes (red blood cell and white blood cell counts, feed intake, and body weight change) in rabbits induced by surgery returned to normal physiological levels in one week. Furthermore, UBM could provide high-resolution imaging of follicles through the intravital window. Daily monitoring of ovarian dynamic processes for 6 days displayed variabilities in follicle counts and size. Collectively, these results provide a relatively new method to monitor ovarian dynamic processes and to understand the reproductive physiology of female rabbits.
Collapse
Affiliation(s)
- Lihan Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Chang Yan
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Siming Tao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Yifeilong He
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Jing Zhao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Yanya Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Yingjie Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Yinghe Qin
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; (L.Y.); (C.Y.); (S.T.); (Y.H.); (J.Z.); (Y.W.); (Y.W.)
- National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| |
Collapse
|
14
|
Anvar Z, Jochum MD, Chakchouk I, Sharif M, Demond H, To AK, Kraushaar DC, Wan YW, Andrews S, Kelsey G, Veyver IB. Maternal loss-of-function of Nlrp2 results in failure of epigenetic reprogramming in mouse oocytes. RESEARCH SQUARE 2024:rs.3.rs-4457414. [PMID: 38883732 PMCID: PMC11177987 DOI: 10.21203/rs.3.rs-4457414/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Background NLRP2 belongs to the subcortical maternal complex (SCMC) of mammalian oocytes and preimplantation embryos. This multiprotein complex, encoded by maternal-effect genes, plays a pivotal role in the zygote-to-embryo transition, early embryogenesis, and epigenetic (re)programming. The maternal inactivation of genes encoding SCMC proteins has been linked to infertility and subfertility in mice and humans. However, the underlying molecular mechanisms for the diverse functions of the SCMC, particularly how this cytoplasmic structure influences DNA methylation, which is a nuclear process, are not fully understood. Results We undertook joint transcriptome and DNA methylome profiling of pre-ovulatory germinal-vesicle oocytes from Nlrp2-null, heterozygous (Het), and wild-type (WT) female mice. We identified numerous differentially expressed genes (DEGs) in Het and Nlrp2-null when compared to WT oocytes. The genes for several crucial factors involved in oocyte transcriptome modulation and epigenetic reprogramming, such as DNMT1, UHRF1, KDM1B and ZFP57 were overexpressed in Het and Nlrp2-null oocytes. Absence or reduction of Nlrp2, did not alter the distinctive global DNA methylation landscape of oocytes, including the bimodal pattern of the oocyte methylome. Additionally, although the methylation profile of germline differentially methylated regions (gDMRs) of imprinted genes was preserved in oocytes of Het and Nlrp2-null mice, we found altered methylation in oocytes of both genotypes at a small percentage of the oocyte-characteristic hyper- and hypomethylated domains. Through a tiling approach, we identified specific DNA methylation differences between the genotypes, with approximately 1.3% of examined tiles exhibiting differential methylation in Het and Nlrp2-null compared to WT oocytes. Conclusions Surprisingly, considering the well-known correlation between transcription and DNA methylation in developing oocytes, we observed no correlation between gene expression differences and gene-body DNA methylation differences in Nlrp2-null versus WT oocytes or Het versus WT oocytes. We therefore conclude that post-transcriptional changes in the stability of transcripts rather than altered transcription is primarily responsible for transcriptome differences in Nlrp2-null and Het oocytes.
