1
|
Wang N, Lv L, Huang X, Shi M, Dai Y, Wei Y, Xu B, Fu C, Huang H, Shi H, Liu Y, Hu X, Qin D. Gene editing in monogenic autism spectrum disorder: animal models and gene therapies. Front Mol Neurosci 2022; 15:1043018. [PMID: 36590912 PMCID: PMC9794862 DOI: 10.3389/fnmol.2022.1043018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a lifelong neurodevelopmental disease, and its diagnosis is dependent on behavioral manifestation, such as impaired reciprocal social interactions, stereotyped repetitive behaviors, as well as restricted interests. However, ASD etiology has eluded researchers to date. In the past decades, based on strong genetic evidence including mutations in a single gene, gene editing technology has become an essential tool for exploring the pathogenetic mechanisms of ASD via constructing genetically modified animal models which validates the casual relationship between genetic risk factors and the development of ASD, thus contributing to developing ideal candidates for gene therapies. The present review discusses the progress in gene editing techniques and genetic research, animal models established by gene editing, as well as gene therapies in ASD. Future research should focus on improving the validity of animal models, and reliable DNA diagnostics and accurate prediction of the functional effects of the mutation will likely be equally crucial for the safe application of gene therapies.
Collapse
Affiliation(s)
- Na Wang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Longbao Lv
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaoyi Huang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Mingqin Shi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Youwu Dai
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yuanyuan Wei
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Bonan Xu
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Chenyang Fu
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Haoyu Huang
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, Yunnan, China
| | - Hongling Shi
- Department of Rehabilitation Medicine, The Third People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yun Liu
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, Yunnan, China
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
2
|
Rho GTPases Signaling in Zebrafish Development and Disease. Cells 2020; 9:cells9122634. [PMID: 33302361 PMCID: PMC7762611 DOI: 10.3390/cells9122634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023] Open
Abstract
Cells encounter countless external cues and the specificity of their responses is translated through a myriad of tightly regulated intracellular signals. For this, Rho GTPases play a central role and transduce signals that contribute to fundamental cell dynamic and survival events. Here, we review our knowledge on how zebrafish helped us understand the role of some of these proteins in a multitude of in vivo cellular behaviors. Zebrafish studies offer a unique opportunity to explore the role and more specifically the spatial and temporal dynamic of Rho GTPases activities within a complex environment at a level of details unachievable in any other vertebrate organism.
Collapse
|
3
|
Hung IC, Chen TM, Lin JP, Tai YL, Shen TL, Lee SJ. Wnt5b integrates Fak1a to mediate gastrulation cell movements via Rac1 and Cdc42. Open Biol 2020; 10:190273. [PMID: 32097584 PMCID: PMC7058935 DOI: 10.1098/rsob.190273] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Focal adhesion kinase (FAK) mediates vital cellular pathways during development. Despite its necessity, how FAK regulates and integrates with other signals during early embryogenesis remains poorly understood. We found that the loss of Fak1a impaired epiboly, convergent extension and hypoblast cell migration in zebrafish embryos. We also observed a clear disturbance in cortical actin at the blastoderm margin and distribution of yolk syncytial nuclei. In addition, we investigated a possible link between Fak1a and a well-known gastrulation regulator, Wnt5b, and revealed that the overexpression of fak1a or wnt5b could cross-rescue convergence defects induced by a wnt5b or fak1a antisense morpholino (MO), respectively. Wnt5b and Fak1a were shown to converge in regulating Rac1 and Cdc42, which could synergistically rescue wnt5b and fak1a morphant phenotypes. Furthermore, we generated several alleles of fak1a mutants using CRISPR/Cas9, but those mutants only revealed mild gastrulation defects. However, injection of a subthreshold level of the wnt5b MO induced severe gastrulation defects in fak1a mutants, which suggested that the upregulated expression of wnt5b might complement the loss of Fak1a. Collectively, we demonstrated that a functional interaction between Wnt and FAK signalling mediates gastrulation cell movements via the possible regulation of Rac1 and Cdc42 and subsequent actin dynamics.
