1
|
Meng X, Su S, Wei X, Wang S, Guo T, Li J, Song H, Wang M, Wang Z. Exposure to bisphenol A alternatives bisphenol AF and fluorene-9-bisphenol induces gonadal injuries in male zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114634. [PMID: 36801538 DOI: 10.1016/j.ecoenv.2023.114634] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Bisphenol A (BPA), present in many household products, can damage the male reproductive system. Accordingly, we summarized urine samples from 6921 human in National Health and Nutrition Examination Survey and found urinary BPA levels were inversely linked with blood testosterone in the children group. Currently, BPA replacements, such as fluorene-9-bisphenol (BHPF) and Bisphenol AF (BPAF), have been introduced to produce "BPA-free" products. Here we demonstrated that BPAF and BHPF could induce delayed gonadal migration and reduce the number of progenitors of germ cell lineage in zebrafish larvae. A close receptor analysis study reveals that BHPF and BPAF can strongly bind to androgen receptors, leading to the downregulation of meiosis-related genes and the overexpression of inflammatory markers. Furthermore, BPAF and BPHF can induce activation of the gonadal axis via negative feedback, leading to the hypersecretion of some upstream hormones and an increase in the expression of upstream hormone receptors. Our findings call for further research on the toxicological effects of BHPF and BPAF on human health and recommend that BPA replacements be investigated for anti-estrogenic action.
Collapse
Affiliation(s)
- Xiangyu Meng
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Shifeng Su
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Xiyi Wei
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Shangqian Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Tao Guo
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Junjian Li
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang Province, China
| | - Huaidong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Mengjing Wang
- State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon 999077, Hong Kong, China.
| | - Zengjun Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China.
| |
Collapse
|
2
|
Heikes KL, Game M, Smith FW, Goldstein B. The embryonic origin of primordial germ cells in the tardigrade Hypsibius exemplaris. Dev Biol 2023; 497:42-58. [PMID: 36893882 DOI: 10.1016/j.ydbio.2023.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/09/2023]
Abstract
Primordial germ cells (PGCs) give rise to gametes - cells necessary for the propagation and fertility of diverse organisms. Current understanding of PGC development is limited to the small number of organisms whose PGCs have been identified and studied. Expanding the field to include little-studied taxa and emerging model organisms is important to understand the full breadth of the evolution of PGC development. In the phylum Tardigrada, no early cell lineages have been identified to date using molecular markers. This includes the PGC lineage. Here, we describe PGC development in the model tardigrade Hypsibius exemplaris. The four earliest-internalizing cells (EICs) exhibit PGC-like behavior and nuclear morphology. The location of the EICs is enriched for mRNAs of conserved PGC markers wiwi1 (water bear piwi 1) and vasa. At early stages, both wiwi1 and vasa mRNAs are detectable uniformly in embryos, which suggests that these mRNAs do not serve as localized determinants for PGC specification. Only later are wiwi1 and vasa enriched in the EICs. Finally, we traced the cells that give rise to the four PGCs. Our results reveal the embryonic origin of the PGCs of H. exemplaris and provide the first molecular characterization of an early cell lineage in the tardigrade phylum. We anticipate that these observations will serve as a basis for characterizing the mechanisms of PGC development in this animal.
Collapse
Affiliation(s)
- Kira L Heikes
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mandy Game
- Biology Department, University of North Florida, Jacksonville, FL, USA
| | - Frank W Smith
- Biology Department, University of North Florida, Jacksonville, FL, USA
| | - Bob Goldstein
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Heikes KL, Game M, Smith FW, Goldstein B. The Embryonic Origin of Primordial Germ Cells in the Tardigrade Hypsibius exemplaris. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.02.522500. [PMID: 36824831 PMCID: PMC9948961 DOI: 10.1101/2023.01.02.522500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Primordial germ cells (PGCs) give rise to gametes â€" cells necessary for the propagation and fertility of diverse organisms. Current understanding of PGC development is limited to the small number of organisms whose PGCs have been identified and studied. Expanding the field to include little-studied taxa and emerging model organisms is important to understand the full breadth of the evolution of PGC development. In the phylum Tardigrada, no early cell lineages have been identified to date using molecular markers. This includes the PGC lineage. Here, we describe PGC development in the model tardigrade Hypsibius exemplaris . The four earliest-internalizing cells (EICs) exhibit PGC-like behavior and nuclear morphology. The location of the EICs is enriched for mRNAs of conserved PGC markers wiwi1 (water bear piwi 1) and vasa . At early stages, both wiwi1 and vasa mRNAs are detectable uniformly in embryos, which suggests that these mRNAs do not serve as localized determinants for PGC specification. Only later are wiwi1 and vasa enriched in the EICs. Finally, we traced the cells that give rise to the four PGCs. Our results reveal the embryonic origin of the PGCs of H. exemplaris and provide the first molecular characterization of an early cell lineage in the tardigrade phylum. We anticipate that these observations will serve as a basis for characterizing the mechanisms of PGC development in this animal.
Collapse
Affiliation(s)
- Kira L. Heikes
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mandy Game
- Biology Department, University of North Florida, Jacksonville, FL, USA
| | - Frank W. Smith
- Biology Department, University of North Florida, Jacksonville, FL, USA
| | - Bob Goldstein
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
4
|
Jiang Z, El-Brolosy MA, Serobyan V, Welker JM, Retzer N, Dooley CM, Jakutis G, Juan T, Fukuda N, Maischein HM, Balciunas D, Stainier DY. Parental mutations influence wild-type offspring via transcriptional adaptation. SCIENCE ADVANCES 2022; 8:eabj2029. [PMID: 36427314 PMCID: PMC9699682 DOI: 10.1126/sciadv.abj2029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
Transgenerational epigenetic inheritance (TEI) is mostly discussed in the context of physiological or environmental factors. Here, we show intergenerational and transgenerational inheritance of transcriptional adaptation (TA), a process whereby mutant messenger RNA (mRNA) degradation affects gene expression, in nematodes and zebrafish. Wild-type offspring of animals heterozygous for mRNA-destabilizing alleles display increased expression of adapting genes. Notably, offspring of animals heterozygous for nontranscribing alleles do not display this response. Germline-specific mutations are sufficient to induce TA in wild-type offspring, indicating that, at least for some genes, mutations in somatic tissues are not necessary for this process. Microinjecting total RNA from germ cells of TA-displaying heterozygous zebrafish can trigger TA in wild-type embryos and in their progeny, suggesting a model whereby mutant mRNAs in the germline trigger a TA response that can be epigenetically inherited. In sum, this previously unidentified mode of TEI reveals a means by which parental mutations can modulate the offspring's transcriptome.
Collapse
Affiliation(s)
- Zhen Jiang
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Mohamed A. El-Brolosy
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Vahan Serobyan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Jordan M. Welker
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Nicholas Retzer
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Christopher M. Dooley
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Gabrielius Jakutis
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Thomas Juan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Nana Fukuda
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Darius Balciunas
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- Life Sciences Centre, Vilnius University, Vilnius, Lithuania
| | - Didier Y.R. Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| |
Collapse
|
5
|
Schick J, Raz E. Blebs—Formation, Regulation, Positioning, and Role in Amoeboid Cell Migration. Front Cell Dev Biol 2022; 10:926394. [PMID: 35912094 PMCID: PMC9337749 DOI: 10.3389/fcell.2022.926394] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
In the context of development, tissue homeostasis, immune surveillance, and pathological conditions such as cancer metastasis and inflammation, migrating amoeboid cells commonly form protrusions called blebs. For these spherical protrusions to inflate, the force for pushing the membrane forward depends on actomyosin contraction rather than active actin assembly. Accordingly, blebs exhibit distinct dynamics and regulation. In this review, we first examine the mechanisms that control the inflation of blebs and bias their formation in the direction of the cell’s leading edge and present current views concerning the role blebs play in promoting cell locomotion. While certain motile amoeboid cells exclusively form blebs, others form blebs as well as other protrusion types. We describe factors in the environment and cell-intrinsic activities that determine the proportion of the different forms of protrusions cells produce.
Collapse
|
6
|
Robertson TF, Huttenlocher A. Real-time imaging of inflammation and its resolution: It's apparent because it's transparent. Immunol Rev 2022; 306:258-270. [PMID: 35023170 PMCID: PMC8855992 DOI: 10.1111/imr.13061] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
The ability to directly observe leukocyte behavior in vivo has dramatically expanded our understanding of the immune system. Zebrafish are particularly amenable to the high-resolution imaging of leukocytes during both homeostasis and inflammation. Due to its natural transparency, intravital imaging in zebrafish does not require any surgical manipulation. As a result, zebrafish are particularly well-suited for the long-term imaging required to observe the temporal and spatial events during the onset and resolution of inflammation. Here, we review major insights about neutrophil and macrophage function gained from real-time imaging of zebrafish. We discuss neutrophil reverse migration, the process whereby neutrophils leave sites of tissue damage and resolve local inflammation. Further, we discuss the current tools available for investigating immune function in zebrafish and how future studies that simultaneously image multiple leukocyte subsets can be used to further dissect mechanisms that regulate both the onset and resolution of inflammation.