Collapse
|
15
|
Iyer RR, Žurauskas M, Rao Y, Chaney EJ, Boppart SA. Bichromatic tetraphasic full-field optical coherence microscopy. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:S22704. [PMID: 38584966 PMCID: PMC10996847 DOI: 10.1117/1.jbo.29.s2.s22704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024]
Abstract
Significance Full-field optical coherence microscopy (FF-OCM) is a prevalent technique for backscattering and phase imaging with epi-detection. Traditional methods have two limitations: suboptimal utilization of functional information about the sample and complicated optical design with several moving parts for phase contrast. Aim We report an OCM setup capable of generating dynamic intensity, phase, and pseudo-spectroscopic contrast with single-shot full-field video-rate imaging called bichromatic tetraphasic (BiTe) full-field OCM with no moving parts. Approach BiTe OCM resourcefully uses the phase-shifting properties of anti-reflection (AR) coatings outside the rated bandwidths to create four unique phase shifts, which are detected with two emission filters for spectroscopic contrast. Results BiTe OCM overcomes the disadvantages of previous FF-OCM setup techniques by capturing both the intensity and phase profiles without any artifacts or speckle noise for imaging scattering samples in three-dimensional (3D). BiTe OCM also utilizes the raw data effectively to generate three complementary contrasts: intensity, phase, and color. We demonstrate BiTe OCM to observe cellular dynamics, image live, and moving micro-animals in 3D, capture the spectroscopic hemodynamics of scattering tissues along with dynamic intensity and phase profiles, and image the microstructure of fall foliage with two different colors. Conclusions BiTe OCM can maximize the information efficiency of FF-OCM while maintaining overall simplicity in design for quantitative, dynamic, and spectroscopic characterization of biological samples.
Collapse
Affiliation(s)
- Rishyashring R. Iyer
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Mantas Žurauskas
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
| | - Yug Rao
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Eric J. Chaney
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
| | - Stephen A. Boppart
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Department of Bioengineering, Urbana, Illinois, United States
- University of Illinois Urbana Champaign, NIH/NIBIB P41 Center for Label-free Imaging and Multiscale Biophotonics (CLIMB), Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Cancer Center at Illinois, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Carle Illinois College of Medicine, Urbana, Illinois, United States
| |
Collapse
|
16
|
Hunter MI, Thies KM, Winuthayanon W. Hormonal regulation of cilia in the female reproductive tract. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2024; 34:100503. [PMID: 38293616 PMCID: PMC10824531 DOI: 10.1016/j.coemr.2024.100503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
This review intends to bridge the gap between our knowledge of steroid hormone regulation of motile cilia and the potential involvement of the primary cilium focusing on the female reproductive tract functions. The review emphasizes hormonal regulation of the motile and primary cilia in the oviduct and uterus. Steroid hormones including estrogen, progesterone, and testosterone act through their cognate receptors to regulate the development and biological function of the reproductive tracts. These hormones modulate motile ciliary beating and, in some cases, primary cilia function. Dysfunction of motile or primary cilia due to genetic anomalies, hormone imbalances, or loss of steroid hormone receptors impairs mammalian fertility. However, further research on hormone modulation of ciliary function, especially in the primary cilium, and its signaling cascades will provide insights into the pathogenesis of mammalian infertility and the development of contraceptives or infertility treatments targeting primary and/or motile cilia.
Collapse
Affiliation(s)
- Mark I. Hunter
- OB/GYN & Women’s Health Department, School of Medicine, University of Missouri – Columbia, Columbia, MO, 65211, United States
| | - Karen M. Thies
- OB/GYN & Women’s Health Department, School of Medicine, University of Missouri – Columbia, Columbia, MO, 65211, United States
| | - Wipawee Winuthayanon
- OB/GYN & Women’s Health Department, School of Medicine, University of Missouri – Columbia, Columbia, MO, 65211, United States
| |
Collapse
|
17
|
Umezu K, Musina GR, Larina IV. In vivo dynamic volumetric imaging of mouse testis and epididymis with optical coherence tomography†. Biol Reprod 2024; 110:365-376. [PMID: 37971359 PMCID: PMC10873499 DOI: 10.1093/biolre/ioad158] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/19/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
The implementation of live imaging in reproductive research is crucial for studying the physiological dynamics. Sperm transport is a highly dynamic process regulated by tubular contractions and luminal flows within the male reproductive tract. However, due to the lack of imaging techniques to capture these dynamics in vivo, there is little information on the physiological and biomechanical regulation of sperm transport through the male reproductive tract. Here, we present a functional in vivo imaging approach using optical coherence tomography, enabling live, label-free, depth-resolved, three-dimensional, high-resolution visualization of the mouse testis and epididymis. With this approach, we spatiotemporally captured tubular contractility in mouse testis and epididymis, as well as microstructures of these reproductive organs. Our findings demonstrated that the contraction frequency varies significantly depending on the epididymal regions, suggesting the spatial regulation of epididymal contractility. Furthermore, we implemented quantitative measurements of the contraction wave and luminal transport through the epididymal duct, revealing the physiological dynamics within the male reproductive tract. The results show that the contraction wave propagates along the epididymal duct and the wave propagation velocity was estimated in vivo. In conclusion, this is the first study to develop in vivo dynamic volumetric imaging of the male reproductive tract, which allows for quantitative analysis of the dynamics associated with sperm transport. This study sets a platform for various studies investigating normal and abnormal male reproductive physiology as well as the pharmacological and environmental effects on reproductive functions in mouse models, ultimately contributing to a comprehensive understanding of male reproductive disorders.