Collapse
Affiliation(s)
- I-Chen Hung
- Department of Life Science, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan
| | - Tsung-Ming Chen
- Department of Life Science, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Department and Graduate Institute of Aquaculture, National Kaohsiung Marine University, Kaohsiung, Taiwan
| | - Jing-Ping Lin
- Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan
| | - Yu-Ling Tai
- Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shyh-Jye Lee
- Department of Life Science, National Taiwan University, No. 1, Roosevelt Road, Section 4, Taipei 10617, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Center for Biotechnology, National Taiwan University, Taipei, Taiwan.,Center for Systems Biology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
4
|
IRE1α governs cytoskeleton remodelling and cell migration
through a direct interaction with filamin A. Nat Cell Biol 2018; 20:942-953. [DOI: 10.1038/s41556-018-0141-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/13/2018] [Indexed: 02/07/2023]
|
5
|
Sepúlveda-Ramírez SP, Toledo-Jacobo L, Henson JH, Shuster CB. Cdc42 controls primary mesenchyme cell morphogenesis in the sea urchin embryo. Dev Biol 2018; 437:140-151. [PMID: 29555242 PMCID: PMC5973877 DOI: 10.1016/j.ydbio.2018.03.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/05/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022]
Abstract
In the sea urchin embryo, gastrulation is characterized by the ingression and directed cell migration of primary mesenchyme cells (PMCs), as well as the primary invagination and convergent extension of the endomesoderm. Like all cell shape changes, individual and collective cell motility is orchestrated by Rho family GTPases and their modulation of the actomyosin cytoskeleton. And while endomesoderm specification has been intensively studied in echinoids, much less is known about the proximate regulators driving cell motility. Toward these ends, we employed anti-sense morpholinos, mutant alleles and pharmacological inhibitors to assess the role of Cdc42 during sea urchin gastrulation. While inhibition of Cdc42 expression or activity had only mild effects on PMC ingression, PMC migration, alignment and skeletogenesis were disrupted in the absence of Cdc42, as well as elongation of the archenteron. PMC migration and patterning of the larval skeleton relies on the extension of filopodia, and Cdc42 was required for filopodia in vivo as well as in cultured PMCs. Lastly, filopodial extension required both Arp2/3 and formin actin-nucleating factors, supporting models of filopodial nucleation observed in other systems. Together, these results suggest that Cdc42 plays essential roles during PMC cell motility and organogenesis.
Collapse
Affiliation(s)
- Silvia P Sepúlveda-Ramírez
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, United States; University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States
| | - Leslie Toledo-Jacobo
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, United States; University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States
| | - John H Henson
- University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States; Department of Biology, Dickinson College, Carlisle, PA 17013, United States
| | - Charles B Shuster
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, United States; University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States.
