1
|
Lin SJ, Huang K, Petree C, Qin W, Varshney P, Varshney GK. Optimizing gRNA selection for high-penetrance F0 CRISPR screening for interrogating disease gene function. Nucleic Acids Res 2025; 53:gkaf180. [PMID: 40103232 PMCID: PMC11915512 DOI: 10.1093/nar/gkaf180] [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: 06/07/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Genes and genetic variants associated with human disease are continually being discovered, but validating their causative roles and mechanisms remains a significant challenge. CRISPR/Cas9 genome editing in model organisms like zebrafish can enable phenotypic characterization of founder generation (F0) knockouts (Crispants), but existing approaches are not amenable to high-throughput genetic screening due to high variability, cost, and low phenotype penetrance. To overcome these challenges, here we provide guide RNA (gRNA) selection rules that enable high phenotypic penetrance of up to three simultaneous knockouts in F0 animals following injection of 1-2 gRNAs per gene. We demonstrate a strong transcriptomic overlap in our F0 knockouts and stable knockout lines that take several months to generate. We systematically evaluated this approach across 324 gRNAs targeting 125 genes and demonstrated its utility in studying epistasis, characterizing paralogous genes, and validating human disease gene phenotypes across multiple tissues. Applying our approach in a high-throughput manner, we screened and identified 10 novel neurodevelopmental disorders and 50 hearing genes not previously studied in zebrafish. Altogether, our approach achieves high phenotypic penetrance using low numbers of gRNAs per gene in F0 zebrafish, offering a robust pipeline for rapidly characterizing candidate human disease genes.
Collapse
Affiliation(s)
- Sheng-Jia Lin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Kevin Huang
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Cassidy Petree
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Wei Qin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Pratishtha Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| |
Collapse
|
2
|
Healey HM, Penn HB, Small CM, Bassham S, Goyal V, Woods MA, Cresko WA. Single-cell sequencing provides clues about the developmental genetic basis of evolutionary adaptations in syngnathid fishes. eLife 2025; 13:RP97764. [PMID: 39898521 PMCID: PMC11790252 DOI: 10.7554/elife.97764] [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: 02/04/2025] Open
Abstract
Seahorses, pipefishes, and seadragons are fishes from the family Syngnathidae that have evolved extraordinary traits including male pregnancy, elongated snouts, loss of teeth, and dermal bony armor. The developmental genetic and cellular changes that led to the evolution of these traits are largely unknown. Recent syngnathid genome assemblies revealed suggestive gene content differences and provided the opportunity for detailed genetic analyses. We created a single-cell RNA sequencing atlas of Gulf pipefish embryos to understand the developmental basis of four traits: derived head shape, toothlessness, dermal armor, and male pregnancy. We completed marker gene analyses, built genetic networks, and examined the spatial expression of select genes. We identified osteochondrogenic mesenchymal cells in the elongating face that express regulatory genes bmp4, sfrp1a, and prdm16. We found no evidence for tooth primordia cells, and we observed re-deployment of osteoblast genetic networks in developing dermal armor. Finally, we found that epidermal cells expressed nutrient processing and environmental sensing genes, potentially relevant for the brooding environment. The examined pipefish evolutionary innovations are composed of recognizable cell types, suggesting that derived features originate from changes within existing gene networks. Future work addressing syngnathid gene networks across multiple stages and species is essential for understanding how the novelties of these fish evolved.
Collapse
Affiliation(s)
- Hope M Healey
- Institute of Ecology and Evolution, University of Oregon, Eugene, United States
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, United States
| | - Hayden B Penn
- Institute of Ecology and Evolution, University of Oregon, Eugene, United States
| | - Clayton M Small
- Institute of Ecology and Evolution, University of Oregon, Eugene, United States
- School of Computer and Data Science, University of Oregon, Eugene, United States
| | - Susan Bassham
- Institute of Ecology and Evolution, University of Oregon, Eugene, United States
| | - Vithika Goyal
- Institute of Ecology and Evolution, University of Oregon, Eugene, United States
| | - Micah A Woods
- Institute of Ecology and Evolution, University of Oregon, Eugene, United States
| | - William A Cresko
- Institute of Ecology and Evolution, University of Oregon, Eugene, United States
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, United States
| |
Collapse
|
3
|
Sołek JM, Nowicka Z, Fendler W, Sadej R, Romanska H, Braun M. Prognostic value of FGFR2 in ER-positive breast cancer is influenced by the profile of stromal gene expression: an in silico analysis based on TCGA data. Contemp Oncol (Pozn) 2025; 28:341-349. [PMID: 39935756 PMCID: PMC11809570 DOI: 10.5114/wo.2024.147003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/22/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction Fibroblast growth factor receptor 2 (FGFR2) activation is associated with endocrine therapy resistance in luminal breast cancer (BC) in vitro, but clinical evidence remains inconsistent. Given the role of FGFRs in mediating tumour microenvironment (TME) interactions, the prognostic value of FGFR2 may depend on the stromal component. This study aimed to validate the association between FGFR-related profile of the stroma and FGFR2 prognostic value in oestrogen receptor-positive invasive ductal carcinoma (IDC). Material and methods An in silico gene expression analysis identified 12 stromal factors (FAP, CXCL12, PDGFRA, COL1A1, HSPG2, CCL2, MMP14, S100A4, MMP9, PDGFA, MCAM, IL6) forming an "FGFR-related profile of the stroma". A cohort of 257 ER+ IDC patients from The Cancer Genome Atlas (TCGA) was analysed. Tumours were clustered using k-means based on stromal gene expression, and Cox proportional hazards regression models were used to assess the association between FGFR2 and overall survival (OS). Results Two clusters of ER+ IDC tumours were identified based on the stromal gene expression profile. While both clusters had similar tumour stages and hormone receptor statuses, multivariable analysis adjusted for clinical factors revealed a significant association between FGFR2 expression and cluster assignment. In Cluster I (high expression of stromal genes), high FGFR2 was linked to poor prognosis, whereas in Cluster II (low expression), high FGFR2 indicated favourable prognosis. FGFR1, FGFR3, and FGFR4 showed no significant prognostic value. Conclusions Stromal profiles modulate the prognostic significance of FGFR2 in luminal breast carcinoma, highlighting the importance of TME profiling for biomarker assessment and explaining inconsistencies in FGFR2 studies.
Collapse
Affiliation(s)
- Julia M. Sołek
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland
| | - Zuzanna Nowicka
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Rafal Sadej
- Laboratory of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Hanna Romanska
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland
| | - Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
4
|
Tasnim M, Wahlquist P, Hill JT. Zebrafish: unraveling genetic complexity through duplicated genes. Dev Genes Evol 2024; 234:99-116. [PMID: 39079985 PMCID: PMC11612004 DOI: 10.1007/s00427-024-00720-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/19/2024] [Indexed: 12/06/2024]
Abstract
The zebrafish is an invaluable model organism for genetic, developmental, and disease research. Although its high conservation with humans is often cited as justification for its use, the zebrafish harbors oft-ignored genetic characteristics that may provide unique insights into gene structure and function. Zebrafish, along with other teleost fish, underwent an additional round of whole genome duplication after their split from tetrapods-resulting in an abundance of duplicated genes when compared to other vertebrates. These duplicated genes have evolved in distinct ways over the ensuing 350 million years. Thus, each gene within a duplicated gene pair has nuanced differences that create a unique identity. By investigating both members of the gene pair together, we can elucidate the mechanisms that underly protein structure and function and drive the complex interplay within biological systems, such as signal transduction cascades, genetic regulatory networks, and evolution of tissue and organ function. It is crucial to leverage such studies to explore these molecular dynamics, which could have far-reaching implications for both basic science and therapeutic development. Here, we will review the role of gene duplications and the existing models for gene divergence and retention following these events. We will also highlight examples within each of these models where studies comparing duplicated genes in the zebrafish have yielded key insights into protein structure, function, and regulation.
