1
|
Mikami K, Kozono Y, Masukawa M, Kobayashi S. A fast in situ hybridization chain reaction method in Drosophila embryos and ovaries. Fly (Austin) 2025; 19:2428499. [PMID: 39639000 PMCID: PMC11633216 DOI: 10.1080/19336934.2024.2428499] [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: 08/23/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
The in situ hybridization chain reaction (isHCR) is a powerful method for visualizing mRNA in many species. We present a rapid isHCR method for Drosophila embryos and ovaries. Ethylene carbonate was added to the hybridization buffer to facilitate the hybridization reaction, and a modified short hairpin DNA was used in the amplification reaction; these modifications decreased the RNA staining time from 3 days to 1 day. This method is compatible with immunohistochemistry and can detect multiple mRNAs. The proposed method could significantly reduce staining time for Drosophila researchers using isHCR.
Collapse
Affiliation(s)
- Kyohei Mikami
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yasuhiro Kozono
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masaki Masukawa
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Kobayashi
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
2
|
Lion AT, Bodine SM, McCutcheon KR, Ghogale M, Chandragiri S, Abayawardena D, Shrestha BD, Descoteaux A, Alvarez K, Balkman JA, Cocke B, Wikramanayake AH, Schlezinger J, Wong JY, Prakash VN, Bradham CA. PFAS Compounds PFOA and Gen X are Teratogenic to Sea Urchin Embryos. Dev Biol 2025; 525:S0012-1606(25)00163-0. [PMID: 40480305 DOI: 10.1016/j.ydbio.2025.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 05/23/2025] [Accepted: 06/03/2025] [Indexed: 06/11/2025]
Abstract
Per-and polyfluorinated substances (PFAS) are synthetic compounds used in the production of fluoropolymer coatings found in products such as non-stick pans, clothing, cosmetics, and food packaging. These highly persistent molecules are known as "forever chemicals" since they neither degrade environmentally nor break down enzymatically within biological systems. PFAS compounds readily contaminate water sources, and as a result, certain PFAS molecules have bioaccumulated in exposed species including humans. The purpose of this study was to define the effect of two PFAS molecules, the ostensibly more toxic perfluorooctanoic acid (PFOA) and the more recent, reportedly safer chemical hexafluoropropylene oxide dimer acid (Gen X), on the development of Lytechinus variegatus sea urchin embryos. We examined the effects of PFOA and Gen X on development and patterning using morphological analysis, immunostaining, HCR-FISH, and Particle Image Velocimetry (PIV). The results show that both PFAS compounds are teratogenic to sea urchin embryos. PFOA and Gen X each function at different intervals during development and provoke distinct phenotypic and gene expression outcomes. Despite beliefs that Gen X would be a safer alternative, our findings indicate that Gen X has earlier and more severe effects on endomesoderm and dorsal-ventral axis specification, neural development and function, and pattern formation compared to PFOA. These results illustrate the dangerous teratogenic potential of environmentally accumulating PFAS like Gen X, underscoring the negative ecological implications that accompany continuing commercial and industrial use of PFAS in the absence of remediation strategies.
Collapse
Affiliation(s)
- Alexandra T Lion
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston MA, USA
| | | | | | - Mayank Ghogale
- Bioinformatics Program, Boston University, Boston MA, USA
| | | | | | | | - Abigail Descoteaux
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston MA, USA; Biological Design Center, College of Engineering, Boston University, Boston MA, USA
| | - Kathryn Alvarez
- Department of Physics, University of Miami, Coral Gables FL, USA
| | | | - Breelyn Cocke
- Department of Physics, University of Miami, Coral Gables FL, USA
| | | | | | - Joyce Y Wong
- Department of Biomedical Engineering, Boston University, Boston MA, USA
| | - Vivek N Prakash
- Department of Physics, University of Miami, Coral Gables FL, USA; Department of Biology, University of Miami, Coral Gables FL, USA; Department of Marine Biology and Ecology, University of Miami, Miami FL, USA
| | - Cynthia A Bradham
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston MA, USA; Biology Department, Boston University, Boston MA, USA; Bioinformatics Program, Boston University, Boston MA, USA; Biological Design Center, College of Engineering, Boston University, Boston MA, USA.
| |
Collapse
|
3
|
Moore PJ, Hoffman K, Ahmed S, Fletcher JR, Wiggen TD, Lucas SK, Arif SJ, Gilbertsen AJ, Kent LA, Fiege JK, Langlois RA, O'Grady SM, Hunter RC. Dual oxic-anoxic co-culture enables direct study of anaerobe-host interactions at the airway epithelial interface. mBio 2025; 16:e0133824. [PMID: 40366160 PMCID: PMC12077211 DOI: 10.1128/mbio.01338-24] [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] [Received: 05/01/2024] [Accepted: 03/10/2025] [Indexed: 05/15/2025] Open
Abstract
Strict and facultative anaerobic bacteria are widely associated with both acute and chronic airway diseases. However, their potential role(s) in disease pathophysiology remains poorly understood due to inherent limitations of existing laboratory models and conflicting oxygen demands between anaerobes and host cells. To address these limitations, here, we describe a dual oxic-anoxic culture (DOAC) approach that maintains an oxygen-limited microenvironment at the apical epithelial interface while host cells are oxygenated basolaterally. This platform enables epithelial-anaerobe co-culture for ~48 h, and we demonstrate its utility by evaluating reciprocal interactions between the oxygen-sensitive anaerobic bacterium, Fusobacterium nucleatum, and oxygen-demanding airway epithelial cells at the transcriptional level. Using bulk RNAseq, we demonstrate that epithelial colonization results in altered gene expression by F. nucleatum, highlighted by the differential expression of genes associated with virulence, ethanolamine and lysine metabolism, metal uptake, and other transport processes. We also combine DOAC with single-cell RNA sequencing to reveal a cell type-specific transcriptional response of the airway epithelium to F. nucleatum infection, including the increased expression of inflammatory marker genes and cancer-associated pathways. Together, these data illustrate the versatility of DOAC while revealing new insights into anaerobe-host interactions and their mechanistic contributions to airway disease pathophysiology.IMPORTANCEConflicting oxygen demands between anaerobes and host cells present a significant barrier to in vitro modeling of how these cell types interact. To this end, the significance of our dual oxic-anoxic culture (DOAC) approach lies in its ability to maintain anaerobe and epithelial viability during co-culture, paving the way for new insights into the role(s) of anaerobic microbiota in disease. We use DOAC to interrogate reciprocal interactions between the airway epithelium and Fusobacterium nucleatum-an anaerobic commensal with pathogenic potential. Given its link to a range of diseases, from localized infections to various cancers, these data showing how F. nucleatum bacterium re-shapes its metabolism and virulence upon epithelial colonization provide new mechanistic insight into F. nucleatum physiology and how the host responds. We use F. nucleatum as our model, but the DOAC platform motivates additional studies of the gut, lung, and oral cavity, where host-anaerobe interactions and the underlying mechanisms of pathogenesis are poorly understood.
Collapse
Affiliation(s)
- Patrick J. Moore
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kayla Hoffman
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sara Ahmed
- Department of Microbiology and Immunology, SUNY at Buffalo, Buffalo, New York, USA
| | - Joshua R. Fletcher
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, North Carolina, USA
| | - Talia D. Wiggen
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sarah K. Lucas
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biology, Syracuse University, Syracuse, New York, USA
| | - Sabrina J. Arif
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Adam J. Gilbertsen
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Leslie A. Kent
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jessica K. Fiege
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ryan A. Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Scott M. O'Grady
- Department of Animal Science, University of Minnesota, Saint Paul, Minnesota, USA
| | - Ryan C. Hunter
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Microbiology and Immunology, SUNY at Buffalo, Buffalo, New York, USA
| |
Collapse
|
4
|
Muntzar R, Carvajal-Agudelo JD, Tench L, Ofosua Apienti TA, Franz-Odendaal TA. BMP and WNT signaling are involved in tracheal cartilage development in chicken embryos. Dev Biol 2025; 524:69-79. [PMID: 40324741 DOI: 10.1016/j.ydbio.2025.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
The respiratory system consists of the tracheal tube which extends from the back of the mouth, down the neck and branches into the bronchial tubes of the lungs, which are the major sites of respiration. Cartilaginous rings are present along the length of the trachea and are essential for keeping the airways open. Understanding trachea and cartilage development is crucial to better understanding respiratory disorders and diseases. We analyzed cartilage development across different chicken embryonic stages using a combination of cellular and molecular biology tools. We found that chondrogenic condensations are barely visible histologically from HH35 (E8.5) while differentiated cartilage rings are distinct by HH36 (E10). In situ hybridisation data shows that Bmp2 is expressed in a ring-like pattern along the length of the tracheal tube from HH36. We further show that BMP2, but not BMP4, colocalises with COL2A1. qPCR data shows that genes such as COL2A1, and WNT are substantially upregulated between HH34 to HH37, while other genes (e.g. SHH, TBX4/5) are downregulated. Within this three to four day period, we detected a reduction in SHH and WNT during cartilage condensation formation and an upregulation of COL2A1 and WNT during overt cartilage differentiation. We next used a known Bmp2 type 1 receptor inhibitor (DMH1) to attempt to disrupt tracheal cartilage development. Following inhibition, we observed a widening and lengthening of the tracheal cartilage rings with some narrowing of the spaces between the rings. Our gene expression analysis reveals significant increases in COL2A1, WNT, and BMP4 following BMP receptor blocking compared to sham controls and indicates that these genes are important for the molecular response that results in patterning the tracheal cartilage ring morphology. This study provides a framework to further understand development of these important airway cartilages in the avian model.
Collapse
Affiliation(s)
- Romman Muntzar
- Department of Biology, Mount Saint Vincent University, Halifax, Nova Scotia, B3M 2J6, Canada
| | | | - Langston Tench
- Department of Biology, Mount Saint Vincent University, Halifax, Nova Scotia, B3M 2J6, Canada
| | | | - Tamara A Franz-Odendaal
- Department of Biology, Mount Saint Vincent University, Halifax, Nova Scotia, B3M 2J6, Canada.
| |
Collapse
|
5
|
Reding K, Pick L. Recent approaches lead to a deeper understanding of diverse segmentation mechanisms in insects, with a focus on the pair-rule genes. CURRENT OPINION IN INSECT SCIENCE 2025; 68:101317. [PMID: 39638284 PMCID: PMC11875919 DOI: 10.1016/j.cois.2024.101317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The division of the insect embryo into repeated units - segments - is a fundamental feature of the body plan. The genes controlling this process in Drosophila melanogaster were identified in genetic screens and characterized in that species in numerous studies in the 1980s and 1990s. These genes form a well-established hierarchy and have been leveraged to examine gene regulation, transcriptional machinery, chromatin structure, and more. Much of the genetic toolkit identified in Drosophila is highly conserved throughout the animal kingdom, spearheading the field of evolutionary developmental biology or Evo-Devo. Accordingly, a 'Drosophila-centric' approach has examined the evolutionary conservation of orthologs of Drosophila segmentation genes in closely and distantly related insects. Here, we report on progress in both Drosophila and emerging model insects in recent years (2022 to present), with much of the new research related to the pair-rule subset of segmentation genes. We highlight new findings on 'classic' Drosophila genes, revealing unexpected roles of genes and cis-regulatory elements in this species. We further report on the expanding knowledge about mechanisms regulating to segmentation in emerging model insects that are distantly related to Drosophila, including those that pattern segments sequentially. We also describe technical advances in both Drosophila and nonmodel species that are currently progressing research in this field.