Collapse
Affiliation(s)
- Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
7
|
Zhu Y. Metalloproteases in gonad formation and ovulation. Gen Comp Endocrinol 2021; 314:113924. [PMID: 34606745 PMCID: PMC8576836 DOI: 10.1016/j.ygcen.2021.113924] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 01/13/2023]
Abstract
Changes in expression or activation of various metalloproteases including matrix metalloproteases (Mmp), a disintegrin and metalloprotease (Adam) and a disintegrin and metalloprotease with thrombospondin motif (Adamts), and their endogenous inhibitors (tissue inhibitors of metalloproteases, Timp), have been shown to be critical for ovulation in various species from studies in past decades. Some of these metalloproteases such as Adamts1, Adamts9, Mmp2, and Mmp9 have also been shown to be regulated by luteinizing hormone (LH) and/or progestin, which are essential triggers for ovulation in all vertebrate species. Most of these metalloproteases also express broadly in various tissues and cells including germ cells and somatic gonad cells. Thus, metalloproteases likely play roles in gonad formation processes comprising primordial germ cell (PGC) migration, development of germ and somatic cells, and sex determination. However, our knowledge on the functions and mechanisms of metalloproteases in these processes in vertebrates is still lacking. This review will summarize our current knowledge on the metalloproteases in ovulation and gonad formation with emphasis on PGC migration and germ cell development.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Biology, East Carolina University, Greenville, NC 27858, USA.
| |
Collapse
|
8
|
Carver JJ, He Y, Zhu Y. Delay in primordial germ cell migration in adamts9 knockout zebrafish. Sci Rep 2021; 11:8545. [PMID: 33879810 PMCID: PMC8058341 DOI: 10.1038/s41598-021-88024-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/05/2021] [Indexed: 11/09/2022] Open
Abstract
Adamts9 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 9) is one of a few metalloproteinases structurally conserved from C. elegans to humans and is indispensable in germ cell migration in invertebrates. However, adamts9's roles in germ cell migration in vertebrates has not been examined. In the present study, we found zygotic expression of adamts9 started around the germ ring stage and reached peak levels at 3 days post fertilization (dpf) in zebrafish. The migration of primordial germ cells (PGC) was completed within 24 hours (h) in wildtype siblings, while a delay in PGC migration was found at 15 and 24-h post-fertilization (hpf) in the Adamts9 knockout (KO). However, the delayed PGC migration in Adamts9 KO disappeared at 48 hpf. Our study suggests a conserved function of Adamts9 in germ cell migration among invertebrates and vertebrates. In addition, our results also suggest that Adamts9 is not essential for germ cell migration as reported in C. elegans, possibly due to expansion of Adamts family members and compensatory roles from other metalloproteinases in vertebrates. Further studies are required in order to elucidate the functions and mechanisms of metalloproteinases in germ cell migration and gonad formation in vertebrates.
Collapse
Affiliation(s)
- Jonathan J Carver
- Department of Biology, Howell Science Complex, East Carolina University, 1000 E. 5th Street, Greenville, NC, 27858, USA
| | - Yuanfa He
- Department of Biology, Howell Science Complex, East Carolina University, 1000 E. 5th Street, Greenville, NC, 27858, USA.,College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yong Zhu
- Department of Biology, Howell Science Complex, East Carolina University, 1000 E. 5th Street, Greenville, NC, 27858, USA.
| |
Collapse
|
9
|
Pal DS, Li X, Banerjee T, Miao Y, Devreotes PN. The excitable signal transduction networks: movers and shapers of eukaryotic cell migration. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2020; 63:407-416. [PMID: 31840779 DOI: 10.1387/ijdb.190265pd] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In response to a variety of external cues, eukaryotic cells display varied migratory modes to perform their physiological functions during development and in the adult. Aberrations in cell migration result in embryonic defects and cancer metastasis. The molecular components involved in cell migration are remarkably conserved between the social amoeba Dictyostelium and mammalian cells. This makes the amoeba an excellent model system for studies of eukaryotic cell migration. These migration-associated components can be grouped into three networks: input, signal transduction and cytoskeletal. In migrating cells, signal transduction events such as Ras or PI3K activity occur at the protrusion tips, referred to as 'front', whereas events such as dissociation of PTEN from these regions are referred to as 'back'. Asymmetric distribution of such front and back events is crucial for establishing polarity and guiding cell migration. The triggering of these signaling events displays properties of biochemical excitability including all-or-nothing responsiveness to suprathreshold stimuli, refractoriness, and wave propagation. These signal transduction waves originate from a point and propagate towards the edge of the cell, thereby driving cytoskeletal activity and cellular protrusions. Any change in the threshold for network activation alters the range of the propagating waves and the size of cellular protrusions which gives rise to various migratory modes in cells. Thus, this review highlights excitable signal transduction networks as key players for coordinating cytoskeletal activities to drive cell migration in all eukaryotes.
Collapse
Affiliation(s)
- Dhiman S Pal
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
10
|
Grimaldi C, Raz E. Germ cell migration-Evolutionary issues and current understanding. Semin Cell Dev Biol 2019; 100:152-159. [PMID: 31864795 DOI: 10.1016/j.semcdb.2019.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 11/19/2022]
Abstract
In many organisms, primordial germ cells (PGCs) are specified at a different location than where the gonad forms, meaning that PGCs must migrate toward the gonad within the early developing embryo. Following species-specific paths, PGCs can be passively carried by surrounding tissues and also perform active migration. When PGCs actively migrate through and along a variety of embryonic structures in different organisms, they adopt an ancestral robust migration mode termed "amoeboid motility", which allows cells to migrate within diverse environments. In this review, we discuss the possible significance of the PGC migration process in facilitating the evolution of animal body shape. In addition, we summarize the latest findings relevant for the molecular and cellular mechanisms controlling the movement and the directed migration of PGCs in different species.
Collapse
Affiliation(s)
- Cecilia Grimaldi
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, 48149, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, 48149, Germany.
| |
Collapse
|
11
|
Zhu J, Zhang D, Liu X, Yu G, Cai X, Xu C, Rong F, Ouyang G, Wang J, Xiao W. Zebrafish prmt5 arginine methyltransferase is essential for germ cell development. Development 2019; 146:dev.179572. [PMID: 31533925 DOI: 10.1242/dev.179572] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/04/2019] [Indexed: 01/18/2023]
Abstract
Protein arginine methyltransferase 5 (Prmt5), a type II arginine methyltransferase, symmetrically dimethylates arginine in nuclear and cytoplasmic proteins. Prmt5 is involved in a variety of cellular processes, including ribosome biogenesis, cellular differentiation, germ cell development and tumorigenesis. However, the mechanisms by which prmt5 influences cellular processes have remained unclear. Here, prmt5 loss in zebrafish led to the expression of an infertile male phenotype due to a reduction in germ cell number, an increase in germ cell apoptosis and the failure of gonads to differentiate into normal testes or ovaries. Moreover, arginine methylation of the germ cell-specific proteins Zili and Vasa, as well as histones H3 (H3R8me2s) and H4 (H4R3me2s), was reduced in the gonads of prmt5-null zebrafish. This resulted in the downregulation of several Piwi pathway proteins, including Zili, and Vasa. In addition, various genes related to meiosis, gonad development and sexual differentiation were dysregulated in the gonads of prmt5-null zebrafish. Our results revealed a novel mechanism associated with prmt5, i.e. prmt5 apparently controls germ cell development in vertebrates by catalyzing arginine methylation of the germline-specific proteins Zili and Vasa.