Collapse
Affiliation(s)
- Maliha Tasnim
- Department of Cell Biology and Physiology, Brigham Young University, 701 E. University Pkwy, Provo, UT, 84602, USA
| | - Preston Wahlquist
- Department of Cell Biology and Physiology, Brigham Young University, 701 E. University Pkwy, Provo, UT, 84602, USA
| | - Jonathon T Hill
- Department of Cell Biology and Physiology, Brigham Young University, 701 E. University Pkwy, Provo, UT, 84602, USA.
| |
Collapse
|
5
|
Harrison EN, Jay AN, Kent MR, Sukienik TP, LaVigne CA, Kendall GC. Engineering an fgfr4 knockout zebrafish to study its role in development and disease. PLoS One 2024; 19:e0310100. [PMID: 39576839 PMCID: PMC11584112 DOI: 10.1371/journal.pone.0310100] [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: 05/08/2024] [Accepted: 08/20/2024] [Indexed: 11/24/2024] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) has a role in many biological processes, including lipid metabolism, tissue repair, and vertebrate development. In recent years, FGFR4 overexpression and activating mutations have been associated with numerous adult and pediatric cancers. As such, FGFR4 presents an opportunity for therapeutic targeting which is being pursued in clinical trials. To understand the role of FGFR4 signaling in disease and development, we generated and characterized three alleles of fgfr4 knockout zebrafish strains using CRISPR/Cas9. To generate fgfr4 knockout crispants, we injected single-cell wildtype zebrafish embryos with fgfr4 targeting guide RNA and Cas9 proteins, identified adult founders, and outcrossed to wildtype zebrafish to create an F1 generation. The generated mutations introduce a stop codon within the second Ig-like domain of Fgfr4, resulting in a truncated 215, 223, or 228 amino acid Fgfr4 protein compared to 922 amino acids in the full-length protein. All mutant strains exhibited significantly decreased fgfr4 mRNA expression during development, providing evidence for successful knockout of fgfr4 in mutant zebrafish. We found that, consistent with other Fgfr4 knockout animal models, the fgfr4 mutant fish developed normally; however, homozygous fgfr4 mutant zebrafish were significantly smaller than wildtype fish at three months post fertilization. These fgfr4 knockout zebrafish lines are a valuable tool to study the role of FGFR4 in vertebrate development and its viability as a potential therapeutic target in pediatric and adult cancers, as well as other diseases.
Collapse
Affiliation(s)
- Emma N. Harrison
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, OH, United States of America
| | - Amanda N. Jay
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, OH, United States of America
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States of America
| | - Matthew R. Kent
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, OH, United States of America
| | - Talia P. Sukienik
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, OH, United States of America
| | - Collette A. LaVigne
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Genevieve C. Kendall
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, OH, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
6
|
Li H, Dong X, Wang L, Wen H, Qi X, Zhang K, Li Y. Genome-wide identification of Fgfr genes and function analysis of Fgfr4 in myoblasts differentiation of Lateolabrax maculatus. Gene 2024; 927:148717. [PMID: 38908457 DOI: 10.1016/j.gene.2024.148717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Fibroblast growth factor receptors (Fgfrs) are involved in cell proliferation, differentiation, and migration via complex signaling pathways in different tissues. Our previous studies showed that fibroblast growth factor receptor 4 (fgfr4) was detected in the most significant quantitative trait loci (QTL) for growth traits. However, studies focusing on the function of fgfr4 on the growth of bony fish are still limited. In this study, we identified seven fgfr genes in spotted sea bass (Lateolabrax maculatus) genome, namely fgfr1a, fgfr1b, fgfr2, fgfr3, fgfr4, fgfr5a, and fgfr5b. Phylogenetic analysis, syntenic analysis and gene structure analysis were conducted to further support the accuracy of our annotation and classification results. Additionally, fgfr4 showed the highest expression levels among fgfrs during the proliferation and differentiation stages of spotted sea bass myoblasts. To further study the function of fgfr4 in myogenesis, dual-fluorescence in situ hybridization (ISH) assay was conducted, and the results showed co-localization of fgfr4 with marker gene of skeletal muscle satellite cells. By treating differentiating myoblasts cultured in vitro with BLU-554, the mRNA expressions of myogenin (myog) and the numbers of myotubes formed by myoblasts increased significantly compared to negative control group. These results indicated that Fgfr4 inhibits the differentiation of myoblasts in spotted sea bass. Our findings contributed to filling a research gap on fgfr4 in bony fish myogenesis and the theoretical understanding of growth trait regulation of spotted sea bass.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Ximeng Dong
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Lingyu Wang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Kaiqiang Zhang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China; Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China.
| |
Collapse
|
7
|
Healey HM, Penn HB, Small CM, Bassham S, Goyal V, Woods MA, Cresko WA. Single Cell Sequencing Provides Clues about the Developmental Genetic Basis of Evolutionary Adaptations in Syngnathid Fishes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588518. [PMID: 38645265 PMCID: PMC11030337 DOI: 10.1101/2024.04.08.588518] [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
Seahorses, pipefishes, and seadragons are fishes from the family Syngnathidae that have evolved extraordinary traits including male pregnancy, elongated snouts, loss of teeth, and dermal bony armor. The developmental genetic and cellular changes that led to the evolution of these traits are largely unknown. Recent syngnathid genome assemblies revealed suggestive gene content differences and provide the opportunity for detailed genetic analyses. We created a single cell RNA sequencing atlas of Gulf pipefish embryos to understand the developmental basis of four traits: derived head shape, toothlessness, dermal armor, and male pregnancy. We completed marker gene analyses, built genetic networks, and examined spatial expression of select genes. We identified osteochondrogenic mesenchymal cells in the elongating face that express regulatory genes bmp4, sfrp1a, and prdm16. We found no evidence for tooth primordia cells, and we observed re-deployment of osteoblast genetic networks in developing dermal armor. Finally, we found that epidermal cells expressed nutrient processing and environmental sensing genes, potentially relevant for the brooding environment. The examined pipefish evolutionary innovations are composed of recognizable cell types, suggesting derived features originate from changes within existing gene networks. Future work addressing syngnathid gene networks across multiple stages and species is essential for understanding how their novelties evolved.
Collapse
Affiliation(s)
- Hope M Healey
- Institute of Ecology and Evolution, University of Oregon
| | - Hayden B Penn
- Institute of Ecology and Evolution, University of Oregon
| | - Clayton M Small
- Institute of Ecology and Evolution, University of Oregon
- School of Computer and Data Science, University of Oregon
| | - Susan Bassham
- Institute of Ecology and Evolution, University of Oregon
| | - Vithika Goyal
- Institute of Ecology and Evolution, University of Oregon
| | - Micah A Woods
- Institute of Ecology and Evolution, University of Oregon
| | - William A Cresko
- Institute of Ecology and Evolution, University of Oregon
- Knight Campus for Accelerating Scientific Impact, University of Oregon
| |
Collapse
|
8
|
Pan YK. Structure and function of the larval teleost fish gill. J Comp Physiol B 2024; 194:569-581. [PMID: 38584182 DOI: 10.1007/s00360-024-01550-8] [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: 11/29/2023] [Revised: 02/05/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
The fish gill is a multifunctional organ that is important in multiple physiological processes such as gas transfer, ionoregulation, and chemoreception. This characteristic organ of fishes has received much attention, yet an often-overlooked point is that larval fishes in most cases do not have a fully developed gill, and thus larval gills do not function identically as adult gills. In addition, large changes associated with gas exchange and ionoregulation happen in gills during the larval phase, leading to the oxygen and ionoregulatory hypotheses examining the environmental constraint that resulted in the evolution of gills. This review thus focuses exclusively on the larval fish gill of teleosts, summarizing the development of teleost larval fish gills and its function in gas transfer, ionoregulation, and chemoreception, and comparing and contrasting it to adult gills where applicable, while providing some insight into the oxygen vs ionoregulatory hypotheses debate.