Collapse
Affiliation(s)
- Katie Reding
- Department of Entomology, University of Maryland, 4291 Fieldhouse Drive, College Park, MD 20742, USA
| | - Leslie Pick
- Department of Entomology, University of Maryland, 4291 Fieldhouse Drive, College Park, MD 20742, USA.
| |
Collapse
|
6
|
Passini FS, Bornstein B, Rubin S, Kuperman Y, Krief S, Masschelein E, Mehlman T, Brandis A, Addadi Y, Shalom SHO, Richter EA, Yardeni T, Tirosh A, De Bock K, Zelzer E. Piezo2 in sensory neurons regulates systemic and adipose tissue metabolism. Cell Metab 2025; 37:987-1000.e6. [PMID: 39919739 DOI: 10.1016/j.cmet.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025]
Abstract
Systemic metabolism ensures energy homeostasis through inter-organ crosstalk regulating thermogenic adipose tissue. Unlike the well-described inductive role of the sympathetic system, the inhibitory signal ensuring energy preservation remains poorly understood. Here, we show that, via the mechanosensor Piezo2, sensory neurons regulate morphological and physiological properties of brown and beige fat and prevent systemic hypermetabolism. Targeting runt-related transcription factor 3 (Runx3)/parvalbumin (PV) sensory neurons in independent genetic mouse models resulted in a systemic metabolic phenotype characterized by reduced body fat and increased insulin sensitivity and glucose tolerance. Deletion of Piezo2 in PV sensory neurons reproduced the phenotype, protected against high-fat-diet-induced obesity, and caused adipose tissue browning and beiging, likely driven by elevated norepinephrine levels. Finding that brown and beige fat are innervated by Runx3/PV sensory neurons expressing Piezo2 suggests a model in which mechanical signals, sensed by Piezo2 in sensory neurons, protect energy storage and prevent a systemic hypermetabolic phenotype.
Collapse
Affiliation(s)
- Fabian S Passini
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Bavat Bornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Evi Masschelein
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- MICC Cell Observatory, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Huri-Ohev Shalom
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Erik A Richter
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Tal Yardeni
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amir Tirosh
- The Dalia and David Arabov Endocrinology and Diabetes Research Center, Institute of Endocrinology, Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Katrien De Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
7
|
Wong ZW, New SY. Recent Advances in Biosensors Based on Hybridization Chain Reaction and Silver Nanoclusters. SMALL METHODS 2025; 9:e2401436. [PMID: 39757735 DOI: 10.1002/smtd.202401436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/22/2024] [Indexed: 01/07/2025]
Abstract
Hybridization chain reaction (HCR) and DNA-templated silver nanoclusters (AgNCs) have emerged as powerful tools in biosensing. HCR enables cascade amplification through programmable DNA interactions, while DNA-AgNCs serve as transducing units with unique fluorogenic and electrochemical properties. Integrating these components into a hybrid sensor could significantly enhance sensing capabilities across various fields. Nonetheless, limited studies and the lack of systematic guidelines for HCR-AgNCs systems have hindered research progress, despite their potential. This review aims to address this gap by providing a comprehensive overview of HCR-AgNCs biosensors, facilitating further innovation in this field. The working principles, performance factors, and complementary features are discussed. Thereafter, reported HCR-AgNCs studies are assessed, emphasizing their distinct sensing mechanisms (e.g., fluorogenic, electrochemical), applications across various fields, and challenges in adopting the hybrid sensors. Drawing from the experience developing multiple HCR-AgNCs sensors, insights and guidelines for designing and developing HCR-AgNCs systems are provided for future researchers. Finally, prospective directions in HCR-AgNCs research, including multiplex assays and integration with emerging technologies, are explored to guide future advancements. The synergistic combination of HCR and AgNCs as a hybrid biosensor holds promise for addressing pressing challenges in healthcare, environmental monitoring, and beyond, paving the way for next-generation biosensing technologies.
Collapse
Affiliation(s)
- Zheng Wei Wong
- School of Pharmacy, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor Darul Ehsan, 43500, Malaysia
| | - Siu Yee New
- School of Pharmacy, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor Darul Ehsan, 43500, Malaysia
| |
Collapse
|
8
|
Hutchins NT, Meziane M, Lu C, Mitalipova M, Fischer D, Li P. Reconstructing signaling histories of single cells via perturbation screens and transfer learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.16.643448. [PMID: 40166200 PMCID: PMC11957020 DOI: 10.1101/2025.03.16.643448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Manipulating the signaling environment is an effective approach to alter cellular states for broad-ranging applications, from engineering tissues to treating diseases. Such manipulation requires knowing the signaling states and histories of the cells in situ , for which high-throughput discovery methods are lacking. Here, we present an integrated experimental-computational framework that learns signaling response signatures from a high-throughput in vitro perturbation atlas and infers combinatorial signaling activities in in vivo cell types with high accuracy and temporal resolution. Specifically, we generated signaling perturbation atlas across diverse cell types/states through multiplexed sequential combinatorial screens on human pluripotent stem cells. Using the atlas to train IRIS, a neural network-based model, and predicting on mouse embryo scRNAseq atlas, we discovered global features of combinatorial signaling code usage over time, identified biologically meaningful heterogeneity of signaling states within each cell type, and reconstructed signaling histories along diverse cell lineages. We further demonstrated that IRIS greatly accelerates the optimization of stem cell differentiation protocols by drastically reducing the combinatorial space that needs to be tested. This framework leads to the revelation that different cell types share robust signal response signatures, and provides a scalable solution for mapping complex signaling interactions in vivo to guide targeted interventions.
Collapse
|
9
|
Dong Y, Qi L, Zhao F, Chen Y, Liang L, Wang J, Zhao W, Wang F, Xu H. Uncovering dynamic transcriptional regulation of methanogenesis via single-cell imaging of archaeal gene expression. Nat Commun 2025; 16:2255. [PMID: 40050284 PMCID: PMC11885431 DOI: 10.1038/s41467-025-57159-0] [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] [Received: 05/16/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
Archaeal methanogenesis is a dynamic process regulated by various cellular and environmental signals. However, understanding this regulation is technically challenging due to the difficulty of measuring gene expression dynamics in individual archaeal cells. Here, we develop a multi-round hybridization chain reaction (HCR)-assisted single-molecule fluorescence in situ hybridization (FISH) method to quantify the transcriptional dynamics of 12 genes involved in methanogenesis in individual cells of Methanococcoides orientis. Under optimal growth condition, most of these genes appear to be expressed in a temporal order matching metabolic reaction order. Interestingly, an important environmental factor, Fe(III), stimulates cellular methane production without upregulating methanogenic gene expression, likely through a Fenton-reaction-triggered mechanism. Through single-cell clustering and kinetic analyses, we associate these gene expression patterns to a dynamic mixture of distinct cellular states, potentially regulated by a set of shared factors. Our work provides a quantitative framework for uncovering the mechanisms of metabolic regulation in archaea.
Collapse
Affiliation(s)
- Yijing Dong
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, China
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Lanting Qi
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, China
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Fei Zhao
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, China
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Chen
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Lewen Liang
- Key Laboratory of Polar Ecosystem and Climate Change, Ministry of Education, and School of Oceanography, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wang
- Key Laboratory of Polar Ecosystem and Climate Change, Ministry of Education, and School of Oceanography, Shanghai Jiao Tong University, Shanghai, China
| | - Weishu Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Fengping Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
- Key Laboratory of Polar Ecosystem and Climate Change, Ministry of Education, and School of Oceanography, Shanghai Jiao Tong University, Shanghai, China.
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong, China.
| | - Heng Xu
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
10
|
Van Emmenis L, Mòdol-Caballero G, Harford-Wright E, Power A, Cattin AL, White IJ, Casal G, Boal-Carvalho I, Bennett CL, Lloyd AC. Identification of CCL3 as a Schwann cell chemotactic factor essential for nerve regeneration. Cell Rep 2025; 44:115322. [PMID: 39960833 DOI: 10.1016/j.celrep.2025.115322] [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: 09/11/2024] [Revised: 12/14/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
Peripheral nerves regenerate following injury, in contrast to those of the central nervous system. This involves the collective migration of Schwann cell (SC) cords, which transport regrowing axons across the wound site. The SC cords migrate along a newly formed vasculature, which bridges the wound site in response to vascular endothelial growth factor, secreted by hypoxic macrophages. However, the directional signals by which SC cords navigate the long distances across the wound, in the absence of those that guide axons during development, remain unknown. Here, we identify CCL3 as the SC chemotactic factor, secreted by hypoxic macrophages, responsible for this process. We show that CCL3 promotes collective SC migration and axonal regrowth in vivo and, using genetic mouse models and widely used CCL3 inhibitors, that CCL3 is essential for effective nerve regeneration. These findings have therapeutic implications for both promoting nerve repair and inhibiting the aberrant nerve growth associated with trauma and disease.
Collapse
Affiliation(s)
- Lucie Van Emmenis
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Guillem Mòdol-Caballero
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elizabeth Harford-Wright
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Alex Power
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Anne-Laure Cattin
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ian J White
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Giulia Casal
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Inês Boal-Carvalho
- Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Clare L Bennett
- Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
11
|
Plygawko AT, Stephan-Otto Attolini C, Pitsidianaki I, Cook DP, Darby AC, Campbell K. The Drosophila adult midgut progenitor cells arise from asymmetric divisions of neuroblast-like cells. Dev Cell 2025; 60:429-446.e6. [PMID: 39532106 DOI: 10.1016/j.devcel.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/21/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
The Drosophila adult midgut progenitor cells (AMPs) give rise to all cells in the adult midgut epithelium, including the intestinal stem cells (ISCs). While they share many characteristics with the ISCs, it remains unclear how they are generated in the early embryo. Here, we show that they arise from a population of endoderm cells, which exhibit multiple similarities with Drosophila neuroblasts. These cells, which we have termed endoblasts, are patterned by homothorax (Hth) and undergo asymmetric divisions using the same molecular machinery as neuroblasts. We also show that the conservation of this molecular machinery extends to the generation of the enteroendocrine lineages. Parallels have previously been drawn between the pupal ISCs and larval neuroblasts. Our results suggest that these commonalities exist from the earliest stages of specification of progenitor cells of the intestinal and nervous systems and may represent an ancestral pathway for multipotent progenitor cell specification.
Collapse
Affiliation(s)
- Andrew T Plygawko
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ioanna Pitsidianaki
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - David P Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Alistair C Darby
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Kyra Campbell
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
12
|
Marchini M, Keller G, Khan N, Shah R, Saliceti Galarza A, Starr KB, Apostopoulos A, Sanger TJ. Sonic hedgehog and fibroblast growth factor 8 regulate the evolution of amniote facial proportions. Commun Biol 2025; 8:84. [PMID: 39827295 PMCID: PMC11742871 DOI: 10.1038/s42003-025-07522-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Amniote skulls are diverse in shape and skeletal composition, which is the basis of much adaptive diversification within this clade. Major differences in skull shape are established early in development, at a critical developmental interval spanning the initial outgrowth and fusion of the facial processes. In birds, this is orchestrated by domains of Shh and Fgf8 expression, known as the frontonasal ectodermal zone (FEZ). It is unclear whether this model of facial development applies to species with diverse facial skeletons, especially species possessing a skull morphology representative of early amniotes. By investigating facial morphogenesis in the lizard, Anolis sagrei, we show that reptilian skull development is driven by the same genes as mammals and birds, but the manner in which those genes regulate facial development is clade-specific. These genes are not expressed in the frontal-nasal prominence, the region of the avian FEZ. Downregulating Shh and Fgf8 signaling disrupts normal facial development, but in pathway-specific ways. Our results demonstrate that early facial morphogenesis in lizards does not conform to the FEZ model. Lizard skull development may be more representative of the ancestral amniote than other model species with highly derived facial skeletons suggesting that the FEZ may be an avian-specific novelty.