Collapse
Affiliation(s)
- Kohei Umezu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Guzel R Musina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Irina V Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
18
|
Xia T, Umezu K, Scully DM, Wang S, Larina IV. In vivo volumetric depth-resolved imaging of cilia metachronal waves using dynamic optical coherence tomography. OPTICA 2023; 10:1439-1451. [PMID: 38665775 PMCID: PMC11044847 DOI: 10.1364/optica.499927] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 04/28/2024]
Abstract
Motile cilia are dynamic hair-like structures covering epithelial surfaces in multiple organs. The periodic coordinated beating of cilia creates waves propagating along the surface, known as the metachronal waves, which transport fluids and mucus along the epithelium. Motile ciliopathies result from disrupted coordinated cilia beating and are associated with serious clinical complications, including reproductive disorders. Despite the recognized clinical significance, research of cilia dynamics is extremely limited. Here, we present quantitative imaging of cilia metachronal waves volumetrically through tissue layers using dynamic optical coherence tomography (OCT). Our method relies on spatiotemporal mapping of the phase of intensity fluctuations in OCT images caused by the ciliary beating. We validated our new method ex vivo and implemented it in vivo to visualize cilia metachronal wave propagation within the mouse fallopian tube. This method can be extended to the assessment of physiological cilia function and ciliary dyskinesias in various organ systems, contributing to better management of pathologies associated with motile ciliopathies.
Collapse
Affiliation(s)
- Tian Xia
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Kohei Umezu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Deirdre M. Scully
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Shang Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, USA
| | - Irina V. Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
19
|
McGlade EA, Stephens KK, Winuthayanon S, Anamthathmakula P, Holtzman MJ, Winuthayanon W. Classical Estrogen Signaling in Ciliated Epithelial Cells of the Oviduct Is Nonessential for Fertility in Female Mice. Endocrinology 2023; 165:bqad163. [PMID: 37942801 PMCID: PMC10658216 DOI: 10.1210/endocr/bqad163] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/03/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023]
Abstract
Ciliary action performs a critical role in the oviduct (Fallopian tube) during pregnancy establishment through sperm and egg transport. The disruption of normal ciliary function in the oviduct affects oocyte pick-up and is a contributing factor to female infertility. Estrogen is an important regulator of ciliary action in the oviduct and promotes ciliogenesis in several species. Global loss of estrogen receptor α (ESR1) leads to infertility. We have previously shown that ESR1 in the oviductal epithelial cell layer is required for female fertility. Here, we assessed the role of estrogen on transcriptional regulation of ciliated epithelial cells of the oviduct using single-cell RNA-sequencing analysis. We observed minor variations in ciliated cell genes in the proximal region (isthmus and uterotubal junction) of the oviduct. However, 17β-estradiol treatment had little impact on the gene expression profile of ciliated epithelial cells. We also conditionally ablated Esr1 from ciliated epithelial cells of the oviduct (called ciliated Esr1d/d mice). Our studies showed that ciliated Esr1d/d females had fertility rates comparable to control females, did not display any disruptions in preimplantation embryo development or embryo transport to the uterus, and had comparable cilia formation to control females. However, we observed some incomplete deletion of Esr1 in the ciliated epithelial cells, especially in the ampulla region. Nevertheless, our data suggest that ESR1 expression in ciliated cells of the oviduct is dispensable for ciliogenesis and nonessential for female fertility in mice.