Collapse
Affiliation(s)
- Yihang Kevin Pan
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
9
|
Risha KS, Rasal KD, Reang D, Iquebal MA, Sonwane A, Brahmane M, Chaudhari A, Nagpure N. DNA Methylation Profiling in Genetically Selected Clarias magur (Hamilton, 1822) Provides Insights into the Epigenetic Regulation of Growth and Development. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:776-789. [PMID: 39037491 DOI: 10.1007/s10126-024-10346-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
DNA methylation is an epigenetic alteration that impacts gene expression without changing the DNA sequence affecting an organism's phenotype. This study utilized a reduced representation bisulfite sequencing (RRBS) approach to investigate the patterns of DNA methylation in genetically selected Clarias magur stocks. RRBS generated 249.22 million reads, with an average of 490,120 methylation sites detected in various parts of genes, including exons, introns, and intergenic regions. A total of 896 differentially methylated regions (DMRs) were identified; 356 and 540 were detected as hyper-methylated and hypo-methylated regions, respectively. The DMRs and their association with overlapping genes were explored using whole genome data of magur, which revealed 205 genes in exonic, 210 in intronic, and 480 in intergenic regions. The analysis identified the maximum number of genes enriched in biological processes such as RNA biosynthetic process, response to growth factors, nervous system development, neurogenesis, and anatomical structure morphogenesis. Differentially methylated genes (DMGs) such as myrip, mylk3, mafb, egr3, ndnf, meis2a, foxn3, bmp1a, plxna3, fgf6, sipa1l1, mcu, cnot8, trim55b, and myof were associated with growth and development. The selected DMGs were analyzed using real-time PCR, which showed altered mRNA expression levels. This work offers insights into the epigenetic mechanisms governing growth performance regulation in magur stocks. This work provides a valuable resource of epigenetic data that could be integrated into breeding programs to select high-performing individuals.
Collapse
Affiliation(s)
- K Shasti Risha
- Fish Genetics and Biotechnology, ICAR - Central Institute of Fisheries Education, Mumbai, Maharashtra, 400061, India
| | - Kiran D Rasal
- Fish Genetics and Biotechnology, ICAR - Central Institute of Fisheries Education, Mumbai, Maharashtra, 400061, India.
| | - Dhalongsaih Reang
- Fish Genetics and Biotechnology, ICAR - Central Institute of Fisheries Education, Mumbai, Maharashtra, 400061, India
| | - Mir Asif Iquebal
- Centre for Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, India
| | - Arvind Sonwane
- Fish Genetics and Biotechnology, ICAR - Central Institute of Fisheries Education, Mumbai, Maharashtra, 400061, India
| | - Manoj Brahmane
- Fish Genetics and Biotechnology, ICAR - Central Institute of Fisheries Education, Mumbai, Maharashtra, 400061, India
| | - Aparna Chaudhari
- Fish Genetics and Biotechnology, ICAR - Central Institute of Fisheries Education, Mumbai, Maharashtra, 400061, India
| | - Naresh Nagpure
- Fish Genetics and Biotechnology, ICAR - Central Institute of Fisheries Education, Mumbai, Maharashtra, 400061, India
| |
Collapse
|
10
|
González M, Maurelia F, Aguayo J, Amigo R, Arrué R, Gutiérrez JL, Torrejón M, Farkas C, Caprile T. Uncovering the role of the subcommissural organ in early brain development through transcriptomic analysis. Biol Res 2024; 57:49. [PMID: 39068496 PMCID: PMC11282827 DOI: 10.1186/s40659-024-00524-y] [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: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND The significant role of embryonic cerebrospinal fluid (eCSF) in the initial stages of brain development has been thoroughly studied. This fluid contains crucial molecules for proper brain development such as members of the Wnt and FGF families, apolipoproteins, and retinol binding protein. Nevertheless, the source of these molecules remains uncertain since they are present before the formation of the choroid plexus, which is conventionally known as the primary producer of cerebrospinal fluid. The subcommissural organ (SCO) is a highly conserved gland located in the diencephalon and is one of the earliest differentiating brain structures. The SCO secretes molecules into the eCSF, prior to the differentiation of the choroid plexus, playing a pivotal role in the homeostasis and dynamics of this fluid. One of the key molecules secreted by the SCO is SCO-spondin, a protein involved in maintenance of the normal ventricle size, straight spinal axis, neurogenesis, and axonal guidance. Furthermore, SCO secretes transthyretin and basic fibroblast growth factor 2, while other identified molecules in the eCSF could potentially be secreted by the SCO. Additionally, various transcription factors have been identified in the SCO. However, the precise mechanisms involved in the early SCO development are not fully understood. RESULTS To uncover key molecular players and signaling pathways involved in the role of the SCO during brain development, we conducted a transcriptomic analysis comparing the embryonic chick SCO at HH23 and HH30 stages (4 and 7 days respectively). Additionally, a public transcriptomic data from HH30 entire chick brain was used to compare expression levels between SCO and whole brain transcriptome. These analyses revealed that, at both stages, the SCO differentially expresses several members of bone morphogenic proteins, Wnt and fibroblast growth factors families, diverse proteins involved in axonal guidance, neurogenic and differentiative molecules, cell receptors and transcription factors. The secretory pathway is particularly upregulated at stage HH30 while the proliferative pathway is increased at stage HH23. CONCLUSION The results suggest that the SCO has the capacity to secrete several morphogenic molecules to the eCSF prior to the development of other structures, such as the choroid plexus.
Collapse
Affiliation(s)
- Maryori González
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Maurelia
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jaime Aguayo
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roberto Amigo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Rodrigo Arrué
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - José Leonardo Gutiérrez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Marcela Torrejón
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Carlos Farkas
- Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| | - Teresa Caprile
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
11
|
Rojoni SA, Ahmed MT, Rahman M, Hossain MMM, Ali MS, Haq M. Advances of microplastics ingestion on the morphological and behavioral conditions of model zebrafish: A review. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 272:106977. [PMID: 38820743 DOI: 10.1016/j.aquatox.2024.106977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/20/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024]
Abstract
Concerns have been conveyed regarding the availability and hazards of microplastics (MPs) in aquatic biota due to their widespread presence in aquatic habitats. Zebrafish (Danio rerio) are widely used as a model organism to study the adverse impacts of MPs due to their several compelling advantages, such as their small size, ease of breeding, inexpensive maintenance, short life cycle, year-round spawning, high fecundity, fewer legal restrictions, and genetic resemblances to humans. Exposure of organisms to MPs produces physical and chemical toxic effects, including abnormal behavior, oxidative stress, neurotoxicity, genotoxicity, immune toxicity, reproductive imbalance, and histopathological effects. But the severity of the effects is size and concentration-dependent. It has been demonstrated that smaller particles could reach the gut and liver, while larger particles are only confined to the gill, the digestive tract of adult zebrafish. This thorough review encapsulates the current body of literature concerning research on MPs in zebrafish and demonstrates an overview of MPs size and concentration effects on the physiological, morphological, and behavioral characteristics of zebrafish. Finding gaps in the literature paves the way for further investigation.