Collapse
Affiliation(s)
- Marta Marchini
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Greta Keller
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Naaz Khan
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Rushabh Shah
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | | | | | | | - Thomas J Sanger
- Department of Biology, Loyola University Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Pisterzi P, Martinez Mir C, Dahri O, de Poorter I, Batlles Parera S, Dostanić M, Mastrangeli M, Mummery C, Geijsen N, Sage F. Spatial analysis of transcript and protein levels in skeletal muscle. STAR Protoc 2024; 5:103378. [PMID: 39388355 PMCID: PMC11736001 DOI: 10.1016/j.xpro.2024.103378] [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: 06/07/2024] [Revised: 07/19/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
Skeletal muscle spatial analyses have revealed unexpected regionalized gene expression patterns challenging the understanding of muscle as a homogeneous tissue. Here, we present a protocol for the spatial analysis of transcript and protein levels in murine skeletal muscle. We describe steps for tibialis anterior dissection, formaldehyde fixation, tissue chopper cutting, and hybridization chain reaction (HCR) detection and amplification. We then detail procedures for immunostaining, tissue clearing, and imaging. This protocol is easily adaptable to other tissues.
Collapse
Affiliation(s)
- Paola Pisterzi
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands
| | - Clara Martinez Mir
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands
| | - Ouafa Dahri
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands
| | - Isabel de Poorter
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands
| | - Sandra Batlles Parera
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands
| | - Milica Dostanić
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands
| | - Massimo Mastrangeli
- Microelectronics Department, Delft University of Technology, 2628 CD Delft, the Netherlands
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands
| | - Niels Geijsen
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands.
| | - Fanny Sage
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, the Netherlands.
| |
Collapse
|
14
|
Forbes Beadle L, Sutcliffe C, Ashe HL. A simple MiMIC-based approach for tagging endogenous genes to visualise live transcription in Drosophila. Development 2024; 151:dev204294. [PMID: 39584418 DOI: 10.1242/dev.204294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Live imaging of transcription in the Drosophila embryo using the MS2 or PP7 systems is transforming our understanding of transcriptional regulation. However, insertion of MS2/PP7 stem-loops into endogenous genes requires laborious CRISPR genome editing. Here, we exploit the previously described Minos-mediated integration cassette (MiMIC) transposon system in Drosophila to establish a method for simply and rapidly inserting MS2/PP7 cassettes into any of the thousands of genes carrying a MiMIC insertion. In addition to generating a variety of stem-loop donor fly stocks, we have made new stocks expressing the complementary coat proteins fused to different fluorescent proteins. We show the utility of this MiMIC-based approach by MS2/PP7 tagging of endogenous genes and the long non-coding RNA roX1, then imaging their transcription in living embryos. We also present live transcription data from larval brains, the wing disc and ovary, thereby extending the tissues that can be studied using the MS2/PP7 system. Overall, this first high-throughput method for tagging mRNAs in Drosophila will facilitate the study of transcription dynamics of thousands of endogenous genes in a range of Drosophila tissues.
Collapse
Affiliation(s)
- Lauren Forbes Beadle
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Catherine Sutcliffe
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Hilary L Ashe
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
15
|
Steyn C, Mishi R, Fillmore S, Verhoog MB, More J, Rohlwink UK, Melvill R, Butler J, Enslin JMN, Jacobs M, Sauka-Spengler T, Greco M, Quiñones S, Dulla CG, Raimondo JV, Figaji A, Hockman D. A temporal cortex cell atlas highlights gene expression dynamics during human brain maturation. Nat Genet 2024; 56:2718-2730. [PMID: 39567748 PMCID: PMC11631765 DOI: 10.1038/s41588-024-01990-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
The human brain undergoes protracted postnatal maturation, guided by dynamic changes in gene expression. Most studies exploring these processes have used bulk tissue analyses, which mask cell-type-specific gene expression dynamics. Here, using single-nucleus RNA sequencing on temporal lobe tissue, including samples of African ancestry, we build a joint pediatric and adult atlas of 75 cell subtypes, which we verify with spatial transcriptomics. We explore the differences between pediatric and adult cell subtypes, revealing the genes and pathways that change during brain maturation. Our results highlight excitatory neuron subtypes, including the LTK and FREM subtypes, that show elevated expression of genes associated with cognition and synaptic plasticity in pediatric tissue. The resources we present here improve our understanding of the brain during its development and contribute to global efforts to build an inclusive brain cell map.
Collapse
Affiliation(s)
- Christina Steyn
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ruvimbo Mishi
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Stephanie Fillmore
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Matthijs B Verhoog
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Jessica More
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ursula K Rohlwink
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Roger Melvill
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - James Butler
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Johannes M N Enslin
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Muazzam Jacobs
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Department of Pathology University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Tatjana Sauka-Spengler
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Maria Greco
- Single Cell Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Sadi Quiñones
- Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Graduate School of Biomedical Science, Tufts University School of Medicine, Boston, MA, USA
| | - Chris G Dulla
- Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Joseph V Raimondo
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Anthony Figaji
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Dorit Hockman
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa.
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
16
|
Olivares-Abril J, Joha J, Lee JY, Davis I. Optimization of hybridization chain reaction for imaging single RNA molecules in Drosophila larvae. Fly (Austin) 2024; 18:2409968. [PMID: 39351922 PMCID: PMC11446410 DOI: 10.1080/19336934.2024.2409968] [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: 06/06/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
In situ hybridization techniques are powerful methods for exploring gene expression in a wide range of biological contexts, providing spatial information that is most often lost in traditional biochemical techniques. However, many in situ hybridization methods are costly and time-inefficient, particularly for screening-based projects that follow on from single-cell RNA sequencing data, which rely on of tens of custom-synthetized probes against each specific RNA of interest. Here we provide an optimized pipeline for Hybridization Chain Reaction (HCR)-based RNA visualization, including an open-source code for optimized probe design. Our method achieves high specificity and sensitivity with the option of multiplexing using only five pairs of probes, which greatly lowers the cost and time of the experiment. These features of our HCR protocol are particularly useful and convenient for projects involving screening several genes at medium throughput, especially as the method include an amplification step, which makes the signal readily visible at low magnification imaging.
Collapse
Affiliation(s)
| | - Jana Joha
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Jeffrey Y Lee
- School of Molecular Biosciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Ilan Davis
- School of Molecular Biosciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
17
|
Kothurkar AA, Patient GS, Noel NCL, Krzywańska AM, Carr BJ, Chu CJ, MacDonald RB. 'Iterative Bleaching Extends Multiplexity' facilitates simultaneous identification of all major retinal cell types. J Cell Sci 2024; 137:jcs263407. [PMID: 39540305 PMCID: PMC11827602 DOI: 10.1242/jcs.263407] [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: 06/26/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
To understand the multicellular composition of tissues, and how it is altered during development, ageing and/or disease, we must visualise the complete cellular landscape. Currently, this is hindered by our limited ability to combine multiple cellular markers. To overcome this, we adapted a highly multiplexed immunofluorescence (IF) technique called 'Iterative Bleaching Extends Multiplexity' (IBEX) to the zebrafish retina. We optimised fluorescent antibody micro-conjugation to perform sequential rounds of labelling on a single tissue to simultaneously visualise all major retinal cell types with 11 cell-specific antibodies. We further adapted IBEX to be compatible with fluorescent transgenic reporter lines, in situ hybridisation chain reaction (HCR), and whole-mount immunofluorescence (WMIF). We applied IBEX at multiple stages to study the spatial and temporal relationships between glia and neurons during retinal development. Finally, we demonstrate the utility of IBEX across species by testing it on the turquoise killifish (Nothobranchius furzeri) and African clawed frog (Xenopus laevis) to glean large amounts of information from precious tissues. These techniques will revolutionise our ability to visualise multiple cell types in any organism where antibodies are readily available.
Collapse
Affiliation(s)
| | - Gregory S. Patient
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Nicole C. L. Noel
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | | | - Brittany J. Carr
- Department of Ophthalmology & Visual Sciences, University of Alberta, Edmonton, AB T5H 3V9, Canada
| | - Colin J. Chu
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Ryan B. MacDonald
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| |
Collapse
|
18
|
Lion AT, Bodine SM, McCutcheon KR, Ghogale M, Chandragiri S, Abayawardena D, Shrestha BD, Descoteaux A, Alvarez K, Balkman JA, Cocke B, Wikramanayake AH, Schlezinger J, Wong JY, Prakash VN, Bradham CA. PFAS Compounds PFOA and Gen X are Teratogenic to Sea Urchin Embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624751. [PMID: 39605628 PMCID: PMC11601578 DOI: 10.1101/2024.11.21.624751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Per-and polyfluorinated substances (PFAS) are synthetic chemicals that are used to make fluoropolymer coatings found in many products, such as non-stick pans, clothing, cosmetics, and food packaging. These highly persistent molecules are known as "forever chemicals" since they neither degrade environmentally nor break down enzymatically within biological systems. PFAS compounds readily contaminate water sources, and as a result, certain PFAS molecules have bioaccumulated in exposed species including humans. The purpose of this study was to define the effect of two PFAS molecules, the ostensibly more toxic perfluorooctanoic acid (PFOA) and the more recent, reportedly safer chemical hexafluoropropylene oxide dimer acid (Gen X), on the development of Lytechinus variegatus sea urchin embryos. We examined the effects of PFOA and Gen X on development and patterning using morphological analysis, immunostaining, HCR-FISH, and Particle Image Velocimetry (PIV). The results show that both PFAS compounds are teratogenic to sea urchin embryos. PFOA and Gen X each function at different intervals during development and provoke distinct phenotypic and gene expression outcomes. Despite beliefs that Gen X would be a safer alternative, our findings indicate that Gen X has earlier and more severe effects on endomesoderm and dorsal-ventral axis specification, neural development and function, and pattern formation compared to PFOA. These results illustrate the dangerous teratogenic potential of environmentally accumulating PFAS like Gen X, underscoring the negative ecological implications that accompany continuing commercial and industrial use of PFAS in the absence of remediation strategies.
Collapse
Affiliation(s)
- Alexandra T. Lion
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston MA, USA
| | | | | | - Mayank Ghogale
- Bioinformatics Program, Boston University, Boston MA, USA
| | | | | | | | - Abigail Descoteaux
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston MA, USA
- Biological Design Center, College of Engineering, Boston University, Boston MA, USA
| | - Kathryn Alvarez
- Department of Physics, University of Miami, Coral Gables FL, USA
| | | | - Breelyn Cocke
- Department of Physics, University of Miami, Coral Gables FL, USA
| | | | | | - Joyce Y. Wong
- Department of Biomedical Engineering, Boston University, Boston MA, USA
| | - Vivek N. Prakash
- Department of Physics, University of Miami, Coral Gables FL, USA
- Department of Biology, University of Miami, Coral Gables FL, USA
- Department of Marine Biology and Ecology, University of Miami, Miami FL, USA
| | - Cynthia A. Bradham
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston MA, USA
- Biology Department, Boston University, Boston MA, USA
- Bioinformatics Program, Boston University, Boston MA, USA
- Biological Design Center, College of Engineering, Boston University, Boston MA, USA
| |
Collapse
|
19
|
McDonald BD, Massri AJ, Berrio A, Byrne M, McClay DR, Wray GA. Contrasting the development of larval and adult body plans during the evolution of biphasic lifecycles in sea urchins. Development 2024; 151:dev203015. [PMID: 39465623 PMCID: PMC11529275 DOI: 10.1242/dev.203015] [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: 04/30/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024]
Abstract
Biphasic lifecycles are widespread among animals, but little is known about how the developmental transition between larvae and adults is regulated. Sea urchins are a unique system for studying this phenomenon because of the stark differences between their bilateral larval and pentaradial adult body plans. Here, we use single-cell RNA sequencing to analyze the development of Heliocidaris erythrogramma (He), a sea urchin species with an accelerated, non-feeding mode of larval development. The sequencing time course extends from embryogenesis to roughly a day before the onset of metamorphosis in He larvae, which is a period that has not been covered by previous datasets. We find that the non-feeding developmental strategy of He is associated with several changes in the specification of larval cell types compared to sea urchins with feeding larvae, such as the loss of a larva-specific skeletal cell population. Furthermore, the development of the larval and adult body plans in sea urchins may utilize largely different sets of regulatory genes. These findings lay the groundwork for extending existing developmental gene regulatory networks to cover additional stages of biphasic lifecycles.
