1
|
Korzeniwsky KG, de Mello PL, Liang Y, Feltes M, Farber SA, Parichy DM. Dominant Negative Mitf Allele Impacts Melanophore and Xanthophore Development and Reveals Collaborative Interactions With Tfec in Zebrafish Chromatophore Lineages. Pigment Cell Melanoma Res 2025; 38:e70009. [PMID: 40123122 PMCID: PMC11931198 DOI: 10.1111/pcmr.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
Ectothermic vertebrates exhibit a diverse array of pigment cell types-chromatophores-that provide valuable opportunities to uncover mechanisms of fate specification and how they evolve. Like melanocytes of mammals, the melanophores of teleosts and other ectotherms depend on basic helix-loop-helix leucine zipper transcription factors encoded by orthologues of MITF. A different chromatophore, the iridescent iridophore, depends on the closely related transcription factor Tfec. Requirements for the specification of other chromatophore lineages remain largely uncertain. Here we identify a new allele of the zebrafish Mitf gene, mitfa, that results in a complete absence of not only melanophores but also yellow-orange xanthophores. Harboring a missense substitution in the DNA-binding domain identical to previously isolated alleles of mouse, we show that this new allele has defects in chromatophore precursor survival and xanthophore differentiation that extend beyond those of mitfa loss-of-function. Additional genetic analyses revealed interactions between Mitfa and Tfec as a likely basis for the observed phenotypes. Our findings point to collaborative roles for Mitfa and Tfec in promoting chromatophore development, particularly in xanthophore lineages, and provide new insights into evolutionary aspects of MITF functions across vertebrates.
Collapse
Affiliation(s)
| | | | - Yipeng Liang
- Department of BiologyUniversity of VirginiaVirginiaUSA
| | - McKenna Feltes
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Steven A. Farber
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - David M. Parichy
- Department of BiologyUniversity of VirginiaVirginiaUSA
- Department of Cell BiologyUniversity of VirginiaVirginiaUSA
| |
Collapse
|
2
|
Hao R, Li L, Zhang D, Tian Y, Long H, Li H, Zhu X, Huang Y, Li G, Zhu C. Characterization and functional analysis of pl-miR-2188 in melanin synthesis in leopard coral grouper (Plectropomus leopardus). Comp Biochem Physiol B Biochem Mol Biol 2024; 275:111043. [PMID: 39491612 DOI: 10.1016/j.cbpb.2024.111043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
MicroRNAs (miRNAs) are known to regulate gene expression and play a role in body color formation in fish. However, the molecular mechanisms underlying miRNA involvement in the body color of leopard coral grouper (Plectropomus leopardus) remain largely unexplored. In this study, we investigated the expression levels of miR-2188 in red and black P. leopardus (pl-miR-2188) and found significantly higher expression levels in red fish samples compared to those in black fish samples. Silencing pl-miR-2188 in vivo using a pl-miR-2188 antagomir resulted in increased melanin concentration. Following pl-miR-2188 silencing, the expression levels of melanin-related genes, such as tyrosinase (tyr), TYR-related protein 1 (tyrp1-1 and tyrp1-2) and TYR-related protein 2 (tyrp2), and microphthalmia-associated transcription factor (mitf), were elevated. RNAhybrid predictions and dual-luciferase reporter assays identified sox5 as a target mRNA of pl-miR-2188. Following pl-miR-2188 antagomir injection, sox5 expression was significantly upregulated in the injection group compared to that in control groups (P < 0.05). These results suggest that pl-miR-2188 may regulate melanin synthesis in P. leopardus by targeting sox5. This study provides new insights into the miRNA-mRNA interactions involved in melanin synthesis and body color formation in the leopard coral grouper.
Collapse
Affiliation(s)
- Ruijuan Hao
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China.
| | - Liancheng Li
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China; Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Dongying Zhang
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China
| | - Yali Tian
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China; Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Hongzhao Long
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China; Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Hang Li
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China
| | - Xiaowen Zhu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Zhanjiang 524088, China
| | - Yang Huang
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China; Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Guangli Li
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Zhanjiang 524088, China
| | - Chunhua Zhu
- Development and research center for biological marine resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China; Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
3
|
Abstract
Melanocytes evolved to produce the melanin that gives colour to our hair, eyes and skin. The melanocyte lineage also gives rise to melanoma, the most lethal form of skin cancer. The melanocyte lineage differentiates from neural crest cells during development, and most melanocytes reside in the skin and hair, where they are replenished by melanocyte stem cells. Because the molecular mechanisms necessary for melanocyte specification, migration, proliferation and differentiation are co-opted during melanoma initiation and progression, studying melanocyte development is directly relevant to human disease. Here, through the lens of advances in cellular omic and genomic technologies, we review the latest findings in melanocyte development and differentiation, and how these developmental pathways become dysregulated in disease.