Collapse
Affiliation(s)
- Junji Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Dawei Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Guangqing Yu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Xiaolian Cai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Chenxi Xu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Fangjing Rong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Gang Ouyang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China .,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China.,The Key of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
12
|
Malhotra D, Shin J, Solnica-Krezel L, Raz E. Spatio-temporal regulation of concurrent developmental processes by generic signaling downstream of chemokine receptors. eLife 2018; 7:e33574. [PMID: 29873633 PMCID: PMC5990360 DOI: 10.7554/elife.33574] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/19/2018] [Indexed: 01/09/2023] Open
Abstract
Chemokines are secreted proteins that regulate a range of processes in eukaryotic organisms. Interestingly, different chemokine receptors control distinct biological processes, and the same receptor can direct different cellular responses, but the basis for this phenomenon is not known. To understand this property of chemokine signaling, we examined the function of the chemokine receptors Cxcr4a, Cxcr4b, Ccr7, Ccr9 in the context of diverse processes in embryonic development in zebrafish. Our results reveal that the specific response to chemokine signaling is dictated by cell-type-specific chemokine receptor signal interpretation modules (CRIM) rather than by chemokine-receptor-specific signals. Thus, a generic signal provided by different receptors leads to discrete responses that depend on the specific identity of the cell that receives the signal. We present the implications of employing generic signals in different contexts such as gastrulation, axis specification and single-cell migration.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Cell Movement/genetics
- Embryo, Nonmammalian/cytology
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Receptors, CCR/genetics
- Receptors, CCR/metabolism
- Receptors, CCR7/genetics
- Receptors, CCR7/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Signal Transduction/genetics
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
| | - Jimann Shin
- Department of Developmental BiologyWashington University School of MedicineSt LouisMissouri
| | | | - Erez Raz
- Institute for Cell BiologyZMBEMuensterGermany
| |
Collapse
|
13
|
Stuelten CH, Parent CA, Montell DJ. Cell motility in cancer invasion and metastasis: insights from simple model organisms. Nat Rev Cancer 2018; 18:296-312. [PMID: 29546880 PMCID: PMC6790333 DOI: 10.1038/nrc.2018.15] [Citation(s) in RCA: 352] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metastasis remains the greatest challenge in the clinical management of cancer. Cell motility is a fundamental and ancient cellular behaviour that contributes to metastasis and is conserved in simple organisms. In this Review, we evaluate insights relevant to human cancer that are derived from the study of cell motility in non-mammalian model organisms. Dictyostelium discoideum, Caenorhabditis elegans, Drosophila melanogaster and Danio rerio permit direct observation of cells moving in complex native environments and lend themselves to large-scale genetic and pharmacological screening. We highlight insights derived from each of these organisms, including the detailed signalling network that governs chemotaxis towards chemokines; a novel mechanism of basement membrane invasion; the positive role of E-cadherin in collective direction-sensing; the identification and optimization of kinase inhibitors for metastatic thyroid cancer on the basis of work in flies; and the value of zebrafish for live imaging, especially of vascular remodelling and interactions between tumour cells and host tissues. While the motility of tumour cells and certain host cells promotes metastatic spread, the motility of tumour-reactive T cells likely increases their antitumour effects. Therefore, it is important to elucidate the mechanisms underlying all types of cell motility, with the ultimate goal of identifying combination therapies that will increase the motility of beneficial cells and block the spread of harmful cells.
Collapse
Affiliation(s)
- Christina H. Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Carole A. Parent
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
- Department of Pharmacology, Michigan Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- ;
| | - Denise J. Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA, USA
- ;
| |
Collapse
|
14
|
Pfeiffer J, Tarbashevich K, Bandemer J, Palm T, Raz E. Rapid progression through the cell cycle ensures efficient migration of primordial germ cells - The role of Hsp90. Dev Biol 2018; 436:84-93. [PMID: 29477339 DOI: 10.1016/j.ydbio.2018.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/30/2017] [Accepted: 02/21/2018] [Indexed: 01/21/2023]
Abstract
Zebrafish primordial germ cells (PGCs) constitute a useful in vivo model to study cell migration and to elucidate the role of specific proteins in this process. Here we report on the role of the heat shock protein Hsp90aa1.2, a protein whose RNA level is elevated in the PGCs during their migration. Reducing Hsp90aa1.2 activity slows down the progression through the cell cycle and leads to defects in the control over the MTOC number in the migrating cells. These defects result in a slower migration rate and compromise the arrival of PGCs at their target, the region where the gonad develops. Our results emphasize the importance of ensuring rapid progression through the cell cycle during single-cell migration and highlight the role of heat shock proteins in the process.
Collapse
Affiliation(s)
- Jana Pfeiffer
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Katsiaryna Tarbashevich
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Jan Bandemer
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Thomas Palm
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany.
| |
Collapse
|
15
|
Dalle Nogare D, Chitnis AB. A framework for understanding morphogenesis and migration of the zebrafish posterior Lateral Line primordium. Mech Dev 2017; 148:69-78. [PMID: 28460893 PMCID: PMC10993927 DOI: 10.1016/j.mod.2017.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 12/31/2022]
Abstract
A description of zebrafish posterior Lateral Line (pLL) primordium development at single cell resolution together with the dynamics of Wnt, FGF, Notch and chemokine signaling in this system has allowed us to develop a framework to understand the self-organization of cell fate, morphogenesis and migration during its early development. The pLL primordium migrates under the skin, from near the ear to the tip of the tail, periodically depositing neuromasts. Nascent neuromasts, or protoneuromasts, form sequentially within the migrating primordium, mature, and are deposited from its trailing end. Initially broad Wnt signaling inhibits protoneuromast formation. However, protoneuromasts form sequentially in response to FGF signaling, starting from the trailing end, in the wake of a progressively shrinking Wnt system. While proliferation adds to the number of cells, the migrating primordium progressively shrinks as its trailing cells stop moving and are deposited. As it shrinks, the length of the migrating primordium correlates with the length of the leading Wnt system. Based on these observations we show how measuring the rate at which the Wnt system shrinks, the proliferation rate, the initial size of the primordium, its speed, and a few additional parameters allows us to predict the pattern of neuromast formation and deposition by the migrating primordium in both wild-type and mutant contexts. While the mechanism that links the length of the leading Wnt system to that of the primordium remains unclear, we discuss how it might be determined by access to factors produced in the leading Wnt active zone that are required for collective migration of trailing cells. We conclude by reviewing how FGFs, produced in response to Wnt signaling in leading cells, help determine collective migration of trailing cells, while a polarized response to a self-generated chemokine gradient serves as an efficient mechanism to steer primordium migration along its relatively long journey.
Collapse
Affiliation(s)
- Damian Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA.
| |
Collapse
|
16
|
Golpour A, Siddique MAM, Siqueira-Silva DH, Pšenička M. Induced sterility in fish and its potential and challenges for aquaculture and germ cell transplantation technology: a review. Biologia (Bratisl) 2016. [DOI: 10.1515/biolog-2016-0118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
17
|
Brill JA, Yildirim S, Fabian L. Phosphoinositide signaling in sperm development. Semin Cell Dev Biol 2016; 59:2-9. [PMID: 27321976 DOI: 10.1016/j.semcdb.2016.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/15/2016] [Indexed: 01/15/2023]
Abstract
Phosphatidylinositol phosphates (PIPs)1 are membrane lipids with crucial roles during cell morphogenesis, including the establishment of cytoskeletal organization, membrane trafficking, cell polarity, cell-cycle control and signaling. Recent studies in mice (Mus musculus), fruit flies (Drosophila melanogaster) and other organisms have defined germ cell intrinsic requirements for these lipids and their regulatory enzymes in multiple aspects of sperm development. In particular, PIP levels are crucial in germline stem cell maintenance, spermatogonial proliferation and survival, spermatocyte cytokinesis, spermatid polarization, sperm tail formation, nuclear shaping, and production of mature, motile sperm. Here, we briefly review the stages of spermatogenesis and discuss the roles of PIPs and their regulatory enzymes in male germ cell development.
Collapse
Affiliation(s)
- Julie A Brill
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Sukriye Yildirim
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada.
| | - Lacramioara Fabian
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada.
| |
Collapse
|
18
|
Li H, Liang R, Lu Y, Wang M, Li Z. RTN3 Regulates the Expression Level of Chemokine Receptor CXCR4 and is Required for Migration of Primordial Germ Cells. Int J Mol Sci 2016; 17:382. [PMID: 27070582 PMCID: PMC4848882 DOI: 10.3390/ijms17040382] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/23/2016] [Accepted: 03/03/2016] [Indexed: 12/25/2022] Open
Abstract
CXCR4 is a crucial chemokine receptor that plays key roles in primordial germ cell (PGC) homing. To further characterize the CXCR4-mediated migration of PGCs, we screened CXCR4-interacting proteins using yeast two-hybrid screening. We identified reticulon3 (RTN3), a member of the reticulon family, and considered an apoptotic signal transducer, as able to interact directly with CXCR4. Furthermore, we discovered that the mRNA and protein expression levels of CXCR4 could be regulated by RTN3. We also found that RTN3 altered CXCR4 translocation and localization. Moreover, increasing the signaling of either CXCR4b or RTN3 produced similar PGC mislocalization phenotypes in zebrafish. These results suggested that RTN3 modulates PGC migration through interaction with, and regulation of, CXCR4.