Collapse
Affiliation(s)
| | | | | | - Maria Byrne
- School of Life and Environmental Sciences, A11, University of Sydney, Sydney, NSW, 2006, Australia
| | - David R. McClay
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Gregory A. Wray
- Department of Biology, Duke University, Durham, NC 27708, USA
| |
Collapse
|
20
|
Chang Y, Liu J, Guo M, Ouyang W, Yan J, Xiong L, Li X. Drought-responsive dynamics of H3K9ac-marked 3D chromatin interactions are integrated by OsbZIP23-associated super-enhancer-like promoter regions in rice. Genome Biol 2024; 25:262. [PMID: 39390531 PMCID: PMC11465533 DOI: 10.1186/s13059-024-03408-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND In response to drought stress (DS), plants undergo complex processes that entail significant transcriptome reprogramming. However, the intricate relationship between the dynamic alterations in the three-dimensional (3D) genome and the modulation of gene co-expression in drought responses remains a relatively unexplored area. RESULTS In this study, we reconstruct high-resolution 3D genome maps based on genomic regions marked by H3K9ac, an active histone modification that dynamically responds to soil water variations in rice. We discover a genome-wide disconnection of 3D genome contact upon DS with over 10,000 chromatin loops lost, which are partially recovered in the subsequent re-watering. Loops integrating promoter-promoter interactions (PPI) contribute to gene expression in addition to basal H3K9ac modifications. Moreover, H3K9ac-marked promoter regions with high affinities in mediating PPIs, termed as super-promoter regions (SPRs), integrate spatially clustered PPIs in a super-enhancer-like manner. Interestingly, the knockout mutation of OsbZIP23, a well-defined DS-responsive transcription factor, leads to the disassociation of over 80% DS-specific PPIs and decreased expression of the corresponding genes under DS. As a case study, we show how OsbZIP23 integrates the PPI cluster formation and the co-expression of four dehydrin genes, RAB16A-D, through targeting the RAB16C SPR in a stress signaling-dependent manner. CONCLUSIONS Our high-resolution 3D genome maps unveil the principles and details of dynamic genome folding in response to water supply variations and illustrate OsbZIP23 as an indispensable integrator of the yet unique 3D genome organization that is essential for gene co-expression under DS in rice.
Collapse
Affiliation(s)
- Yu Chang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jiahan Liu
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Minrong Guo
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Weizhi Ouyang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jiapei Yan
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lizhong Xiong
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Xingwang Li
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
21
|
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
|
22
|
Belew MD, Chien E, Wong M, Michael WM. The TOP-2/condensin II axis silences transcription during germline specification in C. elegans. G3 (BETHESDA, MD.) 2024; 14:jkae236. [PMID: 39358855 PMCID: PMC11631511 DOI: 10.1093/g3journal/jkae236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 09/30/2024] [Indexed: 10/04/2024]
Abstract
In C. elegans, the germline is specified via a preformation mechanism that relies on the PIE-1 protein's ability to globally silence mRNA transcription in germline precursor cells, also known as the P lineage. Recent work from our group has identified additional genome silencing events in C. elegans during oogenesis and in starved L1 larvae, and these require the condensin II complex, topoisomerase II (TOP-2), and components of the H3K9me/heterochromatin pathway. Interestingly, silencing in oocytes also requires PIE-1, but this is not the case in starved L1s. Here, we ask if additional genome silencing components besides PIE-1 are required to repress gene expression in the P lineage of early embryos, and we find that condensin II and TOP-2 are required and the H3K9me/heterochromatin pathway is not. We show that depletion of TOP-2/condensin II activates the normally suppressed RNA polymerase II to inappropriately transcribe somatic genes in the P lineage. We also present evidence that while both PIE-1 and TOP-2/condensin II are required for genome silencing in the P lineage, PIE-1 can silence transcription independently of TOP-2/condensin II when misexpressed in somatic cells. Thus, in oocytes, all three genome silencing systems (TOP-2/condensin II, H3K9me, and PIE-1) are operational while in both early embryos and starved L1s two of the three are active. Our data show that multiple, redundantly acting genome silencing mechanisms act in a mix and match manner to repress transcription at different developmental stages in the C. elegans germline.
Collapse
Affiliation(s)
- Mezmur D Belew
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Emilie Chien
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Matthew Wong
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - W Matthew Michael
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
23
|
Tate HM, Barone V, Schrankel CS, Hamdoun A, Lyons DC. Localization and origins of juvenile skeletogenic cells in the sea urchin Lytechinuspictus. Dev Biol 2024; 514:12-27. [PMID: 38862087 DOI: 10.1016/j.ydbio.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024]
Abstract
The development of the sea urchin larval body plan is well understood from extensive studies of embryonic patterning. However, fewer studies have investigated the late larval stages during which the unique pentaradial adult body plan develops. Previous work on late larval development highlights major tissue changes leading up to metamorphosis, but the location of specific cell types during juvenile development is less understood. Here, we improve on technical limitations by applying highly sensitive hybridization chain reaction fluorescent in situ hybridization (HCR-FISH) to the fast-developing and transparent sea urchin Lytechinus pictus, with a focus on skeletogenic cells. First, we show that HCR-FISH can be used in L. pictus to precisely localize skeletogenic cells in the rudiment. In doing so, we provide a detailed staging scheme for the appearance of skeletogenic cells around the rudiment prior to and during biomineralization and show that many skeletogenic cells unassociated with larval rods localize outside of the rudiment prior to localizing inside. Second, we show that downstream biomineralization genes have similar expression patterns during larval and juvenile skeletogenesis, suggesting some conservation of skeletogenic mechanisms during development between stages. Third, we find co-expression of blastocoelar and skeletogenic cell markers around juvenile skeleton located outside of the rudiment, which is consistent with data showing that cells from the non-skeletogenic mesoderm embryonic lineage contribute to the juvenile skeletogenic cell lineage. This work sets the foundation for subsequent studies of other cell types in the late larva of L. pictus to better understand juvenile body plan development, patterning, and evolution.
Collapse
Affiliation(s)
- Heidi M Tate
- Scripps Institution of Oceanography, UC San Diego, La Jolla, CA, USA
| | - Vanessa Barone
- Scripps Institution of Oceanography, UC San Diego, La Jolla, CA, USA
| | - Catherine S Schrankel
- Scripps Institution of Oceanography, UC San Diego, La Jolla, CA, USA; San Diego State University, San Diego, CA, USA
| | - Amro Hamdoun
- Scripps Institution of Oceanography, UC San Diego, La Jolla, CA, USA
| | - Deirdre C Lyons
- Scripps Institution of Oceanography, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
24
|
Hake KH, West PT, McDonald K, Laundon D, Reyes-Rivera J, Garcia De Las Bayonas A, Feng C, Burkhardt P, Richter DJ, Banfield JF, King N. A large colonial choanoflagellate from Mono Lake harbors live bacteria. mBio 2024; 15:e0162324. [PMID: 39140743 PMCID: PMC11389367 DOI: 10.1128/mbio.01623-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
As the closest living relatives of animals, choanoflagellates offer insights into the ancestry of animal cell physiology. Here, we report the isolation and characterization of a colonial choanoflagellate from Mono Lake, California. The choanoflagellate forms large spherical colonies that are an order of magnitude larger than those formed by the closely related choanoflagellate Salpingoeca rosetta. In cultures maintained in the laboratory, the lumen of the spherical colony is filled with a branched network of extracellular matrix and colonized by bacteria, including diverse Gammaproteobacteria and Alphaproteobacteria. We propose to erect Barroeca monosierra gen. nov., sp. nov. Hake, Burkhardt, Richter, and King to accommodate this extremophile choanoflagellate. The physical association between bacteria and B. monosierra in culture presents a new experimental model for investigating interactions among bacteria and eukaryotes. Future work will investigate the nature of these interactions in wild populations and the mechanisms underpinning the colonization of B. monosierra spheres by bacteria. IMPORTANCE The diversity of organisms that live in the extreme environment of Mono Lake (California, USA) is limited. We sought to investigate whether the closest living relatives of animals, the choanoflagellates, exist in Mono Lake, a hypersaline, alkaline, arsenic-rich environment. We repeatedly isolated members of a new species of choanoflagellate, which we have named Barroeca monosierra. Characterization of B. monosierra revealed that it forms large spherical colonies containing diverse co-isolated bacteria, providing an opportunity to investigate mechanisms underlying physical associations between eukaryotes and bacteria.
Collapse
Affiliation(s)
- K H Hake
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - P T West
- Department of Environmental Science, Policy, & Management, University of California, Berkeley, California, USA
| | - K McDonald
- Electron Microscopy Laboratory, University of California, Berkeley, California, USA
| | - D Laundon
- Marine Biological Association of the United Kingdom, Plymouth, United Kingdom
| | - J Reyes-Rivera
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - A Garcia De Las Bayonas
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - C Feng
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - P Burkhardt
- Marine Biological Association of the United Kingdom, Plymouth, United Kingdom
- Michael Sars Centre, University of Bergen, Bergen, Norway
| | - D J Richter
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Barcelona, Spain
| | - J F Banfield
- Department of Environmental Science, Policy, & Management, University of California, Berkeley, California, USA
| | - N King
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| |
Collapse
|
25
|
Maltby CJ, Krans A, Grudzien SJ, Palacios Y, Muiños J, Suárez A, Asher M, Willey S, Van Deynze K, Mumm C, Boyle AP, Cortese A, Ndayisaba A, Khurana V, Barmada SJ, Dijkstra AA, Todd PK. AAGGG repeat expansions trigger RFC1-independent synaptic dysregulation in human CANVAS neurons. SCIENCE ADVANCES 2024; 10:eadn2321. [PMID: 39231235 PMCID: PMC11373605 DOI: 10.1126/sciadv.adn2321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Cerebellar ataxia with neuropathy and vestibular areflexia syndrome (CANVAS) is a recessively inherited neurodegenerative disorder caused by intronic biallelic, nonreference CCCTT/AAGGG repeat expansions within RFC1. To investigate how these repeats cause disease, we generated patient induced pluripotent stem cell-derived neurons (iNeurons). CCCTT/AAGGG repeat expansions do not alter neuronal RFC1 splicing, expression, or DNA repair pathway function. In reporter assays, AAGGG repeats are translated into pentapeptide repeat proteins. However, these proteins and repeat RNA foci were not detected in iNeurons, and overexpression of these repeats failed to induce neuronal toxicity. CANVAS iNeurons exhibit defects in neuronal development and diminished synaptic connectivity that is rescued by CRISPR deletion of a single expanded AAGGG allele. These deficits were neither replicated by RFC1 knockdown in control iNeurons nor rescued by RFC1 reprovision in CANVAS iNeurons. These findings support a repeat-dependent but RFC1 protein-independent cause of neuronal dysfunction in CANVAS, with implications for therapeutic development in this currently untreatable condition.
Collapse
Affiliation(s)
- Connor J. Maltby
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Krans
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Ann Arbor Veterans Administration Healthcare, Ann Arbor, MI, USA
| | - Samantha J. Grudzien
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yomira Palacios
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Postbaccalaureate Research Education Program, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Muiños
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- UM SMART Undergraduate Summer Program, University of Michigan, Ann Arbor, MI, USA
| | - Andrea Suárez
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Postbaccalaureate Research Education Program, University of Michigan, Ann Arbor, MI, USA
| | - Melissa Asher
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Sydney Willey
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Kinsey Van Deynze
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Camille Mumm
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Alan P. Boyle
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Andrea Cortese
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Department of Brain and Behaviour Sciences, University of Pavia, Pavia 27100, Italy
| | - Alain Ndayisaba
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Vikram Khurana
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sami J. Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Anke A. Dijkstra
- Department of Pathology, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Peter K. Todd
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Ann Arbor Veterans Administration Healthcare, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Wang Y, Liu X, Zeng Y, Saka S, Xie W, Goldaracena I, Kohman R, Yin P, Church G. Multiplexed in situ protein imaging using DNA-barcoded antibodies with extended hybridization chain reactions. Nucleic Acids Res 2024; 52:e71. [PMID: 38966983 PMCID: PMC11347153 DOI: 10.1093/nar/gkae592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 05/23/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
Antibodies have long served as vital tools in biological and clinical laboratories for the specific detection of proteins. Conventional methods employ fluorophore or horseradish peroxidase-conjugated antibodies to detect signals. More recently, DNA-conjugated antibodies have emerged as a promising technology, capitalizing on the programmability and amplification capabilities of DNA to enable highly multiplexed and ultrasensitive protein detection. However, the nonspecific binding of DNA-conjugated antibodies has impeded the widespread adoption of this approach. Here, we present a novel DNA-conjugated antibody staining protocol that addresses these challenges and demonstrates superior performance in suppressing nonspecific signals compared to previously published protocols. We further extend the utility of DNA-conjugated antibodies for signal-amplified in situ protein imaging through the hybridization chain reaction (HCR) and design a novel HCR DNA pair to expand the HCR hairpin pool from the previously published 5 pairs to 13, allowing for flexible hairpin selection and higher multiplexing. Finally, we demonstrate highly multiplexed in situ protein imaging using these techniques in both cultured cells and tissue sections.