| |
Collapse
|
6
|
Brolih S, Parks SK, Vial V, Durivault J, Mostosi L, Pouysségur J, Pagès G, Picco V. AKT1 restricts the invasive capacity of head and neck carcinoma cells harboring a constitutively active PI3 kinase activity. BMC Cancer 2018; 18:249. [PMID: 29506489 PMCID: PMC5836445 DOI: 10.1186/s12885-018-4169-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 02/26/2018] [Indexed: 02/08/2023] Open
Abstract
Background In mammals, the AKT/PKB protein kinase family comprises three members (AKT1–3). PI3-Kinase (PI3K), a key oncogene involved in a wide variety of cancers, drives AKT activity. Constitutive activation of the PI3K/AKT pathway has been associated with tumorigenic properties including uncontrolled cell proliferation and survival, angiogenesis, promotion of cellular motility, invasiveness and metastasis. However, AKT1 activity has also been recently shown to repress the invasive properties of breast cancer cells in specific contexts. Methods This study used both pharmacological and shRNA approaches to inhibit AKT function, microscopy to characterize the cellular morphology, 3D spheroid models to assess migratory and invasive cellular capacities and a phenotypic screening approach based on electrical properties of the cells. Results Here we demonstrate that the alternative action of AKT1 on invasive properties of breast cancers can be extended to head and neck carcinomas, which exhibit constitutive activation of the PI3K/AKT pathway. Indeed, inhibition of AKT1 function by shRNA or a specific pharmacological inhibitor resulted in cellular spreading and an invasive phenotype. A phenotypic screening approach based on cellular electrical properties corroborated microscopic observations and provides a foundation for future high-throughput screening studies. This technique further showed that the inhibition of AKT1 signaling is phenocopied by blocking the mTORC1 pathway with rapamycin. Conclusion Our study suggests that the repressive action of PI3K/AKT1 on cellular invasive properties may be a mechanism common to several cancers. Current and future studies involving AKT inhibitors must therefore consider this property to prevent metastases and consequently to improve survival. Electronic supplementary material The online version of this article (10.1186/s12885-018-4169-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanja Brolih
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco
| | - Scott K Parks
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco
| | - Valérie Vial
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco
| | - Jérôme Durivault
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco
| | - Livio Mostosi
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco
| | - Jacques Pouysségur
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco
| | - Gilles Pagès
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco.,UCA, Université Côte d'Azur, Nice-Sophia-Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS-UMR 7284-Inserm U1081, Nice, France
| | - Vincent Picco
- Centre Scientifique de Monaco, Department of Medical Biology, 8 Quai Antoine Ier, Monaco, Principality of Monaco.
| |
Collapse
|
7
|
Hanovice NJ, McMains E, Gross JM. A GAL4-inducible transgenic tool kit for the in vivo modulation of Rho GTPase activity in zebrafish. Dev Dyn 2016; 245:844-53. [PMID: 27105927 PMCID: PMC4946996 DOI: 10.1002/dvdy.24412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Rho GTPases are small monomeric G-proteins that play key roles in many cellular processes. Due to Rho GTPases' widespread expression and broad functions, analyses of their function during late development require tissue-specific modulation of activity. The GAL4/UAS system provides an excellent tool for investigating the function of Rho GTPases in vivo. With this in mind, we created a transgenic tool kit enabling spatial and temporal modulation of Rho GTPase activity in zebrafish. RESULTS Transgenic constructs were assembled driving dominant-negative, constitutively active, and wild-type versions of Cdc42, RhoA, and Rac1 under 10XUAS control. The self-cleaving viral peptide F2A was utilized to allow bicistronic expression of a fluorescent reporter and Rho GTPase. Global heat shock of hsp70l:gal4(+) transgenic embryos confirmed GAL4-specific construct expression. Western blot analysis indicated myc-tagged Rho GTPases were expressed only in the presence of GAL4. Construct expression was confined to proper cells when combined with pou4f3:gal4 or ptf1a:gal4. Finally, transgene expression resulted in reproducible defects in lens formation, indicating that the transgenes are functional in vivo. CONCLUSIONS We generated and validated 10 transgenic lines, creating a versatile tool kit for the temporal-spatial modulation of Cdc42, RhoA, and Rac1 activity in vivo. These lines will enable systematic analysis of Rho GTPase function in any tissue of interest. Developmental Dynamics 245:844-853, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicholas J Hanovice
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Emily McMains