Collapse
Affiliation(s)
- Haitao Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China.
| | - Rong Liang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China.
| | - Yanan Lu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China.
| | - Mengxia Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China.
| | - Zandong Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China.
| |
Collapse
|
19
|
Bar I, Cummins S, Elizur A. Transcriptome analysis reveals differentially expressed genes associated with germ cell and gonad development in the Southern bluefin tuna (Thunnus maccoyii). BMC Genomics 2016; 17:217. [PMID: 26965070 PMCID: PMC4785667 DOI: 10.1186/s12864-016-2397-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/14/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Controlling and managing the breeding of bluefin tuna (Thunnus spp.) in captivity is an imperative step towards obtaining a sustainable supply of these fish in aquaculture production systems. Germ cell transplantation (GCT) is an innovative technology for the production of inter-species surrogates, by transplanting undifferentiated germ cells derived from a donor species into larvae of a host species. The transplanted surrogates will then grow and mature to produce donor-derived seed, thus providing a simpler alternative to maintaining large-bodied broodstock such as the bluefin tuna. Implementation of GCT for new species requires the development of molecular tools to follow the fate of the transplanted germ cells. These tools are based on key reproductive and germ cell-specific genes. RNA-Sequencing (RNA-Seq) provides a rapid, cost-effective method for high throughput gene identification in non-model species. This study utilized RNA-Seq to identify key genes expressed in the gonads of Southern bluefin tuna (Thunnus maccoyii, SBT) and their specific expression patterns in male and female gonad cells. RESULTS Key genes involved in the reproductive molecular pathway and specifically, germ cell development in gonads, were identified using analysis of RNA-Seq transcriptomes of male and female SBT gonad cells. Expression profiles of transcripts from ovary and testis cells were compared, as well as testis germ cell-enriched fraction prepared with Percoll gradient, as used in GCT studies. Ovary cells demonstrated over-expression of genes related to stem cell maintenance, while in testis cells, transcripts encoding for reproduction-associated receptors, sex steroids and hormone synthesis and signaling genes were over-expressed. Within the testis cells, the Percoll-enriched fraction showed over-expression of genes that are related to post-meiosis germ cell populations. CONCLUSIONS Gonad development and germ cell related genes were identified from SBT gonads and their expression patterns in ovary and testis cells were determined. These expression patterns correlate with the reproductive developmental stage of the sampled fish. The majority of the genes described in this study were sequenced for the first time in T. maccoyii. The wealth of SBT gonadal and germ cell-related gene sequences made publicly available by this study provides an extensive resource for further GCT and reproductive molecular biology studies of this commercially valuable fish.
Collapse
Affiliation(s)
- Ido Bar
- Genecology Research Centre, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 4558 Maroochydore DC, Queensland, Australia
| | - Scott Cummins
- Genecology Research Centre, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 4558 Maroochydore DC, Queensland, Australia
| | - Abigail Elizur
- Genecology Research Centre, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 4558 Maroochydore DC, Queensland, Australia
| |
Collapse
|
20
|
Bar I, Smith A, Bubner E, Yoshizaki G, Takeuchi Y, Yazawa R, Chen BN, Cummins S, Elizur A. Assessment of yellowtail kingfish (Seriola lalandi) as a surrogate host for the production of southern bluefin tuna (Thunnus maccoyii) seed via spermatogonial germ cell transplantation. Reprod Fertil Dev 2016. [DOI: 10.1071/rd15136] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Germ cell transplantation is an innovative technology for the production of interspecies surrogates, capable of facilitating easier and more economical management of large-bodied broodstock, such as the bluefin tuna. The present study explored the suitability of yellowtail kingfish (Seriola lalandi) as a surrogate host for transplanted southern bluefin tuna (Thunnus maccoyii) spermatogonial cells to produce tuna donor-derived gametes upon sexual maturity. Germ cell populations in testes of donor T. maccoyii males were described using basic histology and the molecular markers vasa and dead-end genes. The peripheral area of the testis was found to contain the highest proportions of dead-end-expressing transplantable Type A spermatogonia. T. maccoyii Type A spermatogonia-enriched preparations were transplanted into the coelomic cavity of 6–10-day-old post-hatch S. lalandi larvae. Fluorescence microscopy and polymerase chain reaction analysis detected the presence of tuna cells in the gonads of the transplanted kingfish fingerlings at 18, 28, 39 and 75 days after transplantation, indicating that the transplanted cells migrated to the genital ridge and had colonised the developing gonad. T. maccoyii germ cell-derived DNA or RNA was not detected at later stages, suggesting that the donor cells were not maintained in the hosts’ gonads.
Collapse
|
21
|
Li H, Wu X, Hou S, Malek M, Kielkowska A, Noh E, Makondo KJ, Du Q, Wilkins JA, Johnston JB, Gibson SB, Lin F, Marshall AJ. Phosphatidylinositol-3,4-Bisphosphate and Its Binding Protein Lamellipodin Regulate Chemotaxis of Malignant B Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2015; 196:586-95. [DOI: 10.4049/jimmunol.1500630] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 11/16/2015] [Indexed: 01/14/2023]
|
22
|
Abstract
SUMMARY Stimuli that promote cell migration, such as chemokines, cytokines, and growth factors in metazoans and cyclic AMP in Dictyostelium, activate signaling pathways that control organization of the actin cytoskeleton and adhesion complexes. The Rho-family GTPases are a key convergence point of these pathways. Their effectors include actin regulators such as formins, members of the WASP/WAVE family and the Arp2/3 complex, and the myosin II motor protein. Pathways that link to the Rho GTPases include Ras GTPases, TorC2, and PI3K. Many of the molecules involved form gradients within cells, which define the front and rear of migrating cells, and are also established in related cellular behaviors such as neuronal growth cone extension and cytokinesis. The signaling molecules that regulate migration can be integrated to provide a model of network function. The network displays biochemical excitability seen as spontaneous waves of activation that propagate along the cell cortex. These events coordinate cell movement and can be biased by external cues to bring about directed migration.
Collapse
Affiliation(s)
- Peter Devreotes
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Alan Rick Horwitz
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
23
|
Chemokine-Dependent pH Elevation at the Cell Front Sustains Polarity in Directionally Migrating Zebrafish Germ Cells. Curr Biol 2015; 25:1096-103. [PMID: 25843033 DOI: 10.1016/j.cub.2015.02.071] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 01/06/2015] [Accepted: 02/25/2015] [Indexed: 11/21/2022]
Abstract
Directional cell migration requires cell polarization with respect to the distribution of the guidance cue. Cell polarization often includes asymmetric distribution of response components as well as elements of the motility machinery. Importantly, the function and regulation of most of these molecules are known to be pH dependent. Intracellular pH gradients were shown to occur in certain cells migrating in vitro, but the functional relevance of such gradients for cell migration and for the response to directional cues, particularly in the intact organism, is currently unknown. In this study, we find that primordial germ cells migrating in the context of the developing embryo respond to the graded distribution of the chemokine Cxcl12 by establishing elevated intracellular pH at the cell front. We provide insight into the mechanisms by which a polar pH distribution contributes to efficient cell migration. Specifically, we show that Carbonic Anhydrase 15b, an enzyme controlling the pH in many cell types, including metastatic cancer cells, is expressed in migrating germ cells and is crucial for establishing and maintaining an asymmetric pH distribution within them. Reducing the level of the protein and thereby erasing the pH elevation at the cell front resulted in abnormal cell migration and impaired arrival at the target. The basis for the disrupted migration is found in the stringent requirement for pH conditions in the cell for regulating contractility, for the polarization of Rac1 activity, and hence for the formation of actin-rich structures at the leading edge of the migrating cells.
Collapse
|
24
|
Bussmann J, Raz E. Chemokine-guided cell migration and motility in zebrafish development. EMBO J 2015; 34:1309-18. [PMID: 25762592 DOI: 10.15252/embj.201490105] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/04/2015] [Indexed: 12/29/2022] Open
Abstract
Chemokines are vertebrate-specific, structurally related proteins that function primarily in controlling cell movements by activating specific 7-transmembrane receptors. Chemokines play critical roles in a large number of biological processes and are also involved in a range of pathological conditions. For these reasons, chemokines are at the focus of studies in developmental biology and of clinically oriented research aimed at controlling cancer, inflammation, and immunological diseases. The small size of the zebrafish embryos, their rapid external development, and optical properties as well as the large number of eggs and the fast expansion in genetic tools available make this model an extremely useful one for studying the function of chemokines and chemokine receptors in an in vivo setting. Here, we review the findings relevant to the role that chemokines play in the context of directed single-cell migration, primarily in neutrophils and germ cells, and compare it to the collective cell migration of the zebrafish lateral line. We present the current knowledge concerning the formation of the chemokine gradient, its interpretation within the cell, and the molecular mechanisms underlying the cellular response to chemokine signals during directed migration.