Collapse
Affiliation(s)
- Alessandro Brombin
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - E Elizabeth Patton
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
4
|
Tang CY, Zhang X, Xu X, Sun S, Peng C, Song MH, Yan C, Sun H, Liu M, Xie L, Luo SJ, Li JT. Genetic mapping and molecular mechanism behind color variation in the Asian vine snake. Genome Biol 2023; 24:46. [PMID: 36895044 PMCID: PMC9999515 DOI: 10.1186/s13059-023-02887-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Reptiles exhibit a wide variety of skin colors, which serve essential roles in survival and reproduction. However, the molecular basis of these conspicuous colors remains unresolved. RESULTS We investigate color morph-enriched Asian vine snakes (Ahaetulla prasina), to explore the mechanism underpinning color variations. Transmission electron microscopy imaging and metabolomics analysis indicates that chromatophore morphology (mainly iridophores) is the main basis for differences in skin color. Additionally, we assemble a 1.77-Gb high-quality chromosome-anchored genome of the snake. Genome-wide association study and RNA sequencing reveal a conservative amino acid substitution (p.P20S) in SMARCE1, which may be involved in the regulation of chromatophore development initiated from neural crest cells. SMARCE1 knockdown in zebrafish and immunofluorescence verify the interactions among SMARCE1, iridophores, and tfec, which may determine color variations in the Asian vine snake. CONCLUSIONS This study reveals the genetic associations of color variation in Asian vine snakes, providing insights and important resources for a deeper understanding of the molecular and genetic mechanisms related to reptilian coloration.
Collapse
Affiliation(s)
- Chen-Yang Tang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration and Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Xiaohu Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiao Xu
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Shijie Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Changjun Peng
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration and Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meng-Huan Song
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration and Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chaochao Yan
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration and Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Huaqin Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingfeng Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Liang Xie
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Shu-Jin Luo
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jia-Tang Li
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration and Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Yezin Nay Pyi Taw, 05282, Myanmar.
| |
Collapse
|
5
|
Kenny C, Dilshat R, Seberg HE, Van Otterloo E, Bonde G, Helverson A, Franke CM, Steingrímsson E, Cornell RA. TFAP2 paralogs facilitate chromatin access for MITF at pigmentation and cell proliferation genes. PLoS Genet 2022; 18:e1010207. [PMID: 35580127 PMCID: PMC9159589 DOI: 10.1371/journal.pgen.1010207] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/01/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022] Open
Abstract
In developing melanocytes and in melanoma cells, multiple paralogs of the Activating-enhancer-binding Protein 2 family of transcription factors (TFAP2) contribute to expression of genes encoding pigmentation regulators, but their interaction with Microphthalmia transcription factor (MITF), a master regulator of these cells, is unclear. Supporting the model that TFAP2 facilitates MITF's ability to activate expression of pigmentation genes, single-cell seq analysis of zebrafish embryos revealed that pigmentation genes are only expressed in the subset of mitfa-expressing cells that also express tfap2 paralogs. To test this model in SK-MEL-28 melanoma cells we deleted the two TFAP2 paralogs with highest expression, TFAP2A and TFAP2C, creating TFAP2 knockout (TFAP2-KO) cells. We then assessed gene expression, chromatin accessibility, binding of TFAP2A and of MITF, and the chromatin marks H3K27Ac and H3K27Me3 which are characteristic of active enhancers and silenced chromatin, respectively. Integrated analyses of these datasets indicate TFAP2 paralogs directly activate enhancers near genes enriched for roles in pigmentation and proliferation, and directly repress enhancers near genes enriched for roles in cell adhesion. Consistently, compared to WT cells, TFAP2-KO cells proliferate less and adhere to one another more. TFAP2 paralogs and MITF co-operatively activate a subset of enhancers, with the former necessary for MITF binding and chromatin accessibility. By contrast, TFAP2 paralogs and MITF do not appear to co-operatively inhibit enhancers. These studies reveal a mechanism by which TFAP2 profoundly influences the set of genes activated by MITF, and thereby the phenotype of pigment cells and melanoma cells.