Collapse
Affiliation(s)
- Emily A McGlade
- Obstetrics, Gynecology and Women's Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Kalli K Stephens
- Obstetrics, Gynecology and Women's Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | | | | | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St.Louis, MO 63110, USA
| | - Wipawee Winuthayanon
- Obstetrics, Gynecology and Women's Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
20
|
Xin Q, Yu G, Feng I, Dean J. Chromatin remodeling of prostaglandin signaling in smooth muscle enables mouse embryo passage through the female reproductive tract. Dev Cell 2023; 58:1716-1732.e8. [PMID: 37714160 DOI: 10.1016/j.devcel.2023.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/10/2023] [Accepted: 08/22/2023] [Indexed: 09/17/2023]
Abstract
Early mammalian development occurs during embryo transit of the female reproductive tract. Transport is orchestrated by secreted oviduct fluid, unidirectional beating of epithelial cilia, and smooth muscle contractions. Using gene-edited mice, we document that conditional disruption of a component of the SWI/SNF chromatin remodeling complex in smooth muscle cells prevents transport through the oviduct without perturbing embryogenesis. Analysis with RNA sequencing (RNA-seq), transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunocleavage sequencing (ChIC-seq), and pharmacologic rescue experiments implicated prostaglandin signaling pathways. In comparison with controls, gene-edited mice had compromised chromatin accessibility at enhancer/promoters of Ptgs2, Pla2g16, Pla2r1, and Ptger3 (EP3) as well as decreased enhancer-promoter interactive looping critical for Ptgs2 (aka Cox-2) expression in a SWI/SNF complex-dependent manner. Treatment of wild-type mice with prostaglandin inhibitors phenocopied the genetically induced defect.
Collapse
Affiliation(s)
- Qiliang Xin
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Guoyun Yu
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Iris Feng
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
21
|
Hosotani M, Ichii O, Watanabe T, Kon Y. Oocyte cumulus complex quality and oviduct transportation velocity in systemic autoimmune disease model mice. Exp Biol Med (Maywood) 2023; 248:1359-1363. [PMID: 36961243 PMCID: PMC10657593 DOI: 10.1177/15353702231160875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/30/2023] [Indexed: 03/25/2023] Open
Abstract
Oocyte transportation by the oviduct involves the interaction between ciliated epithelial cells and cumulus cells. To determine whether the quality of cumulus-oocyte complexes (COCs) changes the transportation property of COCs, we compared the transportation velocity of COCs (TVC) by the infundibulum ex vivo with various combinations of infundibula and COCs collected from different mice. We used young and aged C57BL/6N and MRL/MpJ, and MRL/MpJ-Faslpr/lpr mice as the strains with intact female reproductive function and the systemic autoimmune disease model exhibiting oocyte pick-up dysfunction owing to the morphofunctional abnormality of ciliated epithelium, respectively. The TVC of aged MRL strains was less than that of aged C57BL/6N mice, suggesting that aging affects the transportation of COCs in MRL strains. The TVC of aged MRL/MpJ-Faslpr/lpr mice was the least among all examined combinations, whereas the TVC accelerated when the infundibulum or COCs were collected from other strains. These results indicate that the transportation property of COCs is determined not only by the ciliary function in the infundibulum but also by the properties of COCs.
Collapse
Affiliation(s)
- Marina Hosotani
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo 060-0818, Japan
| | - Takafumi Watanabe
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Basic Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
22
|
Umezu K, Larina IV. Optical coherence tomography for dynamic investigation of mammalian reproductive processes. Mol Reprod Dev 2023; 90:3-13. [PMID: 36574640 PMCID: PMC9877170 DOI: 10.1002/mrd.23665] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 12/28/2022]
Abstract
The biological events associated with mammalian reproductive processes are highly dynamic and tightly regulated by molecular, genetic, and biomechanical factors. Implementation of live imaging in reproductive research is vital for the advancement of our understanding of normal reproductive physiology and for improving the management of reproductive disorders. Optical coherence tomography (OCT) is emerging as a promising tool for dynamic volumetric imaging of various reproductive processes in mice and other animal models. In this review, we summarize recent studies employing OCT-based approaches toward the investigation of reproductive processes in both, males and females. We describe how OCT can be applied to study structural features of the male reproductive system and sperm transport through the male reproductive tract. We review OCT applications for in vitro and dynamic in vivo imaging of the female reproductive system, staging and tracking of oocytes and embryos, and investigations of the oocyte/embryo transport through the oviduct. We describe how the functional OCT approach can be applied to the analysis of cilia dynamics within the male and female reproductive systems. We also discuss the areas of research, where OCT could find potential applications to progress our understanding of normal reproductive physiology and reproductive disorders.