Collapse
Affiliation(s)
- Jeroen Bussmann
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands Gorlaeus Laboratories, Department of Molecular Cell Biology, Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Erez Raz
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| |
Collapse
|
25
|
Liao WY, Ho CC, Hou HH, Hsu TH, Tsai MF, Chen KY, Chen HY, Lee YC, Yu CJ, Lee CH, Yang PC. Heparin co-factor II enhances cell motility and promotes metastasis in non-small cell lung cancer. J Pathol 2014; 235:50-64. [PMID: 25130770 DOI: 10.1002/path.4421] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 07/08/2014] [Accepted: 08/06/2014] [Indexed: 12/20/2022]
Abstract
Using the Serial Analysis of Gene Expression (SAGE) database from the Cancer Genome Anatomy Project, we identified heparin co-factor II (HCII), which is over-expressed in non-small cell lung cancer (NSCLC). Here, we investigated the clinical significance of HCII and provided molecular evidence to support the suggestion that HCII could enhance cancer metastasis in NSCLC. We found that high HCII expression in tumour tissue was associated with increased cancer recurrence and shorter overall survival times in 75 clinically operable NSCLC patients. High pretreatment plasma concentration of HCII was associated with reduced overall survival in 57 consecutive NSCLC patients. We over-expressed and knocked down HCII expression in lung cancer cell lines and confirmed that HCII could promote cell motility, invasion ability and filopodium dynamics in NSCLC cells in vitro and increased metastatic colonization in an in vivo mouse model. Exogenous treatment of HCII promoted cancer cell migration, and this promigratory effect of HCII was independent of thrombin. We further showed that HCII could up-regulate cancer cell migration through the activation of PI3K, which acts upstream of Rac1 and Cdc42, and this effect could be blocked by heparin. We suggest that HCII is a novel metastasis enhancer and may be used as a prognostic predictor for heparin treatment in NSCLC.
Collapse
Affiliation(s)
- Wei-Yu Liao
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Riesco MF, Valcarce DG, Alfonso J, Herráez MP, Robles V. In vitro generation of zebrafish PGC-like cells. Biol Reprod 2014; 91:114. [PMID: 25253737 DOI: 10.1095/biolreprod.114.121491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The possibility of generating primordial germ cells (PGCs) in vitro from noncommitted embryonic cells represents an extremely useful tool in current research. Primordial germ cell in vitro differentiation has been successfully reported in mammals. However, contrary to fish, PGC specification in mammals is an inductive mechanism. This study is the first to date to describe a rapid method for PGC in vitro differentiation in teleosts. Primordial germ cell-like cells were characterized by several lines of evidence, including gene expression, cell complexity, size, and image analysis for the quantification of fluorescence under vasa promoter. Moreover, differentiated cells were able to colonize the genital ridge after transplantation. Differentiation treatments increased the number of PGCs in culture, causing differentiation of cells rather than inducing their proliferation. These results open up the possibility of differentiating genetically modified embryonic cells to PGC-like cells to ensure their transmission to the progeny and could be crucial for an in-depth understanding of germline differentiation in teleosts.
Collapse
Affiliation(s)
- Marta F Riesco
- Department of Molecular Biology and Cell Biology Area, University of León, León, Spain
| | - David G Valcarce
- Department of Molecular Biology and Cell Biology Area, University of León, León, Spain
| | - Javier Alfonso
- Department of Mechanical, Computing, and Aerospace Engineering, University of León, León, Spain
| | - M Paz Herráez
- Department of Molecular Biology and Cell Biology Area, University of León, León, Spain
| | - Vanesa Robles
- Department of Molecular Biology and Cell Biology Area, University of León, León, Spain
| |
Collapse
|
27
|
Lee HC, Lin YZ, Lai YT, Huang WJ, Hu JR, Tsai JN, Tsai HJ. Glycogen synthase kinase 3 beta in somites plays a role during the angiogenesis of zebrafish embryos. FEBS J 2014; 281:4367-83. [PMID: 25056693 DOI: 10.1111/febs.12942] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 07/15/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
Glycogen synthase kinase 3 beta (Gsk3b) acts as a negative modulator in endothelial cells through the Wnt/β-catenin/PI3K/AKT/Gsk3b axis in cancer-induced angiogenesis. However, the function of Gsk3b during embryonic angiogenesis remains unclear. Here, either gsk3b knockdown by morpholino or Gsk3b loss of activity by LiCl treatment had serious phenotypic consequences, such as defects in the positioning and patterning of intersegmental blood vessels and reduction of vegfaa121 and vegfaa165 transcripts. In embryos treated with the phosphatidylinositol 3-kinase inhibitor, angiogenesis was severely inhibited, along with reduced Wnt, phosphorylated AKT and phosphorylated Gsk3b, suggesting that the remaining Gsk3b in somites could still degrade β-catenin, resulting in decreased vascular endothelial growth factor Aa(VegfAa) expression. However, in gsk3b-mRNA-overexpressed embryos, intersegmental vessels ectopically sprouted by the increase in phosphorylated-Gsk3b which prevented the degradation of β-catenin and promoted the increase in phosphorylated AKT activity, thus increasing VegfAa expression in somites. Interestingly, the Gsk3b-dependent cross-talk between PI3K/AKT and Wnt/β-catenin suggests that Wnt/β-catenin and PI3K/AKT interaction controls embryonic angiogenesis by a positive feedback loop rather than a hierarchical framework such as that found in cancer-induced angiogenesis. Thus, both active and inactive forms of Gsk3b mediate the cooperative signaling between Wnt/β-catenin and PI3K/AKT to control VegfAa expression in somites during angiogenesis in zebrafish embryos.
Collapse
Affiliation(s)
- Hung-Chieh Lee
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
28
|
Lu Y, West FD, Jordan BJ, Jordan ET, West RC, Yu P, He Y, Barrios MA, Zhu Z, Petitte JN, Beckstead RB, Stice SL. Induced Pluripotency in Chicken Embryonic Fibroblast Results in a Germ Cell Fate. Stem Cells Dev 2014; 23:1755-64. [DOI: 10.1089/scd.2014.0080] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Franklin D. West
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Brian J. Jordan
- Department of Poultry Science, University of Georgia, Athens, Georgia
| | - Erin T. Jordan
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Rachel C. West
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Ping Yu
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Ying He
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
- Guangxi Key Laboratory of Animal Vaccines and New Technology, Guangxi Veterinary Research Institute, Nanning, Guangxi, China
| | - Miguel A. Barrios
- Department of Poultry Science, University of Georgia, Athens, Georgia
| | - Ziying Zhu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - James N. Petitte
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, North Carolina
| | | | - Steven L. Stice
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| |
Collapse
|
29
|
Temporal control over the initiation of cell motility by a regulator of G-protein signaling. Proc Natl Acad Sci U S A 2014; 111:11389-94. [PMID: 25049415 DOI: 10.1073/pnas.1400043111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The control over the acquisition of cell motility is central for a variety of biological processes in development, homeostasis, and disease. An attractive in vivo model for investigating the regulation of migration initiation is that of primordial germ cells (PGCs) in zebrafish embryos. In this study, we show that, following PGC specification, the cells can polarize but do not migrate before the time chemokine-encoded directional cues are established. We found that the regulator of G-protein signaling 14a protein, whose RNA is a newly identified germ plasm component, regulates the temporal relations between the appearance of the guidance molecules and the acquisition of cellular motility by regulating E-cadherin levels.
Collapse
|
30
|
Lysko DE, Putt M, Golden JA. SDF1 reduces interneuron leading process branching through dual regulation of actin and microtubules. J Neurosci 2014; 34:4941-62. [PMID: 24695713 PMCID: PMC3972721 DOI: 10.1523/jneurosci.4351-12.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 11/21/2022] Open
Abstract
Normal cerebral cortical function requires a highly ordered balance between projection neurons and interneurons. During development these two neuronal populations migrate from distinct progenitor zones to form the cerebral cortex, with interneurons originating in the more distant ganglionic eminences. Moreover, deficits in interneurons have been linked to a variety of neurodevelopmental disorders underscoring the importance of understanding interneuron development and function. We, and others, have identified SDF1 signaling as one important modulator of interneuron migration speed and leading process branching behavior in mice, although how SDF1 signaling impacts these behaviors remains unknown. We previously found SDF1 inhibited leading process branching while increasing the rate of migration. We have now mechanistically linked SDF1 modulation of leading process branching behavior to a dual regulation of both actin and microtubule organization. We find SDF1 consolidates actin at the leading process tip by de-repressing calpain protease and increasing proteolysis of branched-actin-supporting cortactin. Additionally, SDF1 stabilizes the microtubule array in the leading process through activation of the microtubule-associated protein doublecortin (DCX). DCX stabilizes the microtubule array by bundling microtubules within the leading process, reducing branching. These data provide mechanistic insight into the regulation of interneuron leading process dynamics during neuronal migration in mice and provides insight into how cortactin and DCX, a known human neuronal migration disorder gene, participate in this process.