Collapse
Affiliation(s)
- Colin Kenny
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Ramile Dilshat
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Hannah E. Seberg
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Eric Van Otterloo
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Gregory Bonde
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Annika Helverson
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Christopher M. Franke
- Department of Surgery, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Robert A. Cornell
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
6
|
Min Y, Li Q, Yu H. Heme-Peroxidase 2 Modulated by POU2F1 and SOX5 is Involved in Pigmentation in Pacific Oyster (Crassostrea gigas). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:263-275. [PMID: 35275290 DOI: 10.1007/s10126-022-10098-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
Color polymorphism is frequently observed in molluscan shellfish, while the molecular regulation of shell pigmentation is not well understood. Peroxidase is a key enzyme involved in melanogenesis. Here, we identified a heme-peroxidase 2 gene (CgHPX2), and characterized the expression patterns and transcriptional regulation of CgHPX2 in the Pacific oyster Crassostrea gigas. Tissues expression analysis showed that CgHPX2 was a mantle-specific gene and primarily expressed in the edge mantle in black shell color oyster compared with white shell oyster. In situ hybridization showed that strong signals for CgHPX2 were detected in the both inner and outer surface of the outer fold of mantle in the black shell color oyster, whereas positive signals in white shell oyster were mainly localized in the outer surface of the outer fold of mantle. In the embryos and larvae, a high expression level of CgHPX2 was detected in the trochophore stage in both black and white shell color oysters. The temporal localization of CgHPX2 was mainly detected in the shell gland and edge mantle of trochophore and calcified shell larvae, respectively. In addition, a 2227 bp of 5' flanking region sequence of CgHPX2 was cloned, which contained a presumed core promoter region and many potential transcription factor binding sites. Further luciferase assay experiment confirmed that POU domain, class 2, transcription factor 1 (POU2F1), and SRY-box transcription factor 5 (SOX5) were involved in transcriptional regulation of CgHPX2 gene through binding to its specific promoter region. After CgPOU2F1 and CgSOX5 RNA interference, the CgHPX2 gene expression was significantly decreased. These results suggested that CgPOU2F1 and CgSOX5 might be two important transcription factors that positively regulated the expression of CgHPX2 gene, improving our understanding of the transcriptional regulation of molluscan shell pigmentation.
Collapse
Affiliation(s)
- Yue Min
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Qi Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education College of Fisheries, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - Hong Yu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education College of Fisheries, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
7
|
Zebrafish Syndromic Albinism Models as Tools for Understanding and Treating Pigment Cell Disease in Humans. Cancers (Basel) 2022; 14:cancers14071752. [PMID: 35406524 PMCID: PMC8997128 DOI: 10.3390/cancers14071752] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/18/2022] [Accepted: 03/26/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Zebrafish (Danio rerio) is an emerging model for studying many diseases, including disorders originating in black pigment cells, melanocytes. In this review of the melanocyte literature, we discuss the current knowledge of melanocyte biology relevant to understanding different forms of albinism and the potential of the zebrafish model system for finding novel mechanisms and treatments. Abstract Melanin is the pigment that protects DNA from ultraviolet (UV) damage by absorbing excess energy. Melanin is produced in a process called melanogenesis. When melanogenesis is altered, diseases such as albinism result. Albinism can result in an increased skin cancer risk. Conversely, black pigment cell (melanocyte) development pathways can be misregulated, causing excessive melanocyte growth that leads to melanoma (cancer of melanocytes). Zebrafish is an emerging model organism used to study pigment disorders due to their high fecundity, visible melanin development in melanophores (melanocytes in mammals) from 24 h post-fertilization, and conserved melanogenesis pathways. Here, we reviewed the conserved developmental pathways in zebrafish melanophores and mammalian melanocytes. Additionally, we summarized the progress made in understanding pigment cell disease and evidence supporting the strong potential for using zebrafish to find novel treatment options for albinism.