Collapse
Affiliation(s)
- Suraiya Alam Rojoni
- Department of Fisheries and Marine Bioscience, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Tanvir Ahmed
- Department of Fisheries and Marine Bioscience, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Mostafizur Rahman
- Department of Fisheries and Marine Bioscience, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Mer Mosharraf Hossain
- Department of Fisheries and Marine Bioscience, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Sadek Ali
- Department of Fisheries and Marine Bioscience, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Monjurul Haq
- Department of Fisheries and Marine Bioscience, Jashore University of Science and Technology, Jashore 7408, Bangladesh.
| |
Collapse
|
12
|
Pereur R, Dambroise E. Insights into Craniofacial Development and Anomalies: Exploring Fgf Signaling in Zebrafish Models. Curr Osteoporos Rep 2024; 22:340-352. [PMID: 38739352 DOI: 10.1007/s11914-024-00873-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE OF REVIEW To illustrate the value of using zebrafish to understand the role of the Fgf signaling pathway during craniofacial skeletal development under normal and pathological conditions. RECENT FINDINGS Recent data obtained from studies on zebrafish have demonstrated the genetic redundancy of Fgf signaling pathway and have identified new molecular partners of this signaling during the early stages of craniofacial skeletal development. Studies on zebrafish models demonstrate the involvement of the Fgf signaling pathway at every stage of craniofacial development. They particularly emphasize the central role of Fgf signaling pathway during the early stages of the development, which significantly impacts the formation of the various structures making up the craniofacial skeleton. This partly explains the craniofacial abnormalities observed in disorders associated with FGF signaling. Future research efforts should focus on investigating zebrafish Fgf signaling during more advanced stages, notably by establishing zebrafish models expressing mutations responsible for diseases such as craniosynostoses.
Collapse
Affiliation(s)
- Rachel Pereur
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, Université Paris Cité, INSERM UMR 1163, Imagine Institut, 24 boulevard Montparnasse, 75015, Paris, France
| | - Emilie Dambroise
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, Université Paris Cité, INSERM UMR 1163, Imagine Institut, 24 boulevard Montparnasse, 75015, Paris, France.
| |
Collapse
|
13
|
Jin S, Choe CP. A Potential Role of fgf4, fgf24, and fgf17 in Pharyngeal Pouch Formation in Zebrafish. Dev Reprod 2024; 28:55-65. [PMID: 39055102 PMCID: PMC11268894 DOI: 10.12717/dr.2024.28.2.55] [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: 03/19/2024] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 07/27/2024]
Abstract
In vertebrates, Fgf signaling is essential for the development of pharyngeal pouches, which controls facial skeletal development. Genetically, fgf3 and fgf8 are required for pouch formation in mice and zebrafish. However, loss-of-function phenotypes of fgf3 and fgf8 are milder than expected in mice and zebrafish, which suggests that an additional fgf gene(s) would be involved in pouch formation. Here, we analyzed the expression, regulation, and function of three fgfs, fgf4, fgf24, and fgf17, during pouch development in zebrafish. We find that they are expressed in the distinct regions of pharyngeal endoderm in pouch formation, with fgf4 and fgf17 also being expressed in the adjacent mesoderm, in addition to previously reported endodermal fgf3 and mesodermal fgf8 expression. The endodermal expression of fgf4, fgf24, and fgf17 and the mesodermal expression of fgf4 and fgf17 are positively regulated by Tbx1 but not by Fgf3, in pouch formation. Fgf8 is required to express the endodermal expression of fgf4 and fgf24. Interestingly, however, single mutant, all double mutant combinations, and triple mutant for fgf4, fgf24, and fgf17 do not show any defects in pouches and facial skeletons. Considering a high degree of genetic redundancy in the Fgf signaling components in craniofacial development in zebrafish, our result suggests that fgf4, fgf24, and fgf17 have a potential role for pouch formation, with a redundancy with other fgf gene(s).
Collapse
Affiliation(s)
- Sil Jin
- Division of Applied Life Science,
Gyeongsang National University, Jinju 52828,
Korea
| | - Chong Pyo Choe
- Division of Life Science, Gyeongsang
National University, Jinju 52828,
Korea
- Plant Molecular Biology and Biotechnology
Research Center, Gyeongsang National University,
Jinju 52828, Korea
| |
Collapse
|
14
|
Harrison EN, Jay AN, Kent MR, Sukienik TP, LaVigne CA, Kendall GC. Engineering an fgfr4 knockout zebrafish to study its role in development and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593184. [PMID: 38766056 PMCID: PMC11100669 DOI: 10.1101/2024.05.08.593184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Fibroblast growth factor receptor 4 (FGFR4) has a role in many biological processes, including lipid metabolism, tissue repair, and vertebrate development. In recent years, FGFR4 overexpression and activating mutations have been associated with numerous adult and pediatric cancers. As such, FGFR4 presents an opportunity for therapeutic targeting which is being pursued in clinical trials. To understand the role of FGFR4 signaling in disease and development, we generated and characterized three alleles of fgfr4 knockout zebrafish strains using CRISPR/Cas9. To generate fgfr4 knockout crispants, we injected single-cell wildtype zebrafish embryos with fgfr4 targeting guide RNA and Cas9 proteins, identified adult founders, and outcrossed to wildtype zebrafish to create an F1 generation. The generated mutations introduce a stop codon within the second Ig-like domain of Fgfr4, resulting in a truncated 215, 223, or 228 amino acid Fgfr4 protein compared to 922 amino acids in the full-length protein. All mutant strains exhibited significantly decreased fgfr4 mRNA expression during development, providing evidence for successful knockout of fgfr4 in mutant zebrafish. We found that, consistent with other Fgfr4 knockout animal models, the fgfr4 mutant fish developed normally; however, homozygous fgfr4 mutant zebrafish were significantly smaller than wildtype fish at three months post fertilization. These fgfr4 knockout zebrafish lines are a valuable tool to study the role of FGFR4 in vertebrate development and its viability as a potential therapeutic target in pediatric and adult cancers, as well as other diseases.
Collapse
|
15
|
Zinck NW, McInnis SJL, Franz-Odendaal TA. Intravitreal injection of FGF and TGF-β inhibitors disrupts cranial cartilage development. Differentiation 2023; 133:51-59. [PMID: 37481903 DOI: 10.1016/j.diff.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
Cartilage development is a tightly regulated process that requires the interaction of epithelial and mesenchymal tissues layers to initiate the aggregation of mesenchyme in a condensation. Several signaling molecules have been implicated in cartilage formation including FGFs, WNTs, and members of the TGF-β super family. However, little is known about the earliest signals involved in these initial phases of development. Here we aimed to investigate whether direct intravitreal injection of pharmaceutical inhibitors for FGF and TGF-β signaling would perturb cranial cartilages in zebrafish. Via wholemount bone and cartilage staining, we found effects on multiple cranial cartilage elements. We found no effect on scleral cartilage development, however, the epiphyseal bar, basihyal, and basicapsular cartilages were disrupted. Interestingly, the epiphyseal bar arises from the same progenitor pool as the scleral cartilage, namely, the periocular ectomesenchyme. This study adds to the foundational knowledge about condensation induction of cranial cartilage development and provides insight into the timing and signaling involved in the early development of several craniofacial cartilage elements in zebrafish.