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
| | - Jeffrey M Gross
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
8
|
Kozol RA, Abrams AJ, James DM, Buglo E, Yan Q, Dallman JE. Function Over Form: Modeling Groups of Inherited Neurological Conditions in Zebrafish. Front Mol Neurosci 2016; 9:55. [PMID: 27458342 PMCID: PMC4935692 DOI: 10.3389/fnmol.2016.00055] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022] Open
Abstract
Zebrafish are a unique cell to behavior model for studying the basic biology of human inherited neurological conditions. Conserved vertebrate genetics and optical transparency provide in vivo access to the developing nervous system as well as high-throughput approaches for drug screens. Here we review zebrafish modeling for two broad groups of inherited conditions that each share genetic and molecular pathways and overlap phenotypically: neurodevelopmental disorders such as Autism Spectrum Disorders (ASD), Intellectual Disability (ID) and Schizophrenia (SCZ), and neurodegenerative diseases, such as Cerebellar Ataxia (CATX), Hereditary Spastic Paraplegia (HSP) and Charcot-Marie Tooth Disease (CMT). We also conduct a small meta-analysis of zebrafish orthologs of high confidence neurodevelopmental disorder and neurodegenerative disease genes by looking at duplication rates and relative protein sizes. In the past zebrafish genetic models of these neurodevelopmental disorders and neurodegenerative diseases have provided insight into cellular, circuit and behavioral level mechanisms contributing to these conditions. Moving forward, advances in genetic manipulation, live imaging of neuronal activity and automated high-throughput molecular screening promise to help delineate the mechanistic relationships between different types of neurological conditions and accelerate discovery of therapeutic strategies.
Collapse
Affiliation(s)
- Robert A. Kozol
- Department of Biology, University of MiamiCoral Gables, FL, USA
| | - Alexander J. Abrams
- Department of Human Genetics, John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation, University of MiamiMiami, FL, USA
| | - David M. James
- Department of Biology, University of MiamiCoral Gables, FL, USA
| | - Elena Buglo
- Department of Human Genetics, John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation, University of MiamiMiami, FL, USA
| | - Qing Yan
- Department of Biology, University of MiamiCoral Gables, FL, USA
| | | |
Collapse
|
9
|
Srinivasulu YS, Wang JR, Hsu KT, Tsai MJ, Charoenkwan P, Huang WL, Huang HL, Ho SY. Characterizing informative sequence descriptors and predicting binding affinities of heterodimeric protein complexes. BMC Bioinformatics 2015; 16 Suppl 18:S14. [PMID: 26681483 PMCID: PMC4682391 DOI: 10.1186/1471-2105-16-s18-s14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Protein-protein interactions (PPIs) are involved in various biological processes, and underlying mechanism of the interactions plays a crucial role in therapeutics and protein engineering. Most machine learning approaches have been developed for predicting the binding affinity of protein-protein complexes based on structure and functional information. This work aims to predict the binding affinity of heterodimeric protein complexes from sequences only. Results This work proposes a support vector machine (SVM) based binding affinity classifier, called SVM-BAC, to classify heterodimeric protein complexes based on the prediction of their binding affinity. SVM-BAC identified 14 of 580 sequence descriptors (physicochemical, energetic and conformational properties of the 20 amino acids) to classify 216 heterodimeric protein complexes into low and high binding affinity. SVM-BAC yielded the training accuracy, sensitivity, specificity, AUC and test accuracy of 85.80%, 0.89, 0.83, 0.86 and 83.33%, respectively, better than existing machine learning algorithms. The 14 features and support vector regression were further used to estimate the binding affinities (Pkd) of 200 heterodimeric protein complexes. Prediction performance of a Jackknife test was the correlation coefficient of 0.34 and mean absolute error of 1.4. We further analyze three informative physicochemical properties according to their contribution to prediction performance. Results reveal that the following properties are effective in predicting the binding affinity of heterodimeric protein complexes: apparent partition energy based on buried molar fractions, relations between chemical structure and biological activity in principal component analysis IV, and normalized frequency of beta turn. Conclusions The proposed sequence-based prediction method SVM-BAC uses an optimal feature selection method to identify 14 informative features to classify and predict binding affinity of heterodimeric protein complexes. The characterization analysis revealed that the average numbers of beta turns and hydrogen bonds at protein-protein interfaces in high binding affinity complexes are more than those in low binding affinity complexes.