Collapse
|
8
|
Dawes JHP, Kelsh RN. Cell Fate Decisions in the Neural Crest, from Pigment Cell to Neural Development. Int J Mol Sci 2021; 22:13531. [PMID: 34948326 PMCID: PMC8706606 DOI: 10.3390/ijms222413531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
The neural crest shows an astonishing multipotency, generating multiple neural derivatives, but also pigment cells, skeletogenic and other cell types. The question of how this process is controlled has been the subject of an ongoing debate for more than 35 years. Based upon new observations of zebrafish pigment cell development, we have recently proposed a novel, dynamic model that we believe goes some way to resolving the controversy. Here, we will firstly summarize the traditional models and the conflicts between them, before outlining our novel model. We will also examine our recent dynamic modelling studies, looking at how these reveal behaviors compatible with the biology proposed. We will then outline some of the implications of our model, looking at how it might modify our views of the processes of fate specification, differentiation, and commitment.
Collapse
Affiliation(s)
- Jonathan H. P. Dawes
- Centre for Networks and Collective Behaviour, University of Bath, Bath BA2 7AY, UK;
- Department of Mathematical Sciences, University of Bath, Bath BA2 7AY, UK
| | - Robert N. Kelsh
- Centre for Mathematical Biology, University of Bath, Bath BA2 7AY, UK
- Department of Biology & Biochemistry, University of Bath, Bath BA2 7AY, UK
| |
Collapse
|
9
|
Colombo S, Petit V, Wagner RY, Champeval D, Yajima I, Gesbert F, Aktary Z, Davidson I, Delmas V, Larue L. Stabilization of β-catenin promotes melanocyte specification at the expense of the Schwann cell lineage. Development 2021; 149:274086. [PMID: 34878101 PMCID: PMC8917410 DOI: 10.1242/dev.194407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/25/2021] [Indexed: 11/20/2022]
Abstract
The canonical Wnt/β-catenin pathway governs a multitude of developmental processes in various cell lineages, including the melanocyte lineage. Indeed, β-catenin regulates transcription of Mitf-M, the master regulator of this lineage. The first wave of melanocytes to colonize the skin is directly derived from neural crest cells, whereas the second wave of melanocytes is derived from Schwann cell precursors (SCPs). We investigated the influence of β-catenin in the development of melanocytes of the first and second waves by generating mice expressing a constitutively active form of β-catenin in cells expressing tyrosinase. Constitutive activation of β-catenin did not affect the development of truncal melanoblasts but led to marked hyperpigmentation of the paws. By activating β-catenin at various stages of development (E8.5-E11.5), we showed that the activation of β-catenin in bipotent SCPs favored melanoblast specification at the expense of Schwann cells in the limbs within a specific temporal window. Furthermore, in vitro hyperactivation of the Wnt/β-catenin pathway, which is required for melanocyte development, induces activation of Mitf-M, in turn repressing FoxD3 expression. In conclusion, β-catenin overexpression promotes SCP cell fate decisions towards the melanocyte lineage. Summary: Activation of β-catenin in bipotent Schwann cell precursors during a specific developmental window induces Mitf and represses FoxD3 to promote melanoblast cell fate at the expense of Schwann cells in limbs.
Collapse
Affiliation(s)
- Sophie Colombo
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Valérie Petit
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Roselyne Y Wagner
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Delphine Champeval
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Ichiro Yajima
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Franck Gesbert
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Zackie Aktary
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Irwin Davidson
- Equipes Labellisées Ligue Contre le Cancer, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, 1 Rue Laurent Fries, 67404 Illkirch Cedex. Department of Functional Genomics and Cancer, France
| | - Véronique Delmas
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Lionel Larue
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| |
Collapse
|
10
|
NRG1/ErbB signalling controls the dialogue between macrophages and neural crest-derived cells during zebrafish fin regeneration. Nat Commun 2021; 12:6336. [PMID: 34732706 PMCID: PMC8566576 DOI: 10.1038/s41467-021-26422-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 09/07/2021] [Indexed: 11/12/2022] Open
Abstract
Fish species, such as zebrafish (Danio rerio), can regenerate their appendages after amputation through the formation of a heterogeneous cellular structure named blastema. Here, by combining live imaging of triple transgenic zebrafish embryos and single-cell RNA sequencing we established a detailed cell atlas of the regenerating caudal fin in zebrafish larvae. We confirmed the presence of macrophage subsets that govern zebrafish fin regeneration, and identified a foxd3-positive cell population within the regenerating fin. Genetic depletion of these foxd3-positive neural crest-derived cells (NCdC) showed that they are involved in blastema formation and caudal fin regeneration. Finally, chemical inhibition and transcriptomic analysis demonstrated that these foxd3-positive cells regulate macrophage recruitment and polarization through the NRG1/ErbB pathway. Here, we show the diversity of the cells required for blastema formation, identify a discrete foxd3-positive NCdC population, and reveal the critical function of the NRG1/ErbB pathway in controlling the dialogue between macrophages and NCdC. Some fish can regenerate appendages by formation of a structure called the blastema. Here, the authors use single-cell RNA sequencing to characterize the cells required for blastema formation and fin regeneration and identified neural crest cells that orchestrate regeneration via the NRG1/ErbB axis