Collapse
Affiliation(s)
- Nicholas W Zinck
- Department of Medical Neuroscience, Dalhousie University, 5850 College Street, Halifax, NS, B3H 4R2, Canada; Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS, B3M 2J6, Canada
| | - Shea J L McInnis
- Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS, B3M 2J6, Canada; Department of Biology, Saint Mary's University, 923 Robie Street, Halifax, NS, B3H 3C3, Canada
| | - Tamara A Franz-Odendaal
- Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS, B3M 2J6, Canada.
| |
Collapse
|
16
|
Čapek D, Safroshkin M, Morales-Navarrete H, Toulany N, Arutyunov G, Kurzbach A, Bihler J, Hagauer J, Kick S, Jones F, Jordan B, Müller P. EmbryoNet: using deep learning to link embryonic phenotypes to signaling pathways. Nat Methods 2023; 20:815-823. [PMID: 37156842 PMCID: PMC10250202 DOI: 10.1038/s41592-023-01873-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 04/05/2023] [Indexed: 05/10/2023]
Abstract
Evolutionarily conserved signaling pathways are essential for early embryogenesis, and reducing or abolishing their activity leads to characteristic developmental defects. Classification of phenotypic defects can identify the underlying signaling mechanisms, but this requires expert knowledge and the classification schemes have not been standardized. Here we use a machine learning approach for automated phenotyping to train a deep convolutional neural network, EmbryoNet, to accurately identify zebrafish signaling mutants in an unbiased manner. Combined with a model of time-dependent developmental trajectories, this approach identifies and classifies with high precision phenotypic defects caused by loss of function of the seven major signaling pathways relevant for vertebrate development. Our classification algorithms have wide applications in developmental biology and robustly identify signaling defects in evolutionarily distant species. Furthermore, using automated phenotyping in high-throughput drug screens, we show that EmbryoNet can resolve the mechanism of action of pharmaceutical substances. As part of this work, we freely provide more than 2 million images that were used to train and test EmbryoNet.
Collapse
Affiliation(s)
- Daniel Čapek
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | | | - Hernán Morales-Navarrete
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
- Centre for the Advanced Study of Collective Behaviour, Konstanz, Germany
| | - Nikan Toulany
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | | | - Anica Kurzbach
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
| | - Johanna Bihler
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Julia Hagauer
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Sebastian Kick
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Felicity Jones
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Ben Jordan
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
| | - Patrick Müller
- Systems Biology of Development, University of Konstanz, Konstanz, Germany.
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany.
- Centre for the Advanced Study of Collective Behaviour, Konstanz, Germany.
| |
Collapse
|
17
|
Truong BT, Shull LC, Lencer E, Bend EG, Field M, Blue EE, Bamshad MJ, Skinner C, Everman D, Schwartz CE, Flanagan-Steet H, Artinger KB. PRDM1 DNA-binding zinc finger domain is required for normal limb development and is disrupted in split hand/foot malformation. Dis Model Mech 2023; 16:dmm049977. [PMID: 37083955 PMCID: PMC10151829 DOI: 10.1242/dmm.049977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/09/2023] [Indexed: 04/22/2023] Open
Abstract
Split hand/foot malformation (SHFM) is a rare limb abnormality with clefting of the fingers and/or toes. For many individuals, the genetic etiology is unknown. Through whole-exome and targeted sequencing, we detected three novel variants in a gene encoding a transcription factor, PRDM1, that arose de novo in families with SHFM or segregated with the phenotype. PRDM1 is required for limb development; however, its role is not well understood and it is unclear how the PRDM1 variants affect protein function. Using transient and stable overexpression rescue experiments in zebrafish, we show that the variants disrupt the proline/serine-rich and DNA-binding zinc finger domains, resulting in a dominant-negative effect. Through gene expression assays, RNA sequencing, and CUT&RUN in isolated pectoral fin cells, we demonstrate that Prdm1a directly binds to and regulates genes required for fin induction, outgrowth and anterior/posterior patterning, such as fgfr1a, dlx5a, dlx6a and smo. Taken together, these results improve our understanding of the role of PRDM1 in the limb gene regulatory network and identified novel PRDM1 variants that link to SHFM in humans.
Collapse
Affiliation(s)
- Brittany T. Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lomeli C. Shull
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ezra Lencer
- Biology Department, Lafayette College, Easton, PA 18042, USA
| | - Eric G. Bend
- Greenwood Genetics Center, Greenwood, SC 29646, USA
| | - Michael Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW 2298, AUS
| | - Elizabeth E. Blue
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Michael J. Bamshad
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | - Kristin B. Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
18
|
Cudak N, López-Delgado AC, Keil S, Knopf F. Fibroblast growth factor pathway component expression in the regenerating zebrafish fin. Gene Expr Patterns 2023; 48:119307. [PMID: 36841347 DOI: 10.1016/j.gep.2023.119307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 02/26/2023]
Abstract
Adult zebrafish regenerate their appendages (fins) after amputation including the regeneration of bone structures (fin rays). Fibroblast growth factor (Fgf) signaling, which is involved in morphogenetic processes during development, has been shown to be essential for the process of fin regeneration. Moreover, mutations in Fgf pathway component genes lead to abnormal skeletal growth in teleosts and mammals, including humans, illustrating the importance of Fgf signaling in the growth control of tissues. Here, we revisited Fgf signaling pathway component expression by RNA in situ hybridization to test for the expression of about half of the ligands and all receptors of the pathway in the regenerating zebrafish fin. Expression patterns of fgf7, fgf10b, fgf12b, fgf17b and fgfr1b have not been reported in the literature before. We summarize and discuss known and novel localization of expression and find that all five Fgf receptors (fgfr1a, fgfr1b, fgfr2, fgfr3 and fgfr4) and most of the tested ligands are expressed in specific regions of the regenerate. Our work provides a basis to study domain specific functions of Fgf signaling in the regenerating teleost appendage.
Collapse
Affiliation(s)
- Nicole Cudak
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alejandra Cristina López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Keil
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
19
|
Zhang YM, Lin CY, Li BZ, Xu WB, Dong WR, Shu MA. Characterization of fibroblast growth factor receptor 4 (FGFR4) from the red swamp crayfish Procambarus clarkii and its role in antiviral and antimicrobial immune responses. J Invertebr Pathol 2023; 196:107865. [PMID: 36436575 DOI: 10.1016/j.jip.2022.107865] [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: 07/15/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022]
Abstract
FGFRs involved multiple physiological processes, such as endocrine homeostasis, wound repair, and cellular behaviors including proliferation, differentiation and survival. In the present study, the homologs of fibroblast growth factor receptor 4 (FGFR4) were identified and characterized from the red swamp crayfish Procambarus clarkii for the first time. The full-length cDNAs of pcFGFR4 were 2878 bp with 2451 bp open reading frame (ORF), respectively. The deduced pcFGFR4 protein contained an immunoglobulin, two immunoglobulin C-2 Type, a transmembrane region and a catalytic domain. Real-time PCR analysis showed that pcFGFR4 were highly expressed in muscle and hemocyte. Moreover, the expression levels of pcFGFR4 in the hepatopancreas and hemocyte were positively stimulated after challenge with Aeromonas hydrophila and WSSV, implying the involvement of pcFGFR4 against bacterial and viral infections in innate immune responses. While pcFGFR4 were silenced in vivo, the expression levels of antimicrobial peptide (AMP) genes (pcALF1-5,8 and pcCrustin1-2) and NF-κB signaling components (pcDrosal and pcRelish) were significantly reduced. Additionally, NF-κB signaling could be markedly activated by overexpression of pcFGFR4 in HEK293T cells. Finally, our results indicated that pcFGFR4 regulated crayfish's innate immunity by modulating NF-κB signaling. These findings may provide new insights into pcFGFR4-mediated signaling cascades in crustaceans and provide a better understanding of crustacean innate immune system.