Collapse
Affiliation(s)
- Yu Wang
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Department of System Biology, Harvard Medical School, Boston, MA 02115, USA
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xiaoyu Liu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yitian Zeng
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sinem K Saka
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of System Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Wenxin Xie
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Isabel Goldaracena
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of System Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Richie E Kohman
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of System Biology, Harvard Medical School, Boston, MA 02115, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
27
|
Attenborough T, Rawlinson KA, Diaz Soria CL, Ambridge K, Sankaranarayanan G, Graham J, Cotton JA, Doyle SR, Rinaldi G, Berriman M. A single-cell atlas of the miracidium larva of Schistosoma mansoni reveals cell types, developmental pathways, and tissue architecture. eLife 2024; 13:RP95628. [PMID: 39190022 PMCID: PMC11349301 DOI: 10.7554/elife.95628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024] Open
Abstract
Schistosoma mansoni is a parasitic flatworm that causes the major neglected tropical disease schistosomiasis. The miracidium is the first larval stage of the life cycle. It swims and infects a freshwater snail, transforms into a mother sporocyst, where its stem cells generate daughter sporocysts that give rise to human-infective cercariae larvae. To understand the miracidium at cellular and molecular levels, we created a whole-body atlas of its ~365 cells. Single-cell RNA sequencing identified 19 transcriptionally distinct cell clusters. In situ hybridisation of tissue-specific genes revealed that 93% of the cells in the larva are somatic (57% neural, 19% muscle, 13% epidermal or tegument, 2% parenchyma, and 2% protonephridia) and 7% are stem. Whereas neurons represent the most diverse somatic cell types, trajectory analysis of the two main stem cell populations indicates that one of them is the origin of the tegument lineage and the other likely contains pluripotent cells. Furthermore, unlike the somatic cells, each of these stem populations shows sex-biased transcriptional signatures suggesting a cell-type-specific gene dosage compensation for sex chromosome-linked loci. The miracidium represents a simple developmental stage with which to gain a fundamental understanding of the molecular biology and spatial architecture of schistosome cells.
Collapse
Affiliation(s)
- Teresa Attenborough
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- School of Infection and Immunity, College of Medical, Veterinary & Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Kate A Rawlinson
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- Josephine Bay Paul Center, Marine Biological LaboratoryWoods HoleUnited States
| | | | - Kirsty Ambridge
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | | | - Jennie Graham
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | - James A Cotton
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary & Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Stephen R Doyle
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- Department of Life Sciences, Aberystwyth UniversityAberystwythUnited Kingdom
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- School of Infection and Immunity, College of Medical, Veterinary & Life Sciences, University of GlasgowGlasgowUnited Kingdom
| |
Collapse
|
28
|
Kawaguchi A, Wang J, Knapp D, Murawala P, Nowoshilow S, Masselink W, Taniguchi-Sugiura Y, Fei JF, Tanaka EM. A chromatin code for limb segment identity in axolotl limb regeneration. Dev Cell 2024; 59:2239-2253.e9. [PMID: 38788714 DOI: 10.1016/j.devcel.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/25/2023] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
The salamander limb correctly regenerates missing limb segments because connective tissue cells have segment-specific identities, termed "positional information". How positional information is molecularly encoded at the chromatin level has been unknown. Here, we performed genome-wide chromatin profiling in mature and regenerating axolotl limb connective tissue cells. We find segment-specific levels of histone H3K27me3 as the major positional mark, especially at limb homeoprotein gene loci but not their upstream regulators, constituting an intrinsic segment information code. During regeneration, regeneration-specific regulatory elements became active prior to the re-appearance of developmental regulatory elements. In the hand, the permissive chromatin state of the homeoprotein gene HoxA13 engages with the regeneration program bypassing the upper limb program. Comparison of regeneration regulatory elements with those found in other regenerative animals identified a core shared set of transcription factors, supporting an ancient, conserved regeneration program.
Collapse
Affiliation(s)
- Akane Kawaguchi
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Jingkui Wang
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Dunja Knapp
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Prayag Murawala
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria; DFG Research Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Sergej Nowoshilow
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria; DFG Research Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Wouter Masselink
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Yuka Taniguchi-Sugiura
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Ji-Feng Fei
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Elly M Tanaka
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria.
| |
Collapse
|
29
|
Sui X, Lo JA, Luo S, He Y, Tang Z, Lin Z, Zhou Y, Wang WX, Liu J, Wang X. Scalable spatial single-cell transcriptomics and translatomics in 3D thick tissue blocks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606553. [PMID: 39149316 PMCID: PMC11326170 DOI: 10.1101/2024.08.05.606553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Characterizing the transcriptional and translational gene expression patterns at the single-cell level within their three-dimensional (3D) tissue context is essential for revealing how genes shape tissue structure and function in health and disease. However, most existing spatial profiling techniques are limited to 5-20 μm thin tissue sections. Here, we developed Deep-STARmap and Deep-RIBOmap, which enable 3D in situ quantification of thousands of gene transcripts and their corresponding translation activities, respectively, within 200-μm thick tissue blocks. This is achieved through scalable probe synthesis, hydrogel embedding with efficient probe anchoring, and robust cDNA crosslinking. We first utilized Deep-STARmap in combination with multicolor fluorescent protein imaging for simultaneous molecular cell typing and 3D neuron morphology tracing in the mouse brain. We also demonstrate that 3D spatial profiling facilitates comprehensive and quantitative analysis of tumor-immune interactions in human skin cancer.
Collapse
Affiliation(s)
- Xin Sui
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally
| | - Jennifer A. Lo
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA USA
- These authors contributed equally
| | - Shuchen Luo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yichun He
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Zefang Tang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zuwan Lin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Yiming Zhou
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wendy Xueyi Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Xiao Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
30
|
Ke W, Fujioka M, Schedl P, Jaynes JB. Stem-loop and circle-loop TADs generated by directional pairing of boundary elements have distinct physical and regulatory properties. eLife 2024; 13:RP94114. [PMID: 39110491 PMCID: PMC11305674 DOI: 10.7554/elife.94114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
The chromosomes in multicellular eukaryotes are organized into a series of topologically independent loops called TADs. In flies, TADs are formed by physical interactions between neighboring boundaries. Fly boundaries exhibit distinct partner preferences, and pairing interactions between boundaries are typically orientation-dependent. Pairing can be head-to-tail or head-to-head. The former generates a stem-loop TAD, while the latter gives a circle-loop TAD. The TAD that encompasses the Drosophila even skipped (eve) gene is formed by the head-to-tail pairing of the nhomie and homie boundaries. To explore the relationship between loop topology and the physical and regulatory landscape, we flanked the nhomie boundary region with two attP sites. The attP sites were then used to generate four boundary replacements: λ DNA, nhomie forward (WT orientation), nhomie reverse (opposite of WT orientation), and homie forward (same orientation as WT homie). The nhomie forward replacement restores the WT physical and regulatory landscape: in MicroC experiments, the eve TAD is a 'volcano' triangle topped by a plume, and the eve gene and its regulatory elements are sequestered from interactions with neighbors. The λ DNA replacement lacks boundary function: the endpoint of the 'new' eve TAD on the nhomie side is ill-defined, and eve stripe enhancers activate a nearby gene, eIF3j. While nhomie reverse and homie forward restore the eve TAD, the topology is a circle-loop, and this changes the local physical and regulatory landscape. In MicroC experiments, the eve TAD interacts with its neighbors, and the plume at the top of the eve triangle peak is converted to a pair of 'clouds' of contacts with the next-door TADs. Consistent with the loss of isolation afforded by the stem-loop topology, the eve enhancers weakly activate genes in the neighboring TADs. Conversely, eve function is partially disrupted.
Collapse
Affiliation(s)
- Wenfan Ke
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Miki Fujioka
- Department of Biochemistry and Molecular Biology, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Paul Schedl
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - James B Jaynes
- Department of Biochemistry and Molecular Biology, Thomas Jefferson UniversityPhiladelphiaUnited States
| |
Collapse
|
31
|
Beaulieu MO, Thomas ED, Raible DW. Transdifferentiation is temporally uncoupled from progenitor pool expansion during hair cell regeneration in the zebrafish inner ear. Development 2024; 151:dev202944. [PMID: 39045613 PMCID: PMC11361639 DOI: 10.1242/dev.202944] [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: 04/09/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
Death of mechanosensory hair cells in the inner ear is a common cause of auditory and vestibular impairment in mammals, which have a limited ability to regrow these cells after damage. In contrast, non-mammalian vertebrates, including zebrafish, can robustly regenerate hair cells after severe organ damage. The zebrafish inner ear provides an understudied model system for understanding hair cell regeneration in organs that are highly conserved with their mammalian counterparts. Here, we quantitatively examine hair cell addition during growth and regeneration of the larval zebrafish inner ear. We used a genetically encoded ablation method to induce hair cell death and we observed gradual regeneration with correct spatial patterning over a 2-week period following ablation. Supporting cells, which surround and are a source of new hair cells, divide in response to hair cell ablation, expanding the possible progenitor pool. In parallel, nascent hair cells arise from direct transdifferentiation of progenitor pool cells temporally uncoupled from supporting cell division. These findings reveal a previously unrecognized mechanism of hair cell regeneration with implications for how hair cells may be encouraged to regenerate in the mammalian ear.
Collapse
Affiliation(s)
- Marielle O. Beaulieu
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
| | - Eric D. Thomas
- Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - David W. Raible
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
- Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
32
|
Cayrol B, Colella S, Uzest M. Coupling clearing and hybridization chain reaction approaches to investigate gene expression in organs inside intact insect heads. Microsc Res Tech 2024; 87:1926-1932. [PMID: 38558482 DOI: 10.1002/jemt.24561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
Detecting RNA molecules within their natural environment inside intact arthropods has long been challenging, particularly in small organisms covered by a tanned and pigmented cuticle. Here, we have developed a methodology that enables high-resolution analysis of the spatial distribution of transcripts of interest without having to dissect tiny organs or tissues, thereby preserving their integrity. We have combined an in situ amplification approach based on hybridization chain reaction, which enhances the signal-to-noise ratio, and a clearing approach that allows the visualization of inner organs beneath the cuticle. We have implemented this methodology for the first time in Hemiptera, mapping two salivary aphid (Acyrthosiphon pisum) transcripts, the effector c002 and the salivary sheath protein SHP. With a multiplex approach, we could simultaneously detect different mRNAs in mounted pea aphid head-thorax samples and show that they were distributed in distinct secretory cells of salivary glands. RESEARCH HIGHLIGHTS: Combining hybridisation chain reaction and clearing allows the detection of transcripts in intact aphids heads. The transcripts of the two salivary proteins c002 and SHP are compartmentalized in distinct secretory cells of the principal glands.