Collapse
|
11
|
Jang HS, Chen Y, Ge J, Wilkening AN, Hou Y, Lee HJ, Choi YR, Lowdon RF, Xing X, Li D, Kaufman CK, Johnson SL, Wang T. Epigenetic dynamics shaping melanophore and iridophore cell fate in zebrafish. Genome Biol 2021; 22:282. [PMID: 34607603 PMCID: PMC8489059 DOI: 10.1186/s13059-021-02493-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Zebrafish pigment cell differentiation provides an attractive model for studying cell fate progression as a neural crest progenitor engenders diverse cell types, including two morphologically distinct pigment cells: black melanophores and reflective iridophores. Nontrivial classical genetic and transcriptomic approaches have revealed essential molecular mechanisms and gene regulatory circuits that drive neural crest-derived cell fate decisions. However, how the epigenetic landscape contributes to pigment cell differentiation, especially in the context of iridophore cell fate, is poorly understood. RESULTS We chart the global changes in the epigenetic landscape, including DNA methylation and chromatin accessibility, during neural crest differentiation into melanophores and iridophores to identify epigenetic determinants shaping cell type-specific gene expression. Motif enrichment in the epigenetically dynamic regions reveals putative transcription factors that might be responsible for driving pigment cell identity. Through this effort, in the relatively uncharacterized iridophores, we validate alx4a as a necessary and sufficient transcription factor for iridophore differentiation and present evidence on alx4a's potential regulatory role in guanine synthesis pathway. CONCLUSIONS Pigment cell fate is marked by substantial DNA demethylation events coupled with dynamic chromatin accessibility to potentiate gene regulation through cis-regulatory control. Here, we provide a multi-omic resource for neural crest differentiation into melanophores and iridophores. This work led to the discovery and validation of iridophore-specific alx4a transcription factor.
Collapse
Affiliation(s)
- Hyo Sik Jang
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
- Present address: Department of Epigenetics, Van Andel Institute, Grand Rapids, MI USA
| | - Yujie Chen
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Jiaxin Ge
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Alicia N. Wilkening
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Yiran Hou
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Hyung Joo Lee
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - You Rim Choi
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Rebecca F. Lowdon
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
| | - Charles K. Kaufman
- Department of Medicine, Division of Medical Oncology, and Department of Developmental Biology, Washington University in Saint Louis, St. Louis, MO USA
| | - Stephen L. Johnson
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St Louis, MO USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO USA
| |
Collapse
|
12
|
Petratou K, Spencer SA, Kelsh RN, Lister JA. The MITF paralog tfec is required in neural crest development for fate specification of the iridophore lineage from a multipotent pigment cell progenitor. PLoS One 2021; 16:e0244794. [PMID: 33439865 PMCID: PMC7806166 DOI: 10.1371/journal.pone.0244794] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022] Open
Abstract
Understanding how fate specification of distinct cell-types from multipotent progenitors occurs is a fundamental question in embryology. Neural crest stem cells (NCSCs) generate extraordinarily diverse derivatives, including multiple neural, skeletogenic and pigment cell fates. Key transcription factors and extracellular signals specifying NCSC lineages remain to be identified, and we have only a little idea of how and when they function together to control fate. Zebrafish have three neural crest-derived pigment cell types, black melanocytes, light-reflecting iridophores and yellow xanthophores, which offer a powerful model for studying the molecular and cellular mechanisms of fate segregation. Mitfa has been identified as the master regulator of melanocyte fate. Here, we show that an Mitf-related transcription factor, Tfec, functions as master regulator of the iridophore fate. Surprisingly, our phenotypic analysis of tfec mutants demonstrates that Tfec also functions in the initial specification of all three pigment cell-types, although the melanocyte and xanthophore lineages recover later. We show that Mitfa represses tfec expression, revealing a likely mechanism contributing to the decision between melanocyte and iridophore fate. Our data are consistent with the long-standing proposal of a tripotent progenitor restricted to pigment cell fates. Moreover, we investigate activation, maintenance and function of tfec in multipotent NCSCs, demonstrating for the first time its role in the gene regulatory network forming and maintaining early neural crest cells. In summary, we build on our previous work to characterise the gene regulatory network governing iridophore development, establishing Tfec as the master regulator driving iridophore specification from multipotent progenitors, while shedding light on possible cellular mechanisms of progressive fate restriction.