Collapse
Affiliation(s)
- Yan-Mei Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chen-Yang Lin
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bang-Ze Li
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen-Bin Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wei-Ren Dong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Miao-An Shu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
20
|
Liu Y, Qu M, Jiang H, Schneider R, Qin G, Luo W, Yu H, Zhang B, Wang X, Zhang Y, Zhang H, Zhang Z, Wu Y, Zhang Y, Yin J, Zhang S, Venkatesh B, Roth O, Meyer A, Lin Q. Immunogenetic losses co-occurred with seahorse male pregnancy and mutation in tlx1 accompanied functional asplenia. Nat Commun 2022; 13:7610. [PMID: 36494371 PMCID: PMC9734139 DOI: 10.1038/s41467-022-35338-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
In the highly derived syngnathid fishes (pipefishes, seadragons & seahorses), the evolution of sex-role reversed brooding behavior culminated in the seahorse lineage's male pregnancy, whose males feature a specialized brood pouch into which females deposit eggs during mating. Then, eggs are intimately engulfed by a placenta-like tissue that facilitates gas and nutrient exchange. As fathers immunologically tolerate allogenic embryos, it was suggested that male pregnancy co-evolved with specific immunological adaptations. Indeed, here we show that a specific amino-acid replacement in the tlx1 transcription factor is associated with seahorses' asplenia (loss of spleen, an organ central in the immune system), as confirmed by a CRISPR-Cas9 experiment using zebrafish. Comparative genomics across the syngnathid phylogeny revealed that the complexity of the immune system gene repertoire decreases as parental care intensity increases. The synchronous evolution of immunogenetic alterations and male pregnancy supports the notion that male pregnancy co-evolved with the immunological tolerance of the embryo.
Collapse
Affiliation(s)
- Yali Liu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| | - Meng Qu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Han Jiang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| | - Ralf Schneider
- grid.9764.c0000 0001 2153 9986Marine Evolutionary Ecology, Zoological Institute, Kiel University, 24118 Kiel, Germany
| | - Geng Qin
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Wei Luo
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Haiyan Yu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Bo Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Xin Wang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Yanhong Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Huixian Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Zhixin Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.412785.d0000 0001 0695 6482Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Minato, Tokyo, Japan
| | - Yongli Wu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Yingyi Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| | - Jianping Yin
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Si Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Byrappa Venkatesh
- grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, 138673 Singapore, Singapore
| | - Olivia Roth
- grid.9764.c0000 0001 2153 9986Marine Evolutionary Ecology, Zoological Institute, Kiel University, 24118 Kiel, Germany
| | - Axel Meyer
- grid.9811.10000 0001 0658 7699Department of Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Qiang Lin
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| |
Collapse
|
21
|
Gebuijs L, Wagener FA, Zethof J, Carels CE, Von den Hoff JW, Metz JR. Targeting fibroblast growth factor receptors causes severe craniofacial malformations in zebrafish larvae. PeerJ 2022; 10:e14338. [PMID: 36444384 PMCID: PMC9700454 DOI: 10.7717/peerj.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background and Objective A key pathway controlling skeletal development is fibroblast growth factor (FGF) and FGF receptor (FGFR) signaling. Major regulatory functions of FGF signaling are chondrogenesis, endochondral and intramembranous bone development. In this study we focus on fgfr2, as mutations in this gene are found in patients with craniofacial malformations. The high degree of conservation between FGF signaling of human and zebrafish (Danio rerio) tempted us to investigate effects of the mutated fgfr2 sa10729 allele in zebrafish on cartilage and bone formation. Methods We stained cartilage and bone in 5 days post fertilization (dpf) zebrafish larvae and compared mutants with wildtypes. We also determined the expression of genes related to these processes. We further investigated whether pharmacological blocking of all FGFRs with the inhibitor BGJ398, during 0-12 and 24-36 h post fertilization (hpf), affected craniofacial structure development at 5 dpf. Results We found only subtle differences in craniofacial morphology between wildtypes and mutants, likely because of receptor redundancy. After exposure to BGJ398, we found dose-dependent cartilage and bone malformations, with more severe defects in fish exposed during 0-12 hpf. These results suggest impairment of cranial neural crest cell survival and/or differentiation by FGFR inhibition. Compensatory reactions by upregulation of fgfr1a, fgfr1b, fgfr4, sp7 and dlx2a were found in the 0-12 hpf group, while in the 24-36 hpf group only upregulation of fgf3 was found together with downregulation of fgfr1a and fgfr2. Conclusions Pharmacological targeting of FGFR1-4 kinase signaling causes severe craniofacial malformations, whereas abrogation of FGFR2 kinase signaling alone does not induce craniofacial skeletal abnormalities. These findings enhance our understanding of the role of FGFRs in the etiology of craniofacial malformations.
Collapse
Affiliation(s)
- Liesbeth Gebuijs
- Department of Orthodontics and Craniofacial Biology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands,Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands,Department of Animal Ecology and Physiology, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Frank A. Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands,Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jan Zethof
- Department of Animal Ecology and Physiology, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Carine E. Carels
- Department of Human Genetics and Department of Oral Health Sciences, KU Leuven, Leuven, Belgium
| | - Johannes W. Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands,Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Juriaan R. Metz
- Department of Animal Ecology and Physiology, Radboud University Nijmegen, Nijmegen, Netherlands
| |
Collapse
|
22
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
23
|
Dash S, Trainor PA. Nucleolin loss of function leads to aberrant Fibroblast Growth Factor signaling and craniofacial anomalies. Development 2022; 149:dev200349. [PMID: 35762670 PMCID: PMC9270975 DOI: 10.1242/dev.200349] [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] [Received: 11/09/2021] [Accepted: 04/25/2022] [Indexed: 11/23/2022]
Abstract
Ribosomal RNA (rRNA) transcription and ribosome biogenesis are global processes required for growth and proliferation of all cells, yet perturbation of these processes in vertebrates leads to tissue-specific defects termed ribosomopathies. Mutations in rRNA transcription and processing proteins often lead to craniofacial anomalies; however, the cellular and molecular reasons for these defects are poorly understood. Therefore, we examined the function of the most abundant nucleolar phosphoprotein, Nucleolin (Ncl), in vertebrate development. ncl mutant (ncl-/-) zebrafish present with craniofacial anomalies such as mandibulofacial hypoplasia. We observed that ncl-/- mutants exhibited decreased rRNA synthesis and p53-dependent apoptosis, consistent with a role in ribosome biogenesis. However, we found that Nucleolin also performs functions not associated with ribosome biogenesis. We discovered that the half-life of fgf8a mRNA was reduced in ncl-/- mutants, which perturbed Fgf signaling, resulting in misregulated Sox9a-mediated chondrogenesis and Runx2-mediated osteogenesis. Consistent with this model, exogenous FGF8 treatment significantly rescued the cranioskeletal phenotype in ncl-/- zebrafish, suggesting that Nucleolin regulates osteochondroprogenitor differentiation. Our work has therefore uncovered tissue-specific functions for Nucleolin in rRNA transcription and post-transcriptional regulation of growth factor signaling during embryonic craniofacial development.