Collapse
Affiliation(s)
- Bastien Cayrol
- PHIM Plant Health Institute, Univ Montpellier, INRAE, CIRAD, Institut Agro, IRD, Montpellier, France
| | - Stefano Colella
- PHIM Plant Health Institute, Univ Montpellier, INRAE, CIRAD, Institut Agro, IRD, Montpellier, France
| | - Marilyne Uzest
- PHIM Plant Health Institute, Univ Montpellier, INRAE, CIRAD, Institut Agro, IRD, Montpellier, France
| |
Collapse
|
33
|
Ramadan YH, Hobert O. Visualization of gene expression in Pristionchus pacificus with smFISH and in situ HCR. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001274. [PMID: 39132051 PMCID: PMC11316217 DOI: 10.17912/micropub.biology.001274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/11/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Single molecule fluorescence in situ hybridization (smFISH) and in situ hybridization chain reaction (HCR) have become powerful tools to visualize gene expression in many different animal species. We show here that smFISH and in situ HCR can be put to effective use in the satellite nematode model organism Pristionchus pacificus . Examining the expression of a homeobox gene ( Ppa-unc-30) , we found that HCR is more sensitive than smFISH. We confirmed the robustness of HCR by visualization of the expression of several genes involved in neurotransmitter synthesis or transport ( Ppa-unc-25 /GAD, Ppa-unc-17/VAChT, Ppa-eat-4 /VGLUT). Combined with its relative cost-effectiveness compared to smFISH analysis, in situ HCR constitutes a useful addition to the toolbox for P. pacificus research .
Collapse
Affiliation(s)
| | - Oliver Hobert
- Biological Sciences, Columbia University, Howard Hughes Medical Institute, New York, United States
| |
Collapse
|
34
|
Brooks EC, Han SJY, Bonatto Paese CL, Lewis AA, Aarnio-Peterson M, Brugmann SA. The ciliary protein C2cd3 is required for mandibular musculoskeletal tissue patterning. Differentiation 2024; 138:100782. [PMID: 38810379 PMCID: PMC11227401 DOI: 10.1016/j.diff.2024.100782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
The mandible is composed of several musculoskeletal tissues including bone, cartilage, and tendon that require precise patterning to ensure structural and functional integrity. Interestingly, most of these tissues are derived from one multipotent cell population called cranial neural crest cells (CNCCs). How CNCCs are properly instructed to differentiate into various tissue types remains nebulous. To better understand the mechanisms necessary for the patterning of mandibular musculoskeletal tissues we utilized the avian mutant talpid2 (ta2) which presents with several malformations of the facial skeleton including dysplastic tendons, mispatterned musculature, and bilateral ectopic cartilaginous processes extending off Meckel's cartilage. We found an ectopic epithelial BMP signaling domain in the ta2 mandibular prominence (MNP) that correlated with the subsequent expansion of SOX9+ cartilage precursors. These findings were validated with conditional murine models suggesting an evolutionarily conserved mechanism for CNCC-derived musculoskeletal patterning. Collectively, these data support a model in which cilia are required to define epithelial signal centers essential for proper musculoskeletal patterning of CNCC-derived mesenchyme.
Collapse
Affiliation(s)
- Evan C Brooks
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Simon J Y Han
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Christian Louis Bonatto Paese
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Amya A Lewis
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Megan Aarnio-Peterson
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
35
|
Clarke DN, Formery L, Lowe CJ. See-Star: a versatile hydrogel-based protocol for clearing large, opaque and calcified marine invertebrates. EvoDevo 2024; 15:8. [PMID: 38918798 PMCID: PMC11201320 DOI: 10.1186/s13227-024-00228-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Studies of morphology and developmental patterning in adult stages of many invertebrates are hindered by opaque structures, such as shells, skeletal elements, and pigment granules that block or refract light and necessitate sectioning for observation of internal features. An inherent challenge in studies relying on surgical approaches is that cutting tissue is semi-destructive, and delicate structures, such as axonal processes within neural networks, are computationally challenging to reconstruct once disrupted. To address this problem, we developed See-Star, a hydrogel-based tissue clearing protocol to render the bodies of opaque and calcified invertebrates optically transparent while preserving their anatomy in an unperturbed state, facilitating molecular labeling and observation of intact organ systems. The resulting protocol can clear large (> 1 cm3) specimens to enable deep-tissue imaging, and is compatible with molecular techniques, such as immunohistochemistry and in situ hybridization to visualize protein and mRNA localization. To test the utility of this method, we performed a whole-mount imaging study of intact nervous systems in juvenile echinoderms and molluscs and demonstrate that See-Star allows for comparative studies to be extended far into development, facilitating insights into the anatomy of juveniles and adults that are usually not amenable to whole-mount imaging.
Collapse
Affiliation(s)
- D N Clarke
- Department of Biology, Hopkins Marine Station, Stanford University, Pacific Grove, CA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - L Formery
- Department of Biology, Hopkins Marine Station, Stanford University, Pacific Grove, CA, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - C J Lowe
- Department of Biology, Hopkins Marine Station, Stanford University, Pacific Grove, CA, USA
- Chan Zuckerberg BioHub, San Francisco, CA, USA
| |
Collapse
|
36
|
André M, Dinvaut S, Castellani V, Falk J. 3D exploration of gene expression in chicken embryos through combined RNA fluorescence in situ hybridization, immunofluorescence, and clearing. BMC Biol 2024; 22:131. [PMID: 38831263 PMCID: PMC11149291 DOI: 10.1186/s12915-024-01922-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Fine characterization of gene expression patterns is crucial to understand many aspects of embryonic development. The chicken embryo is a well-established and valuable animal model for developmental biology. The period spanning from the third to sixth embryonic days (E3 to E6) is critical for many organ developments. Hybridization chain reaction RNA fluorescent in situ hybridization (HCR RNA-FISH) enables multiplex RNA detection in thick samples including embryos of various animal models. However, its use is limited by tissue opacity. RESULTS We optimized HCR RNA-FISH protocol to efficiently label RNAs in whole mount chicken embryos from E3.5 to E5.5 and adapted it to ethyl cinnamate (ECi) tissue clearing. We show that light sheet imaging of HCR RNA-FISH after ECi clearing allows RNA expression analysis within embryonic tissues with good sensitivity and spatial resolution. Finally, whole mount immunofluorescence can be performed after HCR RNA-FISH enabling as exemplified to assay complex spatial relationships between axons and their environment or to monitor GFP electroporated neurons. CONCLUSIONS We could extend the use of HCR RNA-FISH to older chick embryos by optimizing HCR RNA-FISH and combining it with tissue clearing and 3D imaging. The integration of immunostaining makes possible to combine gene expression with classical cell markers, to correlate expressions with morphological differentiation and to depict gene expressions in gain or loss of function contexts. Altogether, this combined procedure further extends the potential of HCR RNA-FISH technique for chicken embryology.
Collapse
Affiliation(s)
- Maëlys André
- MeLiS, CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, 8 avenue Rockefeller, 69008, Lyon, France.
| | - Sarah Dinvaut
- MeLiS, CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, 8 avenue Rockefeller, 69008, Lyon, France
| | - Valérie Castellani
- MeLiS, CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, 8 avenue Rockefeller, 69008, Lyon, France
| | - Julien Falk
- MeLiS, CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, 8 avenue Rockefeller, 69008, Lyon, France.
| |
Collapse
|
37
|
Steyn C, Mishi R, Fillmore S, Verhoog MB, More J, Rohlwink UK, Melvill R, Butler J, Enslin JMN, Jacobs M, Sauka-Spengler T, Greco M, Quiñones S, Dulla CG, Raimondo JV, Figaji A, Hockman D. Cell type-specific gene expression dynamics during human brain maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.29.560114. [PMID: 37808657 PMCID: PMC10557738 DOI: 10.1101/2023.09.29.560114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The human brain undergoes protracted post-natal maturation, guided by dynamic changes in gene expression. Most studies exploring these processes have used bulk tissue analyses, which mask cell type-specific gene expression dynamics. Here, using single nucleus (sn)RNA-seq on temporal lobe tissue, including samples of African ancestry, we build a joint paediatric and adult atlas of 75 cell subtypes, which we verify with spatial transcriptomics. We explore the differences between paediatric and adult cell types, revealing the genes and pathways that change during brain maturation. Our results highlight excitatory neuron subtypes, including the LTK and FREM subtypes, that show elevated expression of genes associated with cognition and synaptic plasticity in paediatric tissue. The new resources we present here improve our understanding of the brain during its development and contribute to global efforts to build an inclusive brain cell map.
Collapse
Affiliation(s)
- Christina Steyn
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ruvimbo Mishi
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Stephanie Fillmore
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Matthijs B Verhoog
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Jessica More
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ursula K Rohlwink
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Roger Melvill
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - James Butler
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Johannes M N Enslin
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Muazzam Jacobs
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Department of Pathology University of Cape Town
- National Health Laboratory Service, South Africa
| | - Tatjana Sauka-Spengler
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Maria Greco
- Single Cell Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Sadi Quiñones
- Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Graduate School of Biomedical Science, Tufts University School of Medicine, Boston, MA, USA
| | - Chris G Dulla
- Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Joseph V Raimondo
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Anthony Figaji
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Dorit Hockman
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
38
|
Powell GT, Faro A, Zhao Y, Stickney H, Novellasdemunt L, Henriques P, Gestri G, White ER, Ren J, Lu W, Young RM, Hawkins TA, Cavodeassi F, Schwarz Q, Dreosti E, Raible DW, Li VSW, Wright GJ, Jones EY, Wilson SW. Cachd1 interacts with Wnt receptors and regulates neuronal asymmetry in the zebrafish brain. Science 2024; 384:573-579. [PMID: 38696577 PMCID: PMC7615972 DOI: 10.1126/science.ade6970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/27/2024] [Indexed: 05/04/2024]
Abstract
Neurons on the left and right sides of the nervous system often show asymmetric properties, but how such differences arise is poorly understood. Genetic screening in zebrafish revealed that loss of function of the transmembrane protein Cachd1 resulted in right-sided habenula neurons adopting left-sided identity. Cachd1 is expressed in neuronal progenitors, functions downstream of asymmetric environmental signals, and influences timing of the normally asymmetric patterns of neurogenesis. Biochemical and structural analyses demonstrated that Cachd1 can bind simultaneously to Lrp6 and Frizzled family Wnt co-receptors. Consistent with this, lrp6 mutant zebrafish lose asymmetry in the habenulae, and epistasis experiments support a role for Cachd1 in modulating Wnt pathway activity in the brain. These studies identify Cachd1 as a conserved Wnt receptor-interacting protein that regulates lateralized neuronal identity in the zebrafish brain.
Collapse
Affiliation(s)
- Gareth T. Powell
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
| | - Ana Faro
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Yuguang Zhao
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Heather Stickney
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
- Ambry Genetics; Aliso Viejo, CA 92656, USA
| | - Laura Novellasdemunt
- The Francis Crick Institute; London, NW1 1AT, UK
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology; 08028, Barcelona, Spain
| | - Pedro Henriques
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Gaia Gestri
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | | | - Jingshan Ren
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Weixian Lu
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Rodrigo M. Young
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor; Camino La Piramide 5750, 8580745, Santiago, Chile
| | - Thomas A. Hawkins
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Florencia Cavodeassi
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- St. George’s, University of London; London, SW17 0RE, UK
| | - Quenten Schwarz
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
| | - Elena Dreosti
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - David W. Raible
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
| | | | - Gavin J. Wright
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York; York, YO10 5DD, UK
| | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Stephen W. Wilson
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| |
Collapse
|
39
|
Tjeerdema E, Lee Y, Metry R, Hamdoun A. Semi-automated, high-content imaging of drug transporter knockout sea urchin (Lytechinus pictus) embryos. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2024; 342:313-329. [PMID: 38087422 PMCID: PMC12010930 DOI: 10.1002/jez.b.23231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/08/2023] [Accepted: 11/19/2023] [Indexed: 05/01/2024]
Abstract
A defining feature of sea urchins is their extreme fecundity. Urchins produce millions of transparent, synchronously developing embryos, ideal for spatial and temporal analysis of development. This biological feature has been effectively utilized for ensemble measurement of biochemical changes. However, it has been underutilized in imaging studies, where single embryo measurements are used. Here we present an example of how stable genetics and high content imaging, along with machine learning-based image analysis, can be used to exploit the fecundity and synchrony of sea urchins in imaging-based drug screens. Building upon our recently created sea urchin ABCB1 knockout line, we developed a high-throughput assay to probe the role of this drug transporter in embryos. We used high content imaging to compare accumulation and toxicity of canonical substrates and inhibitors of the transporter, including fluorescent molecules and antimitotic cancer drugs, in homozygous knockout and wildtype embryos. To measure responses from the resulting image data, we used a nested convolutional neural network, which rapidly classified embryos according to fluorescence or cell division. This approach identified sea urchin embryos with 99.8% accuracy and determined two-cell and aberrant embryos with 96.3% and 89.1% accuracy, respectively. The results revealed that ABCB1 knockout embryos accumulated the transporter substrate calcein 3.09 times faster than wildtypes. Similarly, knockouts were 4.71 and 3.07 times more sensitive to the mitotic poisons vinblastine and taxol. This study paves the way for large scale pharmacological screens in the sea urchin embryo.