Collapse
Affiliation(s)
- Kleio Petratou
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - Samantha A. Spencer
- Department of Human and Molecular Genetics and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States of America
| | - Robert N. Kelsh
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - James A. Lister
- Department of Human and Molecular Genetics and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States of America
| |
Collapse
|
13
|
Weigele J, Bohnsack BL. Genetics Underlying the Interactions between Neural Crest Cells and Eye Development. J Dev Biol 2020; 8:jdb8040026. [PMID: 33182738 PMCID: PMC7712190 DOI: 10.3390/jdb8040026] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022] Open
Abstract
The neural crest is a unique, transient stem cell population that is critical for craniofacial and ocular development. Understanding the genetics underlying the steps of neural crest development is essential for gaining insight into the pathogenesis of congenital eye diseases. The neural crest cells play an under-appreciated key role in patterning the neural epithelial-derived optic cup. These interactions between neural crest cells within the periocular mesenchyme and the optic cup, while not well-studied, are critical for optic cup morphogenesis and ocular fissure closure. As a result, microphthalmia and coloboma are common phenotypes in human disease and animal models in which neural crest cell specification and early migration are disrupted. In addition, neural crest cells directly contribute to numerous ocular structures including the cornea, iris, sclera, ciliary body, trabecular meshwork, and aqueous outflow tracts. Defects in later neural crest cell migration and differentiation cause a constellation of well-recognized ocular anterior segment anomalies such as Axenfeld–Rieger Syndrome and Peters Anomaly. This review will focus on the genetics of the neural crest cells within the context of how these complex processes specifically affect overall ocular development and can lead to congenital eye diseases.
Collapse
Affiliation(s)
- Jochen Weigele
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave, Chicago, IL 60611, USA;
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Chicago, IL 60611, USA
| | - Brenda L. Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave, Chicago, IL 60611, USA;
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, 645 N. Michigan Ave, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-312-227-6180; Fax: +1-312-227-9411
| |
Collapse
|
14
|
Sinagoga KL, Larimer-Picciani AM, George SM, Spencer SA, Lister JA, Gross JM. Mitf-family transcription factor function is required within cranial neural crest cells to promote choroid fissure closure. Development 2020; 147:dev187047. [PMID: 32541011 PMCID: PMC7375471 DOI: 10.1242/dev.187047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
A crucial step in eye development is the closure of the choroid fissure (CF), a transient structure in the ventral optic cup through which vasculature enters the eye and ganglion cell axons exit. Although many factors have been identified that function during CF closure, the molecular and cellular mechanisms mediating this process remain poorly understood. Failure of CF closure results in colobomas. Recently, MITF was shown to be mutated in a subset of individuals with colobomas, but how MITF functions during CF closure is unknown. To address this issue, zebrafish with mutations in mitfa and tfec, two members of the Mitf family of transcription factors, were analyzed and their functions during CF closure determined. mitfa;tfec mutants possess severe colobomas and our data demonstrate that Mitf activity is required within cranial neural crest cells (cNCCs) during CF closure. In the absence of Mitf function, cNCC migration and localization in the optic cup are perturbed. These data shed light on the cellular mechanisms underlying colobomas in individuals with MITF mutations and identify a novel role for Mitf function in cNCCs during CF closure.