Collapse
|
10
|
Jeffery J, Neyt C, Moore W, Paterson S, Bower NI, Chenevix‐Trench G, Verkade H, Hogan BM, Khanna KK. Cep55 regulates embryonic growth and development by promoting Akt stability in zebrafish. FASEB J 2015; 29:1999-2009. [DOI: 10.1096/fj.14-265090] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Jessie Jeffery
- Signal Transduction Laboratory, QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Christine Neyt
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Wade Moore
- Zebrafish Developmental Genetics Laboratory, Monash UniversityClaytonVictoriaAustralia
| | - Scott Paterson
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Neil I. Bower
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Georgia Chenevix‐Trench
- Cancer Genetics Laboratory, QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Heather Verkade
- Zebrafish Developmental Genetics Laboratory, Monash UniversityClaytonVictoriaAustralia
| | - Benjamin M. Hogan
- Vascular Biology and Development Laboratory, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQueenslandAustralia
| | - Kum Kum Khanna
- Signal Transduction Laboratory, QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| |
Collapse
|
11
|
Kirjavainen A, Laos M, Anttonen T, Pirvola U. The Rho GTPase Cdc42 regulates hair cell planar polarity and cellular patterning in the developing cochlea. Biol Open 2015; 4:516-26. [PMID: 25770185 PMCID: PMC4400594 DOI: 10.1242/bio.20149753] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hair cells of the organ of Corti (OC) of the cochlea exhibit distinct planar polarity, both at the tissue and cellular level. Planar polarity at tissue level is manifested as uniform orientation of the hair cell stereociliary bundles. Hair cell intrinsic polarity is defined as structural hair bundle asymmetry; positioning of the kinocilium/basal body complex at the vertex of the V-shaped bundle. Consistent with strong apical polarity, the hair cell apex displays prominent actin and microtubule cytoskeletons. The Rho GTPase Cdc42 regulates cytoskeletal dynamics and polarization of various cell types, and, thus, serves as a candidate regulator of hair cell polarity. We have here induced Cdc42 inactivation in the late-embryonic OC. We show the role of Cdc42 in the establishment of planar polarity of hair cells and in cellular patterning. Abnormal planar polarity was displayed as disturbances in hair bundle orientation and morphology and in kinocilium/basal body positioning. These defects were accompanied by a disorganized cell-surface microtubule network. Atypical protein kinase C (aPKC), a putative Cdc42 effector, colocalized with Cdc42 at the hair cell apex, and aPKC expression was altered upon Cdc42 depletion. Our data suggest that Cdc42 together with aPKC is part of the machinery establishing hair cell planar polarity and that Cdc42 acts on polarity through the cell-surface microtubule network. The data also suggest that defects in apical polarization are influenced by disturbed cellular patterning in the OC. In addition, our data demonstrates that Cdc42 is required for stereociliogenesis in the immature cochlea.
Collapse
Affiliation(s)
- Anna Kirjavainen
- Department of Biosciences, Viikinkaari 1, 00014 University of Helsinki, Finland
| | - Maarja Laos
- Department of Biosciences, Viikinkaari 1, 00014 University of Helsinki, Finland
| | - Tommi Anttonen
- Department of Biosciences, Viikinkaari 1, 00014 University of Helsinki, Finland
| | - Ulla Pirvola
- Department of Biosciences, Viikinkaari 1, 00014 University of Helsinki, Finland
| |
Collapse
|
12
|
Stumpf M, Choorapoikayil S, den Hertog J. Pten function in zebrafish: anything but a fish story. Methods 2014; 77-78:191-6. [PMID: 25461815 DOI: 10.1016/j.ymeth.2014.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/28/2014] [Accepted: 11/01/2014] [Indexed: 12/13/2022] Open
Abstract
Zebrafish is an excellent model system for the analysis of gene function. We and others use zebrafish to investigate the function of the tumor suppressor, Pten, in tumorigenesis and embryonic development. Zebrafish have two pten genes, ptena and ptenb. The recently identified N-terminal extension of human PTEN that may facilitate cell membrane transfer, appears not to be conserved in zebrafish Ptena or Ptenb. Mutants that retain a single wild type pten allele develop tumors, predominantly hemangiosarcomas. Homozygous double mutants are embryonic lethal. Zebrafish embryos lacking functional Pten display enhanced proliferation of endothelial cells, resulting in hyperbranching of blood vessels. In addition, ptena-/-ptenb-/- mutant embryos display enhanced proliferation of hematopoietic stem and progenitor cells and concomitant arrest of differentiation, although Pten-deficient cells commit to all blood cell lineages. Zebrafish is an ideal model for intravital imaging and future work using ptena-/-ptenb-/- mutants will enhance our understanding of the function of Pten in vivo.