Collapse
Affiliation(s)
- Soma Dash
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
24
|
Li K, Fan L, Tian Y, Lou S, Li D, Ma L, Wang L, Pan Y. Application of zebrafish in the study of craniomaxillofacial developmental anomalies. Birth Defects Res 2022; 114:583-595. [PMID: 35437950 DOI: 10.1002/bdr2.2014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/18/2022] [Accepted: 04/03/2022] [Indexed: 12/13/2022]
Abstract
Craniomaxillofacial developmental anomalies are one of the most prevalent congenital defects worldwide and could result from any disruption of normal development processes, which is generally influenced by interactions between genes and the environment. Currently, with the advances in genetic screening strategies, an increasing number of novel variants and their roles in orofacial diseases have been explored. Zebrafish is recognized as a powerful animal model, and its homologous genes and similar oral structure and development process provide an ideal platform for studying the contributions of genetic and environmental factors to human craniofacial malformations. Here, we reviewed zebrafish models for the study of craniomaxillofacial developmental anomalies, such as human nonsyndromic cleft lip with or without an affected palate and jaw and tooth developmental anomalies. Due to its potential for gene expression and regulation research, zebrafish may provide new perspectives for understanding craniomaxillofacial diseaseand its treatment.
Collapse
Affiliation(s)
- Kang Li
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Liwen Fan
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.,Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yu Tian
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.,Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Shu Lou
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.,Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Dandan Li
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.,Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Lan Ma
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.,Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Lin Wang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.,Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Yongchu Pan
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.,Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Hidalgo-Sánchez M, Andreu-Cervera A, Villa-Carballar S, Echevarria D. An Update on the Molecular Mechanism of the Vertebrate Isthmic Organizer Development in the Context of the Neuromeric Model. Front Neuroanat 2022; 16:826976. [PMID: 35401126 PMCID: PMC8987131 DOI: 10.3389/fnana.2022.826976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
A crucial event during the development of the central nervous system (CNS) is the early subdivision of the neural tube along its anterior-to-posterior axis to form neuromeres, morphogenetic units separated by transversal constrictions and programed for particular genetic cascades. The narrower portions observed in the developing neural tube are responsible for relevant cellular and molecular processes, such as clonal restrictions, expression of specific regulatory genes, and differential fate specification, as well as inductive activities. In this developmental context, the gradual formation of the midbrain-hindbrain (MH) constriction has been an excellent model to study the specification of two major subdivisions of the CNS containing the mesencephalic and isthmo-cerebellar primordia. This MH boundary is coincident with the common Otx2-(midbrain)/Gbx2-(hindbrain) expressing border. The early interactions between these two pre-specified areas confer positional identities and induce the generation of specific diffusible morphogenes at this interface, in particular FGF8 and WNT1. These signaling pathways are responsible for the gradual histogenetic specifications and cellular identity acquisitions with in the MH domain. This review is focused on the cellular and molecular mechanisms involved in the specification of the midbrain/hindbrain territory and the formation of the isthmic organizer. Emphasis will be placed on the chick/quail chimeric experiments leading to the acquisition of the first fate mapping and experimental data to, in this way, better understand pioneering morphological studies and innovative gain/loss-of-function analysis.
Collapse
Affiliation(s)
- Matías Hidalgo-Sánchez
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
- *Correspondence: Matías Hidalgo-Sánchez Diego Echevarria
| | - Abraham Andreu-Cervera
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Sergio Villa-Carballar
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Diego Echevarria
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
- *Correspondence: Matías Hidalgo-Sánchez Diego Echevarria
| |
Collapse
|
26
|
Paudel S, Gjorcheska S, Bump P, Barske L. Patterning of cartilaginous condensations in the developing facial skeleton. Dev Biol 2022; 486:44-55. [DOI: 10.1016/j.ydbio.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022]
|
27
|
Systematic Pharmacology-Based Strategy to Explore the Molecular Network Mechanism of Modified Taohong Siwu Decoction in the Treatment of Premature Ovarian Failure. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3044463. [PMID: 35096106 PMCID: PMC8799328 DOI: 10.1155/2022/3044463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To explore the molecular network mechanism of modified Taohong Siwu Decoction (MTHSWD) to interfere with premature ovarian failure based on systematic pharmacological strategy. METHODS The network pharmacology strategy was used to explore the potential mechanism of MTHSWD intervention in POF, and then it was verified through animal experiments. Mouse zona pellucida 3 was used as an antigen to subcutaneously immunize BALB/c female mice to establish an immune POF model. Mice were divided into MTHSWD low-, medium-, and high-dose groups, positive control group, model group, and normal group. After 30 days of drug intervention, ovarian tissue was taken for pathological hematoxylin-eosin (HE) staining, and immunohistochemical methods were used to detect the expression of TGF-β1 and TGF-βRII and Smad2/3 protein expression in follicular wall granular cells and ovarian tissue, respectively. RESULTS Network pharmacology studies have shown that MTHSWD may interfere with the TGF-β signaling pathway. Animal experimental research shows that, compared with the model group, the number of ovarian mature follicles in the MTHSWD groups and the positive group was significantly increased, and the number of atresia follicles decreased. Immunohistochemistry showed that, compared with the control group, the expression of TGF-β1, TGF-βRII, and Smad2/3 in the follicular wall granulosa cells and ovarian tissues of MTHSWD groups was significantly higher than that of the model group (P < 0.05). CONCLUSION MTHSWD may improve the ovarian function of POF mice by upregulating the protein expression of granulosa cells TGF-β1, TGF-βRII, and Smad2/3.
Collapse
|
28
|
Jones WD, Mullins MC. Cell signaling pathways controlling an axis organizing center in the zebrafish. Curr Top Dev Biol 2022; 150:149-209. [DOI: 10.1016/bs.ctdb.2022.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
29
|
Klingbeil K, Nguyen TQ, Fahrner A, Guthmann C, Wang H, Schoels M, Lilienkamp M, Franz H, Eckert P, Walz G, Yakulov TA. Corpuscles of Stannius development requires FGF signaling. Dev Biol 2021; 481:160-171. [PMID: 34666023 DOI: 10.1016/j.ydbio.2021.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/06/2021] [Accepted: 10/11/2021] [Indexed: 01/02/2023]
Abstract
The corpuscles of Stannius (CS) represent a unique endocrine organ of teleostean fish that secrets stanniocalcin-1 (Stc1) to maintain calcium homeostasis. Appearing at 20-25 somite stage in the distal zebrafish pronephros, stc1-expressing cells undergo apical constriction, and are subsequently extruded to form a distinct gland on top of the distal pronephric tubules at 50 h post fertilization (hpf). Several transcription factors (e.g. Hnf1b, Irx3b, Tbx2a/b) and signaling pathways (e.g. Notch) control CS development. We report now that Fgf signaling is required to commit tubular epithelial cells to differentiate into stc1-expressing CS cells. Inhibition of Fgf signaling by SU5402, dominant-negative Fgfr1, or depletion of fgf8a prevented CS formation and stc1 expression. Ablation experiments revealed that CS have the ability to partially regenerate via active cell migration involving extensive filopodia and lamellipodia formation. Activation of Wnt signaling curtailed stc1 expression, but had no effect on CS formation. Thus, our observations identify Fgf signaling as a crucial component of CS cell fate commitment.