Collapse
Affiliation(s)
- Katie L Sinagoga
- Departments of Ophthalmology and Developmental Biology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Alessandra M Larimer-Picciani
- Departments of Ophthalmology and Developmental Biology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Stephanie M George
- Departments of Ophthalmology and Developmental Biology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Samantha A Spencer
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - James A Lister
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Jeffrey M Gross
- Departments of Ophthalmology and Developmental Biology, Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
15
|
Liu Q, Cheng J, Lu Y, Zhou J, Wang L, Yang C, Yang G, Yang H, Cao J, Zhang Z, Sun Y. The clinical and genetic research of Waardenburg syndrome type I and II in Chinese families. Int J Pediatr Otorhinolaryngol 2020; 130:109806. [PMID: 31812001 DOI: 10.1016/j.ijporl.2019.109806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/31/2019] [Accepted: 11/26/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Waardenburg Syndrome (WS) is a neurocristopathy with an autosomal dominant mode of inheritance and highly genetic heterogeneity. To date, mutations of PAX3, SOX10, MITF, EDNRB, EDN3 and SNAI2 have been implicated in the pathogenesis of WS. In this study, we aimed to identify pathogenic genes among WS families and to analyze the pathogenic relationship between genotypes and phenotypes. METHODS In this study, all six families studied were from Hubei province, China.WS patients underwent screening for all deafness genes including PAX3, SOX10, MITF, EDNRB, EDN3 and SNAI2 using Massively Parallel Sequencing (MPS) and validation of mutations using Sanger sequencing. RESULTS Clinical evaluation revealed prominent phenotypic variability in Hubei WS patients. Two WS1 families and four WS2 families were diagnosed in six families. Sensorineural hearing loss was the most common, followed by iris pigmentary abnormality. Molecular genetic analysis of the WS genes for six families revealed five novel heterozygous mutations. Two mutations occurred in the PAX3 gene: one nonsense mutation c.667C > T(p.Arg223Ter) and one missense mutation c.220C > T(p.Arg74Cys).One missense mutation c.331T > C (p.Phe111Leu) and one nonsense mutation c.346C > T(p.Gln116Ter) were detected in the SOX10 gene. Two mutations were detected in the MITF gene: one splice site mutation c.859-1G > A and one nonsense mutation c.859G > T(p.Glu287Ter). Among them, the mutations (SOX10 c.331T > C and MITF c.859G > T) were de novo mutations. CONCLUSION In this study, six mutations were found to be associated with the phenotype of patients. Our data helped illuminate the phenotypic and genotypic spectrum of WS in Hubei province and could have implications for the genetic counseling of WS in Hubei province.
Collapse
Affiliation(s)
- Qin Liu
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Jing Cheng
- Medical Genetics Center, The First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Yu Lu
- Medical Genetics Center, The First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Jia Zhou
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Li Wang
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Changliang Yang
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Guang Yang
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Hui Yang
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Jingyuan Cao
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Zhao Zhang
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China
| | - Yi Sun
- Department of Otorhinolaryngology, General Hospital of Central Theater Command, Chinese People's Liberation Army, Wuhan, 430000, China; Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
16
|
Ren S, Chen X, Kong X, Chen Y, Wu Q, Jiao Z, Shi H. Identification of six novel variants in Waardenburg syndrome type II by next-generation sequencing. Mol Genet Genomic Med 2020; 8:e1128. [PMID: 31960627 PMCID: PMC7057110 DOI: 10.1002/mgg3.1128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 01/02/2020] [Indexed: 11/22/2022] Open
Abstract
Background Waardenburg syndrome (WS) is a dominantly inherited, genetically heterogeneous auditory‐pigmentary syndrome characterized by nonprogressive sensorineural hearing loss and iris discoloration. This study aimed to investigate the underlying molecular pathology in Chinese WS families. Methods A total of 13 patients with Waardenburg syndrome type II (WS2) from six unrelated Chinese families were enrolled. We investigated the mutation profile of genes related to congenital deafness in these families through a targeted sequencing technology and validated the candidate variants by Sanger sequencing. Results We identified six novel variants in microphthalmia‐associated transcription factor (MITF) and SRY‐box 10 (SOX10), which were predicted to be disease causing by in silico analysis. Our results showed that mutations in SOX10 and MITF are two major causes of deafness associated with WS, and de novo mutations were frequently found in probands with SOX10 mutations but not in those with MITF mutations. Conclusion Results showed that targeted next‐generation sequencing (NGS) enabled us to detect disease‐causing mutations with high accuracy, stability, speed and throughput. Our study extends the pathogenic mutation spectrum of MITF and SOX10.