Collapse
Affiliation(s)
- Kohei Umezu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Irina V Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
23
|
Faubert AC, Larina IV, Wang S. Open-source, highly efficient, post-acquisition synchronization for 4D dual-contrast imaging of the mouse embryonic heart over development with optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2023; 14:163-181. [PMID: 36698661 PMCID: PMC9842004 DOI: 10.1364/boe.475027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 05/28/2023]
Abstract
Dynamic imaging of the beating embryonic heart in 3D is critical for understanding cardiac development and defects. Optical coherence tomography (OCT) plays an important role in embryonic heart imaging with its unique imaging scale and label-free contrasts. In particular, 4D (3D + time) OCT imaging enabled biomechanical analysis of the developing heart in various animal models. While ultrafast OCT systems allow for direct volumetric imaging of the beating heart, the imaging speed remains limited, leading to an image quality inferior to that produced by post-acquisition synchronization. As OCT systems become increasingly available to a wide range of biomedical researchers, a more accessible 4D reconstruction method is required to enable the broader application of OCT in the dynamic, volumetric assessment of embryonic heartbeat. Here, we report an open-source, highly efficient, post-acquisition synchronization method for 4D cardiodynamic and hemodynamic imaging of the mouse embryonic heart. Relying on the difference between images to characterize heart wall movements, the method provides good sensitivity to the cardiac activity when aligning heartbeat phases, even at early stages when the heart wall occupies only a small number of pixels. The method works with a densely sampled single 3D data acquisition, which, unlike the B-M scans required by other methods, is readily available in most commercial OCT systems. Compared with an existing approach for the mouse embryonic heart, this method shows superior reconstruction quality. We present the robustness of the method through results from different embryos with distinct heart rates, ranging from 1.24 Hz to 2.13 Hz. Since the alignment process operates on a 1D signal, the method has a high efficiency, featuring sub-second alignment time while utilizing ∼100% of the original image files. This allows us to achieve repeated, dual-contrast imaging of mouse embryonic heart development. This new, open-source method could facilitate research using OCT to study early cardiogenesis.
Collapse
Affiliation(s)
- Andre C. Faubert
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Irina V. Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shang Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
24
|
Biswas A, Ng BH, Prabhakaran VS, Chan CJ. Squeezing the eggs to grow: The mechanobiology of mammalian folliculogenesis. Front Cell Dev Biol 2022; 10:1038107. [PMID: 36531957 PMCID: PMC9756970 DOI: 10.3389/fcell.2022.1038107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/16/2022] [Indexed: 08/25/2023] Open
Abstract
The formation of functional eggs (oocyte) in ovarian follicles is arguably one of the most important events in early mammalian development since the oocytes provide the bulk genetic and cytoplasmic materials for successful reproduction. While past studies have identified many genes that are critical to normal ovarian development and function, recent studies have highlighted the role of mechanical force in shaping folliculogenesis. In this review, we discuss the underlying mechanobiological principles and the force-generating cellular structures and extracellular matrix that control the various stages of follicle development. We also highlight emerging techniques that allow for the quantification of mechanical interactions and follicular dynamics during development, and propose new directions for future studies in the field. We hope this review will provide a timely and useful framework for future understanding of mechano-signalling pathways in reproductive biology and diseases.
Collapse
Affiliation(s)
- Arikta Biswas
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Boon Heng Ng
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | | | - Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Chan CJ, Hirashima T. Tissue hydraulics in reproduction. Semin Cell Dev Biol 2022; 131:124-133. [PMID: 35606275 DOI: 10.1016/j.semcdb.2022.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The development of functional eggs and sperm are critical processes in mammalian development as they ensure successful reproduction and species propagation. While past studies have identified important genes that regulate these processes, the roles of luminal flow and fluid stress in reproductive biology remain less well understood. Here, we discuss recent evidence that support the diverse functions of luminal fluid in oogenesis, spermatogenesis and embryogenesis. We also review emerging techniques that allow for precise quantification and perturbation of tissue hydraulics in female and male reproductive systems, and propose new questions and approaches in this field. We hope this review will provide a useful resource to inspire future research in tissue hydraulics in reproductive biology and diseases.