Collapse
Affiliation(s)
- Daniel E. Lysko
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Mary Putt
- Department of Biostatistics and Epidemiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, and
| | - Jeffrey A. Golden
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| |
Collapse
|
31
|
Wei KH, Liu IH. Heparan sulfate glycosaminoglycans modulate migration and survival in zebrafish primordial germ cells. Theriogenology 2014; 81:1275-85.e1-2. [PMID: 24629592 DOI: 10.1016/j.theriogenology.2014.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 01/09/2014] [Accepted: 02/06/2014] [Indexed: 11/17/2022]
Abstract
Early in embryonic development, primordial germ cells (PGCs) are specified and migrate from the site of their origin to where the gonad develops, following a specific route. Heparan sulfate glycosaminoglycans (HS-GAGs) are ubiquitous in extracellular matrix and the cell surface and have long been speculated to play a role during the migration of PGCs. In line with this speculation, whole-mount immunohistochemistry revealed the existence of HS-GAGs in the vicinity of migrating PGCs in early zebrafish embryos. To examine the roles of HS-GAGs during PGC migration, zebrafish heparanase 1 (hpse1), which degrades HS-GAGs, was cloned and overexpressed specifically in PGCs. The guidance signal for the migration of PGCs was disrupted with the overexpression of hpse1, as cluster formation and marginal localization at the blastoderm were significantly perturbed at 6 hours postfertilization. Furthermore, the number of PGCs was significantly decreased with the lack of vicinal HS-GAGs, as observed in the whole-mount in situ hybridization and quantitative PCR of the PGC marker gene vasa. Terminal deoxynucleotidyl transferase dUTP nick-end labeling indicated significantly increased apoptosis in PGCs overexpressing hpse1, suggesting that HS-GAGs contribute to the maintenance of PGC survival. In conclusion, HS-GAGs play multifaceted roles in PGCs during migration and are required both for guidance signals and multiplication of PGCs.
Collapse
Affiliation(s)
- Ke-Hsuan Wei
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
32
|
Glover JD, Taylor L, Sherman A, Zeiger-Poli C, Sang HM, McGrew MJ. A novel piggyBac transposon inducible expression system identifies a role for AKT signalling in primordial germ cell migration. PLoS One 2013; 8:e77222. [PMID: 24223709 PMCID: PMC3817190 DOI: 10.1371/journal.pone.0077222] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/09/2013] [Indexed: 01/15/2023] Open
Abstract
In this work, we describe a single piggyBac transposon system containing both a tet-activator and a doxycycline-inducible expression cassette. We demonstrate that a gene product can be conditionally expressed from the integrated transposon and a second gene can be simultaneously targeted by a short hairpin RNA contained within the transposon, both in vivo and in mammalian and avian cell lines. We applied this system to stably modify chicken primordial germ cell (PGC) lines in vitro and induce a reporter gene at specific developmental stages after injection of the transposon-modified germ cells into chicken embryos. We used this vector to express a constitutively-active AKT molecule during PGC migration to the forming gonad. We found that PGC migration was retarded and cells could not colonise the forming gonad. Correct levels of AKT activation are thus essential for germ cell migration during early embryonic development.
Collapse
Affiliation(s)
- James D Glover
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | | | | | | | | | | |
Collapse
|
33
|
Wong TT, Collodi P. Effects of specific and prolonged expression of zebrafish growth factors, Fgf2 and Lif in primordial germ cells in vivo. Biochem Biophys Res Commun 2012. [PMID: 23178298 DOI: 10.1016/j.bbrc.2012.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Primordial germ cells (PGCs), specified early in development, proliferate and migrate to the developing gonad before sexual differentiation occurs in the embryo and eventually give rise to spermatogonia or oogonia. In this study, we discovered that nanos3 3'UTR, a common method used to label PGCs, not only directed PGC-specific expression of DsRed but also prolonged this expression up to 26 days post fertilization (dpf) when DsRed-nanos3 3'UTR hybrid mRNAs were introduced into 1- to 2-cell-stage embryos. As such, we employed this knowledge to express zebrafish leukemia inhibitory factor (Lif), basic fibroblast growth factor (Fgf2) and bone morphogenetic protein 4 (Bmp4) in the PGCs and evaluate their effects on PGC development in vivo for over a period of 3 weeks. The results show that expression of Fgf2 significantly increased PGC number at 14- and 21-dpf while Bmp4 resulted in severe ventralization and death of the embryos by 3 days. Expression of Lif resulted in a significant disruption of PGC migration. Mopholino knockdown experiments indicated that Lif illicited its effect on PGC migration through Lif receptor a (Lifra) but not Lifrb. The general approach described in this study could be used to achieve prolonged PGC-specific expression of other proteins to investigate their roles in germ cell and gonad development. The results also indicate that zebrafish PGCs have a mechanism to stabilize and prolong the expression of mRNA that carries nanos3 3'UTR. Understanding this mechanism may make it possible to achieve prolonged RNA expression in other cell types.
Collapse
Affiliation(s)
- Ten-Tsao Wong
- Department of Animal Sciences, Purdue University, 901 W. State Street, West Lafayette, IN 47907, USA.
| | | |
Collapse
|
34
|
Stulberg MJ, Lin A, Zhao H, Holley SA. Crosstalk between Fgf and Wnt signaling in the zebrafish tailbud. Dev Biol 2012; 369:298-307. [PMID: 22796649 PMCID: PMC3423502 DOI: 10.1016/j.ydbio.2012.07.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 06/26/2012] [Accepted: 07/02/2012] [Indexed: 01/09/2023]
Abstract
Fibroblast growth factor (Fgf) and Wnt signaling are necessary for the intertwined processes of tail elongation, mesodermal development and somitogenesis. Here, we use pharmacological modifiers and time-resolved quantitative analysis of both nascent transcription and protein phosphorylation in the tailbud, to distinguish early effects of signal perturbation from later consequences related to cell fate changes. We demonstrate that Fgf activity elevates Wnt signaling by inhibiting transcription of the Wnt antagonists dkk1 and notum1a. PI3 kinase signaling also increases Wnt signaling via phosphorylation of Gsk3β. Conversely, Wnt can increase signaling within the Mapk branch of the Fgf pathway as Gsk3β phosphorylation elevates phosphorylation levels of Erk. Despite the reciprocal positive regulation between Fgf and Wnt, the two pathways generally have opposing effects on the transcription of co-regulated genes. This opposing regulation of target genes may represent a rudimentary relationship that manifests as out-of-phase oscillation of Fgf and Wnt target genes in the mouse and chick tailbud. In summary, these data suggest that Fgf and Wnt signaling are tightly integrated to maintain proportional levels of activity in the zebrafish tailbud, and this balance is important for axis elongation, cell fate specification and somitogenesis.
Collapse
Affiliation(s)
- Michael J. Stulberg
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Aiping Lin
- Keck Biostatistics Resource, Yale University, New Haven, CT 06511, USA
| | - Hongyu Zhao
- Keck Biostatistics Resource, Yale University, New Haven, CT 06511, USA
- Department of Epidemiology and Public Health, Yale University, New Haven, CT 06511, USA
| | - Scott A. Holley
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
35
|
Petrie RJ, Gavara N, Chadwick RS, Yamada KM. Nonpolarized signaling reveals two distinct modes of 3D cell migration. ACTA ACUST UNITED AC 2012; 197:439-55. [PMID: 22547408 PMCID: PMC3341168 DOI: 10.1083/jcb.201201124] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The elastic behavior of the 3D extracellular matrix determines the relative polarization of intracellular signaling and whether cells migrate using lamellipodia or lobopodia. We search in this paper for context-specific modes of three-dimensional (3D) cell migration using imaging for phosphatidylinositol (3,4,5)-trisphosphate (PIP3) and active Rac1 and Cdc42 in primary fibroblasts migrating within different 3D environments. In 3D collagen, PIP3 and active Rac1 and Cdc42 were targeted to the leading edge, consistent with lamellipodia-based migration. In contrast, elongated cells migrating inside dermal explants and the cell-derived matrix (CDM) formed blunt, cylindrical protrusions, termed lobopodia, and Rac1, Cdc42, and PIP3 signaling was nonpolarized. Reducing RhoA, Rho-associated protein kinase (ROCK), or myosin II activity switched the cells to lamellipodia-based 3D migration. These modes of 3D migration were regulated by matrix physical properties. Specifically, experimentally modifying the elasticity of the CDM or collagen gels established that nonlinear elasticity supported lamellipodia-based migration, whereas linear elasticity switched cells to lobopodia-based migration. Thus, the relative polarization of intracellular signaling identifies two distinct modes of 3D cell migration governed intrinsically by RhoA, ROCK, and myosin II and extrinsically by the elastic behavior of the 3D extracellular matrix.
Collapse
Affiliation(s)
- Ryan J Petrie
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
36
|
Xu H, Kardash E, Chen S, Raz E, Lin F. Gβγ signaling controls the polarization of zebrafish primordial germ cells by regulating Rac activity. Development 2011; 139:57-62. [PMID: 22096073 DOI: 10.1242/dev.073924] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
During development, primordial germ cells (PGCs) migrate from the sites of their specification towards the region in which the future gonad develops. This cell migration requires polarization of PGCs and their responsiveness to external guidance cues. In zebrafish, the directed migration and polarization of PGCs are regulated independently, by the chemokine Cxcl12a and the Rho GTPase Rac1, respectively. However, the upstream signals controlling Rac activity in this context have not yet been identified. By investigating the role of G proteins in PGC migration, we found that signaling mediated by G protein subunits Gβγ is required to regulate cell polarization. PGCs that are defective for Gβγ signaling failed to polarize, and developed multiple protrusions in random locations, resembling the defects observed in PGCs with decreased Rac activity. These defects render PGCs incapable of migrating actively and responding to directional cues. FRET-based assays showed that PGCs require Gβγ signaling for polarized Rac activation and actin organization at the leading front, as well as for maintaining overall Rac levels in these cells. Conversely, overexpression of Gβγ in PGCs increases Rac activity. Our results indicate that during PGC migration in vivo, Gβγ signaling regulates Rac activity to control cell polarity, which is required for the responsiveness to chemokine signaling.