Collapse
Affiliation(s)
- Evan Tjeerdema
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Yoon Lee
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Rachel Metry
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Amro Hamdoun
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
40
|
Beaulieu MO, Thomas ED, Raible DW. Transdifferentiation is uncoupled from progenitor pool expansion during hair cell regeneration in the zebrafish inner ear. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588777. [PMID: 38645220 PMCID: PMC11030336 DOI: 10.1101/2024.04.09.588777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Death of mechanosensory hair cells in the inner ear is a common cause of auditory and vestibular impairment in mammals, which have a limited ability to regrow these cells after damage. In contrast, non-mammalian vertebrates including zebrafish can robustly regenerate hair cells following severe organ damage. The zebrafish inner ear provides an understudied model system for understanding hair cell regeneration in organs that are highly conserved with their mammalian counterparts. Here we quantitatively examine hair cell addition during growth and regeneration of the larval zebrafish inner ear. We used a genetically encoded ablation method to induce hair cell death and observed gradual regeneration with correct spatial patterning over two weeks following ablation. Supporting cells, which surround and are a source of new hair cells, divide in response to hair cell ablation, expanding the possible progenitor pool. In parallel, nascent hair cells arise from direct transdifferentiation of progenitor pool cells uncoupled from progenitor division. These findings reveal a previously unrecognized mechanism of hair cell regeneration with implications for how hair cells may be encouraged to regenerate in the mammalian ear.
Collapse
Affiliation(s)
- Marielle O. Beaulieu
- Molecular and Cellular Biology Graduate Program, Seattle, WA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, Seattle, WA
| | - Eric D. Thomas
- Neuroscience Graduate Program, Seattle, WA
- Department of Biological Structure University of Washington, Seattle, WA
| | - David W. Raible
- Molecular and Cellular Biology Graduate Program, Seattle, WA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, Seattle, WA
- Neuroscience Graduate Program, Seattle, WA
- Department of Biological Structure University of Washington, Seattle, WA
| |
Collapse
|
41
|
Truong BT, Shull LC, Zepeda BJ, Lencer E, Artinger KB. Human split hand/foot variants are not as functional as wildtype human PRDM1 in the rescue of craniofacial defects. Birth Defects Res 2024; 116:e2327. [PMID: 38456586 PMCID: PMC10949536 DOI: 10.1002/bdr2.2327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Split hand/foot malformation (SHFM) is a congenital limb disorder presenting with limb anomalies, such as missing, hypoplastic, or fused digits, and often craniofacial defects, including a cleft lip/palate, microdontia, micrognathia, or maxillary hypoplasia. We previously identified three novel variants in the transcription factor, PRDM1, that are associated with SHFM phenotypes. One individual also presented with a high arch palate. Studies in vertebrates indicate that PRDM1 is important for development of the skull; however, prior to our study, human variants in PRDM1 had not been associated with craniofacial anomalies. METHODS Using transient mRNA overexpression assays in prdm1a-/- mutant zebrafish, we tested whether the PRDM1 SHFM variants were functional and could lead to a rescue of the craniofacial defects observed in prdm1a-/- mutants. We also mined previously published CUT&RUN and RNA-seq datasets that sorted EGFP-positive cells from a Tg(Mmu:Prx1-EGFP) transgenic line that labels the pectoral fin, pharyngeal arches, and dorsal part of the head to examine Prdm1a binding and the effect of Prdm1a loss on craniofacial genes. RESULTS The prdm1a-/- mutants exhibit craniofacial defects including a hypoplastic neurocranium, a loss of posterior ceratobranchial arches, a shorter palatoquadrate, and an inverted ceratohyal. Injection of wildtype (WT) hPRDM1 in prdm1a-/- mutants partially rescues the palatoquadrate phenotype. However, injection of each of the three SHFM variants fails to rescue this skeletal defect. Loss of prdm1a leads to a decreased expression of important craniofacial genes by RNA-seq, including emilin3a, confirmed by hybridization chain reaction expression. Other genes including dlx5a/dlx6a, hand2, sox9b, col2a1a, and hoxb genes are also reduced. Validation by real-time quantitative PCR in the anterior half of zebrafish embryos failed to confirm the expression changes suggesting that the differences are enriched in prx1 expressing cells. CONCLUSION These data suggest that the three SHFM variants are likely not functional and may be associated with the craniofacial defects observed in the humans. Finally, they demonstrate how Prdm1a can directly bind and regulate genes involved in craniofacial development.
Collapse
Affiliation(s)
- Brittany T Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lomeli C Shull
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bryan J Zepeda
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| | - Ezra Lencer
- Biology Department, Lafayette College, Easton, Pennsylvania, USA
| | - Kristin B Artinger
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| |
Collapse
|
42
|
Schulte S, Shin B, Rothenberg EV, Pierce NA. Multiplex, Quantitative, High-Resolution Imaging of Protein:Protein Complexes via Hybridization Chain Reaction. ACS Chem Biol 2024; 19:280-288. [PMID: 38232374 PMCID: PMC10877569 DOI: 10.1021/acschembio.3c00431] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 11/18/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
Signal amplification based on the mechanism of hybridization chain reaction (HCR) facilitates spatial exploration of gene regulatory networks by enabling multiplex, quantitative, high-resolution imaging of RNA and protein targets. Here, we extend these capabilities to the imaging of protein:protein complexes, using proximity-dependent cooperative probes to conditionally generate a single amplified signal if and only if two target proteins are colocalized within the sample. HCR probes and amplifiers combine to provide automatic background suppression throughout the protocol, ensuring that even if reagents bind nonspecifically in the sample, they will not generate amplified background. We demonstrate protein:protein imaging with a high signal-to-background ratio in human cells, mouse proT cells, and highly autofluorescent formalin-fixed paraffin-embedded (FFPE) human breast tissue sections. Further, we demonstrate multiplex imaging of three different protein:protein complexes simultaneously and validate that HCR enables accurate and precise relative quantitation of protein:protein complexes with subcellular resolution in an anatomical context. Moreover, we establish a unified framework for simultaneous multiplex, quantitative, high-resolution imaging of RNA, protein, and protein:protein targets, with one-step, isothermal, enzyme-free HCR signal amplification performed for all target classes simultaneously.
Collapse
Affiliation(s)
- Samuel
J. Schulte
- Division
of Biology and Biological Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Boyoung Shin
- Division
of Biology and Biological Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Ellen V. Rothenberg
- Division
of Biology and Biological Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Niles A. Pierce
- Division
of Biology and Biological Engineering, California
Institute of Technology, Pasadena, California 91125, United States
- Division
of Engineering and Applied Science, California
Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
43
|
Schulte SJ, Fornace ME, Hall JK, Shin GJ, Pierce NA. HCR spectral imaging: 10-plex, quantitative, high-resolution RNA and protein imaging in highly autofluorescent samples. Development 2024; 151:dev202307. [PMID: 38415752 PMCID: PMC10941662 DOI: 10.1242/dev.202307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/21/2023] [Indexed: 02/29/2024]
Abstract
Signal amplification based on the mechanism of hybridization chain reaction (HCR) provides a unified framework for multiplex, quantitative, high-resolution imaging of RNA and protein targets in highly autofluorescent samples. With conventional bandpass imaging, multiplexing is typically limited to four or five targets owing to the difficulty in separating signals generated by fluorophores with overlapping spectra. Spectral imaging has offered the conceptual promise of higher levels of multiplexing, but it has been challenging to realize this potential in highly autofluorescent samples, including whole-mount vertebrate embryos. Here, we demonstrate robust HCR spectral imaging with linear unmixing, enabling simultaneous imaging of ten RNA and/or protein targets in whole-mount zebrafish embryos and mouse brain sections. Further, we demonstrate that the amplified and unmixed signal in each of the ten channels is quantitative, enabling accurate and precise relative quantitation of RNA and/or protein targets with subcellular resolution, and RNA absolute quantitation with single-molecule resolution, in the anatomical context of highly autofluorescent samples.
Collapse
Affiliation(s)
- Samuel J. Schulte
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mark E. Fornace
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - John K. Hall
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Grace J. Shin
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Niles A. Pierce
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Division of Engineering & Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
44
|
Schwarz M, Hamburger K. Memory effects of visual and olfactory landmark information in human wayfinding. Cogn Process 2024; 25:37-51. [PMID: 38032500 PMCID: PMC10827900 DOI: 10.1007/s10339-023-01169-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023]
Abstract
Non-human animals are exceptionally good at using smell to find their way through the environment. However, the use of olfactory cues for human navigation is often underestimated. Although the sense of smell is well-known for its distinct connection to memory and emotion, memory effects in human navigation using olfactory landmarks have not been studied yet. Therefore, this article compares wayfinding and recognition performance for visual and olfactory landmarks learned by 52 participants in a virtual maze. Furthermore, it is one of the first empirical studies investigating differences in memory effects on human navigation by using two separate test situations 1 month apart. The experimental task was to find the way through a maze-like virtual environment with either olfactory or visual cues at the intersections that served as decision points. Our descriptive results show that performance was above chance level for both conditions (visual and olfactory landmarks). Wayfinding performance did not decrease 1 month later when using olfactory landmarks. In contrast, when using visual landmarks wayfinding performance decreased significantly, while visual landmarks overall lead to better recognition than olfactory landmarks at both times of testing. The results demonstrate the unique character of human odor memory and support the conclusion that olfactory cues may be used in human spatial orientation. Furthermore, the present study expands the research field of human wayfinding by providing a study that investigates memory for landmark knowledge and route decisions for the visual and olfactory modality. However, more studies are required to put this important research strand forward.
Collapse
Affiliation(s)
- Mira Schwarz
- Experimental Psychology and Cognitive Science, Department of Psychology and Sport Science, Justus Liebig University, Otto-Behagel-Str. 10F, 35394, Giessen, Germany.
| | - Kai Hamburger
- Experimental Psychology and Cognitive Science, Department of Psychology and Sport Science, Justus Liebig University, Otto-Behagel-Str. 10F, 35394, Giessen, Germany
| |
Collapse
|
45
|
Gautam S, Fenner JL, Wang B, Range RC. Evolutionarily conserved Wnt/Sp5 signaling is critical for anterior-posterior axis patterning in sea urchin embryos. iScience 2024; 27:108616. [PMID: 38179064 PMCID: PMC10765061 DOI: 10.1016/j.isci.2023.108616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/30/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Studies across a diverse group of metazoan embryos indicate that Wnt signaling often activates the transcription factor Sp5, forming a signaling 'cassette' that plays critical roles in many developmental processes. This study explores the role of Wnt/Sp5 signaling during the specification and patterning of the primary germ layers during early anterior-posterior axis formation in the deuterostome sea urchin embryo. Our functional analyses show that Sp5 is critical for endomesoderm specification downstream of Wnt/β-catenin in posterior cells as well as anterior neuroectoderm patterning downstream of non-canonical Wnt/JNK signaling in anterior cells. Interestingly, expression and functional data comparisons show that Wnt/Sp5 signaling often plays similar roles in posterior endomesoderm as well as neuroectoderm patterning along the AP axis of several deuterostome embryos, including vertebrates. Thus, our findings provide strong support for the idea that Wnt-Sp5 signaling cassettes were critical for the establishment of early germ layers in the common deuterostome ancestor.