Collapse
Affiliation(s)
- Konstantin Klingbeil
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Thanh Quang Nguyen
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Andreas Fahrner
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Clara Guthmann
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Hui Wang
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Maximilian Schoels
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Miriam Lilienkamp
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Henriette Franz
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Priska Eckert
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Albertstrasse 19, 79104, Freiburg, Germany
| | - Toma Antonov Yakulov
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
30
|
Nguyen TN, Hoang HD. Exome sequencing revealed the potential causal mutation in a Vietnamese patient with Apert syndrome. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2020.100995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
31
|
Truong BT, Artinger KB. The power of zebrafish models for understanding the co-occurrence of craniofacial and limb disorders. Genesis 2021; 59:e23407. [PMID: 33393730 PMCID: PMC8153179 DOI: 10.1002/dvg.23407] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Craniofacial and limb defects are two of the most common congenital anomalies in the general population. Interestingly, these defects are not mutually exclusive. Many patients with craniofacial phenotypes, such as orofacial clefting and craniosynostosis, also present with limb defects, including polydactyly, syndactyly, brachydactyly, or ectrodactyly. The gene regulatory networks governing craniofacial and limb development initially seem distinct from one another, and yet these birth defects frequently occur together. Both developmental processes are highly conserved among vertebrates, and zebrafish have emerged as an advantageous model due to their high fecundity, relative ease of genetic manipulation, and transparency during development. Here we summarize studies that have used zebrafish models to study human syndromes that present with both craniofacial and limb phenotypes. We discuss the highly conserved processes of craniofacial and limb/fin development and describe recent zebrafish studies that have explored the function of genes associated with human syndromes with phenotypes in both structures. We attempt to identify commonalities between the two to help explain why craniofacial and limb anomalies often occur together.
Collapse
Affiliation(s)
- Brittany T. Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
32
|
Domenichini A, Casari I, Simpson PV, Desai NM, Chen L, Dustin C, Edmands JS, van der Vliet A, Mohammadi M, Massi M, Falasca M. Rhenium N-heterocyclic carbene complexes block growth of aggressive cancers by inhibiting FGFR- and SRC-mediated signalling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:276. [PMID: 33287862 PMCID: PMC7720599 DOI: 10.1186/s13046-020-01777-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Platinum-based anticancer drugs have been at the frontline of cancer therapy for the last 40 years, and are used in more than half of all treatments for different cancer types. However, they are not universally effective, and patients often suffer severe side effects because of their lack of cellular selectivity. There is therefore a compelling need to investigate the anticancer activity of alternative metal complexes. Here we describe the potential anticancer activity of rhenium-based complexes with preclinical efficacy in different types of solid malignancies. METHODS Kinase profile assay of rhenium complexes. Toxicology studies using zebrafish. Analysis of the growth of pancreatic cancer cell line-derived xenografts generated in zebrafish and in mice upon exposure to rhenium compounds. RESULTS We describe rhenium complexes which block cancer proliferation in vitro by inhibiting the signalling cascade induced by FGFR and Src. Initially, we tested the toxicity of rhenium complexes in vivo using a zebrafish model and identified one compound that displays anticancer activity with low toxicity even in the high micromolar range. Notably, the rhenium complex has anticancer activity in very aggressive cancers such as pancreatic ductal adenocarcinoma and neuroblastoma. We demonstrate the potential efficacy of this complex via a significant reduction in cancer growth in mouse xenografts. CONCLUSIONS Our findings provide a basis for the development of rhenium-based chemotherapy agents with enhanced selectivity and limited side effects compared to standard platinum-based drugs.
Collapse
Affiliation(s)
- Alice Domenichini
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia
| | - Ilaria Casari
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia
| | - Peter V Simpson
- Curtin Institute of Functional Molecules and Interfaces, Department of Chemistry, Curtin University, Perth, WA, 6102, Australia
| | - Nima Maheshkumar Desai
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia
| | - Lingfeng Chen
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Christopher Dustin
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Jeanne S Edmands
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Moosa Mohammadi
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Massimiliano Massi
- Curtin Institute of Functional Molecules and Interfaces, Department of Chemistry, Curtin University, Perth, WA, 6102, Australia
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia.
| |
Collapse
|
33
|
Zhang J, Qi J, Shi F, Pan H, Liu M, Tian R, Geng Y, Li H, Qu Y, Chen J, Seim I, Li M. Insights into the Evolution of Neoteny from the Genome of the Asian Icefish Protosalanx chinensis. iScience 2020; 23:101267. [PMID: 32593955 PMCID: PMC7327861 DOI: 10.1016/j.isci.2020.101267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/28/2020] [Accepted: 06/08/2020] [Indexed: 12/23/2022] Open
Abstract
Salangids, known as Asian icefishes, represent a peculiar radiation within the bony fish order Protacanthopterygii where adult fish retain larval characteristics such as transparent and miniaturized bodies and a cartilaginous endoskeleton into adulthood. Here, we report a de novo genome of Protosalanx chinensis, the most widely distributed salangid lineage. The P. chinensis genome assembly is more contiguous and complete than a previous assembly. We estimate that P. chinensis, salmons, trouts, and pikes diverged from a common ancestor 185 million years ago. A juxtaposition with other fish genomes revealed loss of the genes encoding ectodysplasin-A receptor (EDAR), SCPP1, and four Hox proteins and likely lack of canonical fibroblast growth factor 5 (FGF5) function. We also report genomic variations of P. chinensis possibly reflecting the immune system repertoire of a species with a larval phenotype in sexually mature individuals. The new Asian icefish reference genome provides a solid foundation for future studies.
Collapse
Affiliation(s)
- Jie Zhang
- Chinese Academy of Sciences Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Beijing 100101, China.
| | - Jiwei Qi
- Chinese Academy of Sciences Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Beijing 100101, China
| | - Fanglei Shi
- Chinese Academy of Sciences Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huijuan Pan
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing 100083, China
| | - Meng Liu
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Ran Tian
- Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing 210046, China
| | - Yuepan Geng
- Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing 210046, China
| | - Huaying Li
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Yujie Qu
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Jinping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource, Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Guangdong Institute of Applied Biological Resources, Guangzhou 510260, China.
| | - Inge Seim
- Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing 210046, China; Comparative and Endocrine Biology Laboratory, Translational Research Institute-Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Woolloongabba, QLD 4102, Australia.
| | - Ming Li
- Chinese Academy of Sciences Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Beijing 100101, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
34
|
Multiple epithelia are required to develop teeth deep inside the pharynx. Proc Natl Acad Sci U S A 2020; 117:11503-11512. [PMID: 32398375 DOI: 10.1073/pnas.2000279117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
To explain the evolutionary origin of vertebrate teeth from odontodes, it has been proposed that competent epithelium spread into the oropharyngeal cavity via the mouth and other possible channels such as the gill slits [Huysseune et al., 2009, J. Anat. 214, 465-476]. Whether tooth formation deep inside the pharynx in extant vertebrates continues to require external epithelia has not been addressed so far. Using zebrafish we have previously demonstrated that cells derived from the periderm penetrate the oropharyngeal cavity via the mouth and via the endodermal pouches and connect to periderm-like cells that subsequently cover the entire endoderm-derived pharyngeal epithelium [Rosa et al., 2019, Sci. Rep. 9, 10082]. We now provide conclusive evidence that the epithelial component of pharyngeal teeth in zebrafish (the enamel organ) is derived from medial endoderm, as hitherto assumed based on position deep in the pharynx. Yet, dental morphogenesis starts only after the corresponding endodermal pouch (pouch 6) has made contact with the skin ectoderm, and only after periderm-like cells have covered the prospective tooth-forming endodermal epithelium. Manipulation of signaling pathways shown to adversely affect tooth development indicates they act downstream of these events. We demonstrate that pouch-ectoderm contact and the presence of a periderm-like layer are both required, but not sufficient, for tooth initiation in the pharynx. We conclude that the earliest interactions to generate pharyngeal teeth encompass those between different epithelial populations (skin ectoderm, endoderm, and periderm-like cells in zebrafish), in addition to the epithelial-mesenchymal interactions that govern the formation of all vertebrate teeth.
Collapse
|