Collapse
Affiliation(s)
- Miriam Stumpf
- Hubrecht Institute - KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Suma Choorapoikayil
- Hubrecht Institute - KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; CNRS, UMR 5235, Dynamique des Interactions Membranaires Normales et Pathologiques, Univ Montpellier 2, 34095 Montpellier, France
| | - Jeroen den Hertog
- Hubrecht Institute - KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Institute of Biology Leiden, Leiden University, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
13
|
Lee SJ. Dynamic regulation of the microtubule and actin cytoskeleton in zebrafish epiboly. Biochem Biophys Res Commun 2014; 452:1-7. [PMID: 25117442 DOI: 10.1016/j.bbrc.2014.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/01/2014] [Indexed: 11/17/2022]
Abstract
Gastrulation is a key developmental stage with striking changes in morphology. Coordinated cell movements occur to bring cells to their correct positions in a timely manner. Cell movements and morphological changes are accomplished by precisely controlling dynamic changes in cytoskeletal proteins, microtubules, and actin filaments. Among those cellular movements, epiboly produces the first distinct morphological changes in teleosts. In this review, I describe epiboly and its mechanics, and the dynamic changes in microtubule networks and actin structures, mainly in zebrafish embryos. The factors regulating those cytoskeletal changes will also be discussed.
Collapse
Affiliation(s)
- Shyh-Jye Lee
- Department of Life Science, National Taiwan University, 1 Roosevelt Rd., Sec., 4, Taipei 10617, Taiwan, ROC; Center for Biotechnology, National Taiwan University, 1 Roosevelt Rd., Sec., 4, Taipei 10617, Taiwan, ROC; Center for Developmental Biology and Regenerative Medicine, National Taiwan University, 1 Roosevelt Rd., Sec., 4, Taipei 10617, Taiwan, ROC; Center for System Biology, National Taiwan University, 1 Roosevelt Rd., Sec., 4, Taipei 10617, Taiwan, ROC.
| |
Collapse
|
14
|
Pten regulates homeostasis and inflammation-induced migration of myelocytes in zebrafish. J Hematol Oncol 2014; 7:17. [PMID: 24598081 PMCID: PMC4015859 DOI: 10.1186/1756-8722-7-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 02/27/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Loss of the tumor suppressor phosphatase and tensin homolog (PTEN) is frequently observed in hematopoietic malignancies. Although PTEN has been implicated in maintaining the quiescence of hematopoietic stem cells (HSCs), its role in hematopoiesis during ontogeny remains largely unexplored. METHODS The expression of hematopoietic marker genes was analyzed via whole mount in situ hybridization assay in ptena and ptenb double mutant zebrafish. The embryonic myelopoiesis was characterized by living imaging and whole mount in situ immunofluorescence with confocal microscopy, as well as cell-specific chemical staining for neutrophils and macrophages. Analyses of the involved signaling pathway were carried out by inhibitor treatment and mRNA injection. RESULTS Pten-deficient zebrafish embryos exhibited a strikingly increased number of myeloid cells, which were further characterized as being immune deficient. In accordance with this finding, the inhibition of phosphoinositide 3-kinase (PI3K) or the mechanistic target of rapamycin (mTOR) corrected the expansive myelopoiesis in the pten-deficient embryos. Further mechanistic studies revealed that the expression of cebpa, a critical transcription factor in myeloid precursor cells, was downregulated in the pten-deficient myeloid cells, whereas the injection of cebpa mRNA markedly ameliorated the dysmyelopoiesis induced by the loss of pten. CONCLUSIONS Our data provide in vivo evidence that definitive myelopoiesis in zebrafish is critically regulated by pten via the elevation of cebpa expression.