Collapse
Affiliation(s)
- Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore.
| | - Tsuyoshi Hirashima
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; The Hakubi Center/Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Japan Science and Technology Agency, PRESTO, Kawaguchi, Japan.
| |
Collapse
|
26
|
Wagner LE, Melnyk O, Duffett BE, Linnemann AK. Mouse models and human islet transplantation sites for intravital imaging. Front Endocrinol (Lausanne) 2022; 13:992540. [PMID: 36277698 PMCID: PMC9579277 DOI: 10.3389/fendo.2022.992540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/09/2022] [Indexed: 01/12/2023] Open
Abstract
Human islet transplantations into rodent models are an essential tool to aid in the development and testing of islet and cellular-based therapies for diabetes prevention and treatment. Through the ability to evaluate human islets in an in vivo setting, these studies allow for experimental approaches to answer questions surrounding normal and disease pathophysiology that cannot be answered using other in vitro and in vivo techniques alone. Intravital microscopy enables imaging of tissues in living organisms with dynamic temporal resolution and can be employed to measure biological processes in transplanted human islets revealing how experimental variables can influence engraftment, and transplant survival and function. A key consideration in experimental design for transplant imaging is the surgical placement site, which is guided by the presence of vasculature to aid in functional engraftment of the islets and promote their survival. Here, we review transplantation sites and mouse models used to study beta cell biology in vivo using intravital microscopy and we highlight fundamental observations made possible using this methodology.
Collapse
Affiliation(s)
- Leslie E. Wagner
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Olha Melnyk
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bryce E. Duffett
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Amelia K. Linnemann
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
27
|
Scully DM, Larina IV. Mouse embryo phenotyping with optical coherence tomography. Front Cell Dev Biol 2022; 10:1000237. [PMID: 36158219 PMCID: PMC9500480 DOI: 10.3389/fcell.2022.1000237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023] Open
Abstract
With the explosion of gene editing tools in recent years, there has been a much greater demand for mouse embryo phenotyping, and traditional methods such as histology and histochemistry experienced a methodological renaissance as they became the principal tools for phenotyping. However, it is important to explore alternative phenotyping options to maximize time and resources and implement volumetric structural analysis for enhanced investigation of phenotypes. Cardiovascular phenotyping, in particular, is important to perform in vivo due to the dramatic structural and functional changes that occur in heart development over relatively short periods of time. Optical coherence tomography (OCT) is one of the most exciting advanced imaging techniques emerging within the field of developmental biology, and this review provides a summary of how it is currently being implemented in mouse embryo investigations and phenotyping. This review aims to provide an understanding of the approaches used in optical coherence tomography and how they can be applied in embryology and developmental biology, with the overall aim of bridging the gap between biology and technology.
Collapse
|
28
|
Umezu K, Xia T, Larina IV. Dynamic volumetric imaging and cilia beat mapping in the mouse male reproductive tract with optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2022; 13:3672-3684. [PMID: 35781970 PMCID: PMC9208606 DOI: 10.1364/boe.459937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Spermatozoa transport within the male reproductive tract is a highly dynamic and biologically important reproductive event. However, due to the lack of live volumetric imaging technologies and quantitative measurements, there is little information on the dynamic aspect and regulation of this process. Here, we presented ex vivo dynamic volumetric imaging of the mouse testis, efferent duct, epididymis, and vas deferens at a micro-scale spatial resolution with optical coherence tomography (OCT). Micro computed tomography imaging is presented as a reference for the proposed OCT imaging. Application of functional OCT analysis allowed for 3D mapping of the cilia beat frequency in the efferent duct, which volumetrically visualized the spatial distribution of the ciliated cells and corresponding ciliary activities. Potentially these analyses could be expanded to in vivo settings through intravital approach. In summary, this study demonstrated that OCT has a great potential to investigate the microstructure and dynamics, such as cilia beating, muscle contractions, and sperm transport, within the male reproductive tract.
Collapse
|