Collapse
Affiliation(s)
- Sujan Gautam
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Jennifer L. Fenner
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Boyuan Wang
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Ryan C. Range
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
46
|
Rayamajhi D, Ege M, Ukhanov K, Ringers C, Zhang Y, Jung I, D’Gama PP, Li SS, Cosacak MI, Kizil C, Park HC, Yaksi E, Martens JR, Brody SL, Jurisch-Yaksi N, Roy S. The forkhead transcription factor Foxj1 controls vertebrate olfactory cilia biogenesis and sensory neuron differentiation. PLoS Biol 2024; 22:e3002468. [PMID: 38271330 PMCID: PMC10810531 DOI: 10.1371/journal.pbio.3002468] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024] Open
Abstract
In vertebrates, olfactory receptors localize on multiple cilia elaborated on dendritic knobs of olfactory sensory neurons (OSNs). Although olfactory cilia dysfunction can cause anosmia, how their differentiation is programmed at the transcriptional level has remained largely unexplored. We discovered in zebrafish and mice that Foxj1, a forkhead domain-containing transcription factor traditionally linked with motile cilia biogenesis, is expressed in OSNs and required for olfactory epithelium (OE) formation. In keeping with the immotile nature of olfactory cilia, we observed that ciliary motility genes are repressed in zebrafish, mouse, and human OSNs. Strikingly, we also found that besides ciliogenesis, Foxj1 controls the differentiation of the OSNs themselves by regulating their cell type-specific gene expression, such as that of olfactory marker protein (omp) involved in odor-evoked signal transduction. In line with this, response to bile acids, odors detected by OMP-positive OSNs, was significantly diminished in foxj1 mutant zebrafish. Taken together, our findings establish how the canonical Foxj1-mediated motile ciliogenic transcriptional program has been repurposed for the biogenesis of immotile olfactory cilia, as well as for the development of the OSNs.
Collapse
Affiliation(s)
- Dheeraj Rayamajhi
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Mert Ege
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kirill Ukhanov
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Christa Ringers
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yiliu Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Inyoung Jung
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biomedical Sciences, Korea University, Ansan, South Korea
| | - Percival P. D’Gama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Summer Shijia Li
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Dresden, Germany
| | - Caghan Kizil
- Department of Neurology and The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University, Ansan, South Korea
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
- Koç University Research Center for Translational Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Jeffrey R. Martens
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Steven L. Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
- Department of Paediatrics, National University of Singapore, Singapore
| |
Collapse
|
47
|
Nguyen TT, Mitchell JM, Kiel MD, Kenny CP, Li H, Jones KL, Cornell RA, Williams TJ, Nichols JT, Van Otterloo E. TFAP2 paralogs regulate midfacial development in part through a conserved ALX genetic pathway. Development 2024; 151:dev202095. [PMID: 38063857 PMCID: PMC10820886 DOI: 10.1242/dev.202095] [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: 06/16/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Cranial neural crest development is governed by positional gene regulatory networks (GRNs). Fine-tuning of the GRN components underlies facial shape variation, yet how those networks in the midface are connected and activated remain poorly understood. Here, we show that concerted inactivation of Tfap2a and Tfap2b in the murine neural crest, even during the late migratory phase, results in a midfacial cleft and skeletal abnormalities. Bulk and single-cell RNA-seq profiling reveal that loss of both TFAP2 family members dysregulates numerous midface GRN components involved in midface morphogenesis, patterning and differentiation. Notably, Alx1, Alx3 and Alx4 (ALX) transcript levels are reduced, whereas ChIP-seq analyses suggest TFAP2 family members directly and positively regulate ALX gene expression. Tfap2a, Tfap2b and ALX co-expression in midfacial neural crest cells of both mouse and zebrafish implies conservation of this regulatory axis across vertebrates. Consistent with this notion, tfap2a zebrafish mutants present with abnormal alx3 expression patterns, Tfap2a binds ALX loci and tfap2a-alx3 genetic interactions are observed. Together, these data demonstrate TFAP2 paralogs regulate vertebrate midfacial development in part by activating expression of ALX transcription factor genes.
Collapse
Affiliation(s)
- Timothy T. Nguyen
- Iowa Institute for Oral Health Research, College of Dentistry and Dental Clinics, University of Iowa, Iowa City, IA 52242, USA
- Department of Periodontics, College of Dentistry and Dental Clinics, University of Iowa, Iowa City, IA 52242, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Jennyfer M. Mitchell
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michaela D. Kiel
- Iowa Institute for Oral Health Research, College of Dentistry and Dental Clinics, University of Iowa, Iowa City, IA 52242, USA
- Department of Periodontics, College of Dentistry and Dental Clinics, University of Iowa, Iowa City, IA 52242, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Colin P. Kenny
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Hong Li
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L. Jones
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Robert A. Cornell
- Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA 98195, USA
| | - Trevor J. Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James T. Nichols
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eric Van Otterloo
- Iowa Institute for Oral Health Research, College of Dentistry and Dental Clinics, University of Iowa, Iowa City, IA 52242, USA
- Department of Periodontics, College of Dentistry and Dental Clinics, University of Iowa, Iowa City, IA 52242, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Craniofacial Anomalies Research Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
48
|
Maltby CJ, Krans A, Grudzien SJ, Palacios Y, Muiños J, Suárez A, Asher M, Khurana V, Barmada SJ, Dijkstra AA, Todd PK. AAGGG repeat expansions trigger RFC1-independent synaptic dysregulation in human CANVAS Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571345. [PMID: 38168171 PMCID: PMC10760133 DOI: 10.1101/2023.12.13.571345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Cerebellar ataxia with neuropathy and vestibular areflexia syndrome (CANVAS) is a late onset, recessively inherited neurodegenerative disorder caused by biallelic, non-reference pentameric AAGGG(CCCTT) repeat expansions within the second intron of replication factor complex subunit 1 (RFC1). To investigate how these repeats cause disease, we generated CANVAS patient induced pluripotent stem cell (iPSC) derived neurons (iNeurons) and utilized calcium imaging and transcriptomic analysis to define repeat-elicited gain-of-function and loss-of-function contributions to neuronal toxicity. AAGGG repeat expansions do not alter neuronal RFC1 splicing, expression, or DNA repair pathway functions. In reporter assays, AAGGG repeats are translated into pentapeptide repeat proteins that selectively accumulate in CANVAS patient brains. However, neither these proteins nor repeat RNA foci were detected in iNeurons, and overexpression of these repeats in isolation did not induce neuronal toxicity. CANVAS iNeurons exhibit defects in neuronal development and diminished synaptic connectivity that is rescued by CRISPR deletion of a single expanded allele. These phenotypic deficits were not replicated by knockdown of RFC1 in control neurons and were not rescued by ectopic expression of RFC1. These findings support a repeat-dependent but RFC1-independent cause of neuronal dysfunction in CANVAS, with important implications for therapeutic development in this currently untreatable condition.
Collapse
Affiliation(s)
- Connor J. Maltby
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Krans
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Ann Arbor Veterans Administration Healthcare, Ann Arbor, MI, USA
| | - Samantha J. Grudzien
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yomira Palacios
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Postbaccalaureate Research Education Program, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Muiños
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- UM SMART Undergraduate Summer Program, University of Michigan, Ann Arbor, MI, USA
| | - Andrea Suárez
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Postbaccalaureate Research Education Program, University of Michigan, Ann Arbor, MI, USA
| | - Melissa Asher
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Vikram Khurana
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sami J. Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Anke A. Dijkstra
- Department of Pathology, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter K. Todd
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Ann Arbor Veterans Administration Healthcare, Ann Arbor, MI, USA
| |
Collapse
|
49
|
Sperling AL, Eves-van den Akker S. Whole mount multiplexed visualization of DNA, mRNA, and protein in plant-parasitic nematodes. PLANT METHODS 2023; 19:139. [PMID: 38049899 PMCID: PMC10696717 DOI: 10.1186/s13007-023-01112-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/16/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND Plant-parasitic nematodes compromise the agriculture of a wide variety of the most common crops worldwide. Obtaining information on the fundamental biology of these organisms and how they infect the plant has been restricted by the ability to visualize intact nematodes using small molecule stains, antibodies, or in situ hybridization. Consequently, there is limited information available about the internal composition of the nematodes or the biology of the effector molecules they use to reprogram their host plant. RESULTS We present the Sperling prep - a whole mount method for nematode preparation that enables staining with small molecules, antibodies, or in situ hybridization chain reaction. This method does not require specialized apparatus and utilizes typical laboratory equipment and materials. By dissociating the strong cuticle and interior muscle layers, we enabled entry of the small molecule stains into the tissue. After permeabilization, small molecule stains can be used to visualize the nuclei with the DNA stain DAPI and the internal structures of the digestive tract and longitudinal musculature with the filamentous actin stain phalloidin. The permeabilization even allows entry of larger antibodies, albeit with lower efficiency. Finally, this method works exceptionally well with in situ HCR. Using this method, we have visualized effector transcripts specific to the dorsal gland and the subventral grand of the sugar beet cyst nematode, Heterodera schachtii, multiplexed in the same nematode. CONCLUSION We were able to visualize the internal structures of the nematode as well as key effector transcripts that are used during plant infection and parasitism. Therefore, this method provides an important toolkit for studying the biology of plant-parasitic nematodes.
Collapse
Affiliation(s)
- Alexis L Sperling
- Department of Plant Sciences, Crop Science Centre, University of Cambridge, 93 Lawrence Weaver Road, Cambridge, CB3 0LE, Cambridge, UK
| | - Sebastian Eves-van den Akker
- Department of Plant Sciences, Crop Science Centre, University of Cambridge, 93 Lawrence Weaver Road, Cambridge, CB3 0LE, Cambridge, UK.
| |
Collapse
|
50
|
Chen Q, Leshkowitz D, Li H, van Impel A, Schulte-Merker S, Amit I, Rizzoti K, Levkowitz G. Neural plate progenitors give rise to both anterior and posterior pituitary cells. Dev Cell 2023; 58:2652-2665.e6. [PMID: 37683631 DOI: 10.1016/j.devcel.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023]
Abstract
The pituitary is the master neuroendocrine gland, which regulates body homeostasis. It consists of the anterior pituitary/adenohypophysis harboring hormones producing cells and the posterior pituitary/neurohypophysis, which relays the passage of hormones from the brain to the periphery. It is accepted that the adenohypophysis originates from the oral ectoderm (Rathke's pouch), whereas the neural ectoderm contributes to the neurohypophysis. Single-cell transcriptomics of the zebrafish pituitary showed that cyp26b1-positive astroglial pituicytes of the neurohypophysis and prop1-positive adenohypophyseal progenitors expressed common markers implying lineage relatedness. Genetic tracing identifies that, in contrast to the prevailing dogma, neural plate precursors of zebrafish (her4.3+) and mouse (Sox1+) contribute to both neurohypophyseal and a subset of adenohypophyseal cells. Pituicyte-derived retinoic-acid-degrading enzyme Cyp26b1 fine-tunes differentiation of prop1+ progenitors into hormone-producing cells. These results challenge the notion that adenohypophyseal cells are exclusively derived from non-neural ectoderm and demonstrate that crosstalk between neuro- and adeno-hypophyseal cells affects differentiation of pituitary cells.
Collapse
Affiliation(s)
- Qiyu Chen
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dena Leshkowitz
- Life Science Core Facilities, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Hanjie Li
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Present address: CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Andreas van Impel
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, UK
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel.
| |
Collapse
|