Collapse
|
15
|
Genome wide analysis reveals Zic3 interaction with distal regulatory elements of stage specific developmental genes in zebrafish. PLoS Genet 2013; 9:e1003852. [PMID: 24204288 PMCID: PMC3814314 DOI: 10.1371/journal.pgen.1003852] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 08/19/2013] [Indexed: 02/06/2023] Open
Abstract
Zic3 regulates early embryonic patterning in vertebrates. Loss of Zic3 function is known to disrupt gastrulation, left-right patterning, and neurogenesis. However, molecular events downstream of this transcription factor are poorly characterized. Here we use the zebrafish as a model to study the developmental role of Zic3 in vivo, by applying a combination of two powerful genomics approaches – ChIP-seq and microarray. Besides confirming direct regulation of previously implicated Zic3 targets of the Nodal and canonical Wnt pathways, analysis of gastrula stage embryos uncovered a number of novel candidate target genes, among which were members of the non-canonical Wnt pathway and the neural pre-pattern genes. A similar analysis in zic3-expressing cells obtained by FACS at segmentation stage revealed a dramatic shift in Zic3 binding site locations and identified an entirely distinct set of target genes associated with later developmental functions such as neural development. We demonstrate cis-regulation of several of these target genes by Zic3 using in vivo enhancer assay. Analysis of Zic3 binding sites revealed a distribution biased towards distal intergenic regions, indicative of a long distance regulatory mechanism; some of these binding sites are highly conserved during evolution and act as functional enhancers. This demonstrated that Zic3 regulation of developmental genes is achieved predominantly through long distance regulatory mechanism and revealed that developmental transitions could be accompanied by dramatic changes in regulatory landscape. The Zic3 transcription factor regulates early embryonic patterning, and the loss of its function leads to defects in left-right body asymmetry. Previous studies have only identified a small number of Zic3 targets, which renders the molecular mechanism underlying its activity insufficiently understood. Utilizing two genomics technologies, next generation sequencing and microarray, we profile the genome-wide binding sites of Zic3 and identified its target genes in the developing zebrafish embryo. Our results show that Zic3 regulates its target genes predominantly through regulatory elements located far from promoters. Among the targets of Zic3 are the Nodal and Wnt pathways known to regulate gastrulation and left-right body asymmetry, as well as neural pre-pattern genes regulating proliferation of neural progenitors. Using enhancer activity assay, we further show that genomic regions bound by Zic3 function as enhancers. Our study provides a genome-wide view of the regulatory landscape of Zic3 and its changes during vertebrate development.
Collapse
|
16
|
Hu CW, Tseng CW, Chien CW, Huang HC, Ku WC, Lee SJ, Chen YJ, Juan HF. Quantitative proteomics reveals diverse roles of miR-148a from gastric cancer progression to neurological development. J Proteome Res 2013; 12:3993-4004. [PMID: 23869555 DOI: 10.1021/pr400302w] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that control gene expression either by degradation of mRNAs or inhibition of protein translation. miR-148a has been reported to have the impacts on tumor progression. Here, a quantitative proteomics combined with stable isotope labeling was applied to identify the global profile of miR-148a-regulated downstream proteins. The data have been deposited to the ProteomeXchange with identifier PXD000190. A total of 2938 proteins were quantified, and 55 proteins were considered to be regulated by miR-148a. We found that not only proteins associated with cancer progression but also molecules involved in neural development were regulated by miR-148a. This study is the first to identify the function of miR-148a in neural development by using a proteomic approach. Analysis of a public clinical database also showed that the patients with neural diseases could display abnormal expression of miR-148a. Moreover, silencing of miR-148a led to the abnormal morphology and decreased expression of neuron-related markers in the developing brain of zebrafish. These results provided important insight into the regulation of neurological development elicited by miR-148a.