Collapse
Affiliation(s)
- Hui Xu
- Department of Anatomy and Cell Biology, University of Iowa, 1-400 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242-1109, USA
| | | | | | | | | |
Collapse
|
37
|
Tarbashevich K, Dzementsei A, Pieler T. A novel function for KIF13B in germ cell migration. Dev Biol 2011; 349:169-78. [DOI: 10.1016/j.ydbio.2010.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Revised: 09/19/2010] [Accepted: 10/14/2010] [Indexed: 10/18/2022]
|
38
|
Tarbashevich K, Raz E. The nuts and bolts of germ-cell migration. Curr Opin Cell Biol 2010; 22:715-21. [DOI: 10.1016/j.ceb.2010.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 08/20/2010] [Accepted: 09/16/2010] [Indexed: 12/28/2022]
|
39
|
An evolutionarily conserved arginine is essential for Tre1 G protein-coupled receptor function during germ cell migration in Drosophila melanogaster. PLoS One 2010; 5:e11839. [PMID: 20676220 PMCID: PMC2911388 DOI: 10.1371/journal.pone.0011839] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 06/29/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND G protein-coupled receptors (GPCRs) play central roles in mediating cellular responses to environmental signals leading to changes in cell physiology and behaviors, including cell migration. Numerous clinical pathologies including metastasis, an invasive form of cell migration, have been linked to abnormal GPCR signaling. While the structures of some GPCRs have been defined, the in vivo roles of conserved amino acid residues and their relationships to receptor function are not fully understood. Trapped in endoderm 1 (Tre1) is an orphan receptor of the rhodopsin class that is necessary for primordial germ cell migration in Drosophila melanogaster embryos. In this study, we employ molecular genetic approaches to identify residues in Tre1 that are critical to its functions in germ cell migration. METHODOLOGY/PRINCIPAL FINDINGS First, we show that the previously reported scattershot mutation is an allele of tre1. The scattershot allele results in an in-frame deletion of 8 amino acids at the junction of the third transmembrane domain and the second intracellular loop of Tre1 that dramatically impairs the function of this GPCR in germ cell migration. To further refine the molecular basis for this phenotype, we assayed the effects of single amino acid substitutions in transgenic animals and determined that the arginine within the evolutionarily conserved E/N/DRY motif is critical for receptor function in mediating germ cell migration within an intact developing embryo. CONCLUSIONS/SIGNIFICANCE These structure-function studies of GPCR signaling in native contexts will inform future studies into the basic biology of this large and clinically important family of receptors.
Collapse
|
40
|
Swaney KF, Huang CH, Devreotes PN. Eukaryotic chemotaxis: a network of signaling pathways controls motility, directional sensing, and polarity. Annu Rev Biophys 2010; 39:265-89. [PMID: 20192768 DOI: 10.1146/annurev.biophys.093008.131228] [Citation(s) in RCA: 370] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chemotaxis, the directed migration of cells in chemical gradients, is a vital process in normal physiology and in the pathogenesis of many diseases. Chemotactic cells display motility, directional sensing, and polarity. Motility refers to the random extension of pseudopodia, which may be driven by spontaneous actin waves that propagate through the cytoskeleton. Directional sensing is mediated by a system that detects temporal and spatial stimuli and biases motility toward the gradient. Polarity gives cells morphologically and functionally distinct leading and lagging edges by relocating proteins or their activities selectively to the poles. By exploiting the genetic advantages of Dictyostelium, investigators are working out the complex network of interactions between the proteins that have been implicated in the chemotactic processes of motility, directional sensing, and polarity.
Collapse
Affiliation(s)
- Kristen F Swaney
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
41
|
Xu H, Li M, Gui J, Hong Y. Fish germ cells. SCIENCE CHINA-LIFE SCIENCES 2010; 53:435-46. [PMID: 20596909 DOI: 10.1007/s11427-010-0058-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 12/02/2009] [Indexed: 01/15/2023]
Abstract
Fish, like many other animals, have two major cell lineages, namely the germline and soma. The germ-soma separation is one of the earliest events of embryonic development. Germ cells can be specifically labeled and isolated for culture and transplantation, providing tools for reproduction of endangered species in close relatives, such as surrogate production of trout in salmon. Haploid cell cultures, such as medaka haploid embryonic stem cells have recently been obtained, which are capable of mimicking sperm to produce fertile offspring, upon nuclear being directly transferred into normal eggs. Such fish originated from a mosaic oocyte that had a haploid meiotic nucleus and a transplanted haploid mitotic cell culture nucleus. The first semi-cloned fish is Holly. Here we review the current status and future directions of understanding and manipulating fish germ cells in basic research and reproductive technology.
Collapse
Affiliation(s)
- HongYan Xu
- Department of Biological Sciences, National University of Singapore, Singapore 119260, Singapore
| | | | | | | |
Collapse
|
42
|
Tumor angiogenesis: insights and innovations. JOURNAL OF ONCOLOGY 2010; 2010:132641. [PMID: 20445741 PMCID: PMC2860112 DOI: 10.1155/2010/132641] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Revised: 02/12/2010] [Accepted: 02/12/2010] [Indexed: 12/21/2022]
Abstract
Angiogenesis is a vital process resulting in the formation of new blood vessels. It is normally a highly regulated process that occurs during human development, reproduction, and wound repair. However, angiogenesis can also become a fundamental pathogenic process found in cancer and several other diseases. To date, the inhibition of angiogenesis has been researched at both the bench and the bedside. While several studies have found moderate improvements when treating with angiogenesis inhibitors, greater success is being seen when the inhibition of angiogenesis is combined with other traditional forms of available therapy. This review summarizes several important angiogenic factors, examines new research and ongoing clinical trials for such factors, and attempts to explain how this new knowledge may be applied in the fight against cancer and other angiogenic-related diseases.
Collapse
|
43
|
Wessels D, Kuhl S, Soll DR. 2D and 3D quantitative analysis of cell motility and cytoskeletal dynamics. Methods Mol Biol 2010; 586:315-35. [PMID: 19768439 DOI: 10.1007/978-1-60761-376-3_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
2D- and 3D-Dynamic Image Analysis Systems (2D- and 3D-DIAS) for quantitative analysis of cell motility and chemotaxis are described. Particular attention is given to protocols that have proven useful in the quantitation of cell shape changes and pseudopod dynamics during basic cell motility (i.e. crawling in the absence of a chemotactic or other type of extracellular signal) and directed motion. In addition, methods provided, highlight the applicability of this approach to the accurate phenotypic characterizations of cytoskeletal mutations in Dictyostelium discoideum, cytoskeletal alterations in metastatic cells, and cytoskeletal defects in chemotactically defective polymorphonuclear neutrophils.
Collapse
Affiliation(s)
- Deborah Wessels
- Keck Dynamic Image Analysis Facility, Department of Biological Sciences, University of Iowa, Iowa City, IA, USA
| | | | | |
Collapse
|
44
|
Yoo SK, Deng Q, Cavnar PJ, Wu YI, Hahn KM, Huttenlocher A. Differential regulation of protrusion and polarity by PI3K during neutrophil motility in live zebrafish. Dev Cell 2010; 18:226-36. [PMID: 20159593 DOI: 10.1016/j.devcel.2009.11.015] [Citation(s) in RCA: 294] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2009] [Revised: 10/29/2009] [Accepted: 11/23/2009] [Indexed: 11/26/2022]
Abstract
Cell polarity is crucial for directed migration. Here we show that phosphoinositide 3-kinase (PI(3)K) mediates neutrophil migration in vivo by differentially regulating cell protrusion and polarity. The dynamics of PI(3)K products PI(3,4,5)P(3)-PI(3,4)P(2) during neutrophil migration were visualized in living zebrafish, revealing that PI(3)K activation at the leading edge is critical for neutrophil motility in intact tissues. A genetically encoded photoactivatable Rac was used to demonstrate that localized activation of Rac is sufficient to direct migration with precise temporal and spatial control in vivo. Similar stimulation of PI(3)K-inhibited cells did not direct migration. Localized Rac activation rescued membrane protrusion but not anteroposterior polarization of F-actin dynamics of PI(3)K-inhibited cells. Uncoupling Rac-mediated protrusion and polarization suggests a paradigm of two-tiered PI(3)K-mediated regulation of cell motility. This work provides new insight into how cell signaling at the front and back of the cell is coordinated during polarized cell migration in intact tissues within a multicellular organism.