Collapse
Affiliation(s)
- Chia-Wei Hu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Cheng YC, Scotting PJ, Hsu LS, Lin SJ, Shih HY, Hsieh FY, Wu HL, Tsao CL, Shen CJ. Zebrafish rgs4 is essential for motility and axonogenesis mediated by Akt signaling. Cell Mol Life Sci 2013; 70:935-50. [PMID: 23052218 PMCID: PMC11113239 DOI: 10.1007/s00018-012-1178-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 09/19/2012] [Accepted: 09/24/2012] [Indexed: 12/21/2022]
Abstract
The schizophrenia susceptibility gene, Rgs4, is one of the most intensively studied regulators of G-protein signaling members, well known to be fundamental in regulating neurotransmission. However, little is known about its role in the developing nervous system. We have isolated zebrafish rgs4 and shown that it is transcribed in the developing nervous system. Rgs4 knockdown did not affect neuron number and patterning but resulted in locomotion defects and aberrant development of axons. This was confirmed using a selective Rgs4 inhibitor, CCG-4986. Rgs4 knockdown also attenuated the level of phosphorylated-Akt1, and injection of constitutively-activated AKT1 rescued the motility defects and axonal phenotypes in the spinal cord but not in the hindbrain and trigeminal neurons. Our in vivo analysis reveals a novel role for Rgs4 in regulating axonogenesis during embryogenesis, which is mediated by another schizophrenia-associated gene, Akt1, in a region-specific manner.
Collapse
Affiliation(s)
- Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, 33383, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Bloomekatz J, Grego-Bessa J, Migeotte I, Anderson KV. Pten regulates collective cell migration during specification of the anterior-posterior axis of the mouse embryo. Dev Biol 2012; 364:192-201. [PMID: 22342906 DOI: 10.1016/j.ydbio.2012.02.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 02/03/2012] [Accepted: 02/04/2012] [Indexed: 01/23/2023]
Abstract
Pten, the potent tumor suppressor, is a lipid phosphatase that is best known as a regulator of cell proliferation and cell survival. Here we show that mouse embryos that lack Pten have a striking set of morphogenetic defects, including the failure to correctly specify the anterior-posterior body axis, that are not caused by changes in proliferation or cell death. The majority of Pten null embryos express markers of the primitive streak at ectopic locations around the embryonic circumference, rather than at a single site at the posterior of the embryo. Epiblast-specific deletion shows that Pten is not required in the cells of the primitive streak; instead, Pten is required for normal migration of cells of the Anterior Visceral Endoderm (AVE), an extraembryonic organizer that controls the position of the streak. Cells of the wild-type AVE migrate within the visceral endoderm epithelium from the distal tip of the embryo to a position adjacent to the extraembryonic region. In all Pten null mutants, AVE cells move a reduced distance and disperse in random directions, instead of moving as a coordinated group to the anterior of the embryo. Aberrant AVE migration is associated with the formation of ectopic F-actin foci, which indicates that absence of Pten disrupts the actin-based migration of these cells. After the initiation of gastrulation, embryos that lack Pten in the epiblast show defects in the migration of mesoderm and/or endoderm. The findings suggest that Pten has an essential and general role in the control of mammalian collective cell migration.
Collapse
Affiliation(s)
- Joshua Bloomekatz
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | | | | | | |
Collapse
|