Collapse
Affiliation(s)
- Sa Kan Yoo
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
45
|
Neumann JC, Dovey JS, Chandler GL, Carbajal L, Amatruda JF. Identification of a heritable model of testicular germ cell tumor in the zebrafish. Zebrafish 2010; 6:319-27. [PMID: 20047465 DOI: 10.1089/zeb.2009.0613] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Germ cell tumors (GCTs) affect infants, children, and adults and are the most common cancer type in young men. Progress in understanding the molecular basis of GCTs has been hampered by a lack of suitable animal models. Here we report the identification of a zebrafish model of highly penetrant, heritable testicular GCT isolated as part of a forward genetic screen for cancer susceptibility genes. The mutant line develops spontaneous testicular tumors at a median age of 7 months, and pedigree analysis indicates dominant inheritance of the GCT susceptibility trait. The zebrafish model exhibits disruption of testicular tissue architecture and the accumulation of primitive, spermatogonial-like cells with loss of spermatocytic differentiation. Radiation treatment leads to apoptosis of the tumor cells and tumor regression. The GCT-susceptible line can serve as a model for understanding the mechanisms regulating germ cells in normal development and disease and as a platform investigating new therapeutic approaches for GCTs.
Collapse
Affiliation(s)
- Joanie C Neumann
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | |
Collapse
|
46
|
Mulligan T, Blaser H, Raz E, Farber SA. Prenylation-deficient G protein gamma subunits disrupt GPCR signaling in the zebrafish. Cell Signal 2010; 22:221-33. [PMID: 19786091 PMCID: PMC2788088 DOI: 10.1016/j.cellsig.2009.09.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 09/02/2009] [Accepted: 09/14/2009] [Indexed: 01/01/2023]
Abstract
Prenylation of G protein gamma (gamma) subunits is necessary for the membrane localization of heterotrimeric G proteins and for functional heterotrimeric G protein coupled receptor (GPCR) signaling. To evaluate GPCR signaling pathways during development, we injected zebrafish embryos with mRNAs encoding Ggamma subunits mutated so that they can no longer be prenylated. Low-level expression of these prenylation-deficient Ggamma subunits driven either ubiquitously or specifically in the primordial germ cells (PGCs) disrupts GPCR signaling and manifests as a PGC migration defect. This disruption results in a reduction of calcium accumulation in the protrusions of migrating PGCs and a failure of PGCs to directionally migrate. When co-expressed with a prenylation-deficient Ggamma, 8 of the 17 wildtype Ggamma isoforms individually confer the ability to restore calcium accumulation and directional migration. These results suggest that while the Ggamma subunits possess the ability to interact with G Beta (beta) proteins, only a subset of wildtype Ggamma proteins are stable within PGCs and can interact with key signaling components necessary for PGC migration. This in vivo study highlights the functional redundancy of these signaling components and demonstrates that prenylation-deficient Ggamma subunits are an effective tool to investigate the roles of GPCR signaling events during vertebrate development.
Collapse
Affiliation(s)
- Timothy Mulligan
- Carnegie Institution, Department of Embryology, Johns Hopkins University, Baltimore, MD 21218, United States
| | | | | | | |
Collapse
|
47
|
Richardson BE, Lehmann R. Mechanisms guiding primordial germ cell migration: strategies from different organisms. Nat Rev Mol Cell Biol 2010; 11:37-49. [PMID: 20027186 PMCID: PMC4521894 DOI: 10.1038/nrm2815] [Citation(s) in RCA: 350] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The regulated migration of cells is essential for development and tissue homeostasis, and aberrant cell migration can lead to an impaired immune response and the progression of cancer. Primordial germ cells (PGCs), precursors to sperm and eggs, have to migrate across the embryo to reach somatic gonadal precursors, where they carry out their function. Studies of model organisms have revealed that, despite important differences, several features of PGC migration are conserved. PGCs require an intrinsic motility programme and external guidance cues to survive and successfully migrate. Proper guidance involves both attractive and repulsive cues and is mediated by protein and lipid signalling.
Collapse
Affiliation(s)
- Brian E Richardson
- Howard Hughes Medical Institute, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York University, New York, 10016, USA
| | | |
Collapse
|
48
|
Clelland E, Peng C. Endocrine/paracrine control of zebrafish ovarian development. Mol Cell Endocrinol 2009; 312:42-52. [PMID: 19406202 DOI: 10.1016/j.mce.2009.04.009] [Citation(s) in RCA: 198] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 04/16/2009] [Accepted: 04/17/2009] [Indexed: 12/11/2022]
Abstract
Ovarian differentiation and the processes of follicle development, oocyte maturation and ovulation are complex events, requiring the coordinated action of regulatory molecules. In zebrafish, ovarian development is initiated at 10 days after hatching and fish become sexually mature at 3 months. Adult zebrafish have asynchronous ovaries, which contain follicles of all stages of development. Eggs are spawned daily under proper environmental conditions in a population of zebrafish, with individual females spawning irregularly every 4-7 days in mixed sex conditions. Maximal embryo viability is achieved when sexually isolated females are bred in 10-day intervals [Niimi, A.J., LaHam, Q.N., 1974. Influence of breeding time interval on egg number, mortality, and hatching of the zebra fish Brachydanio verio. Can. J. Zool. 52, 515-517]. Similar to other vertebrates, hormones from the hypothalamus-pituitary-gonadal axis play important roles in regulating follicle development. Follicle stimulating hormone (FSH) stimulates estradiol production, which in turn, promotes viteollogenesis. Luteinizing hormone (LH) stimulates the production of 17,20beta-dihydroxy-4-pregnen-3-one (17,20betaP) or maturation inducing hormone (MIH) which acts through membrane progestin receptors to activate maturation promoting factor, leading to oocyte maturation. Recent studies in zebrafish have also provided novel insights into the functions of ovary-derived growth factors in follicle development and oocyte maturation. The present review summarizes the current knowledge on how endocrine and paracrine factors regulate ovarian development in zebrafish. Special emphasis is placed on how follicle development and oocyte maturation in adult females is regulated by gonadotropins, ovarian steroids and growth factors produced by the ovary.
Collapse
Affiliation(s)
- Eric Clelland
- Department of Biology, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada
| | | |
Collapse
|
49
|
Abstract
The movements of Dictyostelium discoideum amoebae translocating on a glass surface in the absence of chemoattractant have been reconstructed at 5-second intervals and motion analyzed by employing 3D-DIAS software. A morphometric analysis of pseudopods, the main cell body, and the uropod provides a comprehensive description of the basic motile behavior of a cell in four dimensions (4D), resulting in a list of 18 characteristics. A similar analysis of the myosin II phosphorylation mutant 3XASP reveals a role for the cortical localization of myosin II in the suppression of lateral pseudopods, formation of the uropod, cytoplasmic distribution of cytoplasm in the main cell body, and efficient motility. The results of the morphometric analysis suggest that pseudopods, the main cell body, and the uropod represent three motility compartments that are coordinated for efficient translocation. It provides a contextual framework for interpreting the effects of mutations, inhibitors, and chemoattractants on the basic motile behavior of D. discoideum. The generality of the characteristics of the basic motile behavior of D. discoideum must now be tested by similar 4D analyses of the motility of amoeboid cells of higher eukaryotic cells, in particular human polymorphonuclear leukocytes.
Collapse
|
50
|
Olesnicky Killian EC, Birkholz DA, Artinger KB. A role for chemokine signaling in neural crest cell migration and craniofacial development. Dev Biol 2009; 333:161-72. [PMID: 19576198 DOI: 10.1016/j.ydbio.2009.06.031] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 06/22/2009] [Accepted: 06/23/2009] [Indexed: 12/13/2022]
Abstract
Neural crest cells (NCCs) are a unique population of multipotent cells that migrate along defined pathways throughout the embryo and give rise to many diverse cell types including pigment cells, craniofacial cartilage and the peripheral nervous system (PNS). Aberrant migration of NCCs results in a wide variety of congenital birth defects including craniofacial abnormalities. The chemokine Sdf1 and its receptors, Cxcr4 and Cxcr7, have been identified as key components in the regulation of cell migration in a variety of tissues. Here we describe a novel role for the zebrafish chemokine receptor Cxcr4a in the development and migration of cranial NCCs (CNCCs). We find that loss of Cxcr4a, but not Cxcr7b, results in aberrant CNCC migration defects in the neurocranium, as well as cranial ganglia dysmorphogenesis. Moreover, overexpression of either Sdf1b or Cxcr4a causes aberrant CNCC migration and results in ectopic craniofacial cartilages. We propose a model in which Sdf1b signaling from the pharyngeal arch endoderm and optic stalk to Cxcr4a expressing CNCCs is important for both the proper condensation of the CNCCs into pharyngeal arches and the subsequent patterning and morphogenesis of the neural crest derived tissues.
Collapse
Affiliation(s)
- Eugenia C Olesnicky Killian
- Department of Craniofacial Biology, University of Colorado Denver School of Dental Medicine, Aurora, CO 80045, USA
| | | | | |
Collapse
|