Collapse
Affiliation(s)
- Shumin Ren
- Department of Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojie Chen
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangdong Kong
- Department of Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yibing Chen
- Department of Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qinghua Wu
- Department of Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihui Jiao
- Department of Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huirong Shi
- Department of obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Gramann AK, Venkatesan AM, Guerin M, Ceol CJ. Regulation of zebrafish melanocyte development by ligand-dependent BMP signaling. eLife 2019; 8:50047. [PMID: 31868592 PMCID: PMC6968919 DOI: 10.7554/elife.50047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/21/2019] [Indexed: 02/06/2023] Open
Abstract
Preventing terminal differentiation is important in the development and progression of many cancers including melanoma. Recent identification of the BMP ligand GDF6 as a novel melanoma oncogene showed GDF6-activated BMP signaling suppresses differentiation of melanoma cells. Previous studies have identified roles for GDF6 orthologs during early embryonic and neural crest development, but have not identified direct regulation of melanocyte development by GDF6. Here, we investigate the BMP ligand gdf6a, a zebrafish ortholog of human GDF6, during the development of melanocytes from the neural crest. We establish that the loss of gdf6a or inhibition of BMP signaling during neural crest development disrupts normal pigment cell development, leading to an increase in the number of melanocytes and a corresponding decrease in iridophores, another neural crest-derived pigment cell type in zebrafish. This shift occurs as pigment cells arise from the neural crest and depends on mitfa, an ortholog of MITF, a key regulator of melanocyte development that is also targeted by oncogenic BMP signaling. Together, these results indicate that the oncogenic role ligand-dependent BMP signaling plays in suppressing differentiation in melanoma is a reiteration of its physiological roles during melanocyte development.
Collapse
Affiliation(s)
- Alec K Gramann
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Department of Molecular Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| | - Arvind M Venkatesan
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Department of Molecular Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| | - Melissa Guerin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Department of Molecular Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| | - Craig J Ceol
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Department of Molecular Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
18
|
Lewis VM, Saunders LM, Larson TA, Bain EJ, Sturiale SL, Gur D, Chowdhury S, Flynn JD, Allen MC, Deheyn DD, Lee JC, Simon JA, Lippincott-Schwartz J, Raible DW, Parichy DM. Fate plasticity and reprogramming in genetically distinct populations of Danio leucophores. Proc Natl Acad Sci U S A 2019; 116:11806-11811. [PMID: 31138706 PMCID: PMC6575160 DOI: 10.1073/pnas.1901021116] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Understanding genetic and cellular bases of adult form remains a fundamental goal at the intersection of developmental and evolutionary biology. The skin pigment cells of vertebrates, derived from embryonic neural crest, are a useful system for elucidating mechanisms of fate specification, pattern formation, and how particular phenotypes impact organismal behavior and ecology. In a survey of Danio fishes, including the zebrafish Danio rerio, we identified two populations of white pigment cells-leucophores-one of which arises by transdifferentiation of adult melanophores and another of which develops from a yellow-orange xanthophore or xanthophore-like progenitor. Single-cell transcriptomic, mutational, chemical, and ultrastructural analyses of zebrafish leucophores revealed cell-type-specific chemical compositions, organelle configurations, and genetic requirements. At the organismal level, we identified distinct physiological responses of leucophores during environmental background matching, and we showed that leucophore complement influences behavior. Together, our studies reveal independently arisen pigment cell types and mechanisms of fate acquisition in zebrafish and illustrate how concerted analyses across hierarchical levels can provide insights into phenotypes and their evolution.
Collapse
Affiliation(s)
- Victor M Lewis
- Department of Biology, University of Virginia, Charlottesville, VA 22903
- Department of Biology, University of Washington, Seattle, WA 98195
| | - Lauren M Saunders
- Department of Biology, University of Virginia, Charlottesville, VA 22903
- Department of Genome Sciences, University of Washington, Seattle, WA 98195
- Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
| | - Tracy A Larson
- Department of Biology, University of Virginia, Charlottesville, VA 22903
| | - Emily J Bain
- Department of Biology, University of Virginia, Charlottesville, VA 22903
| | | | - Dvir Gur
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
- Eunice Kennedy Shriver National Institute for Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Sarwat Chowdhury
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Jessica D Flynn
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michael C Allen
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92093
| | - Dimitri D Deheyn
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92093
| | - Jennifer C Lee
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Julian A Simon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | | | - David W Raible
- Department of Biology, University of Washington, Seattle, WA 98195
- Department of Biological Structure, University of Washington, Seattle, WA 98195
| | - David M Parichy
- Department of Biology, University of Virginia, Charlottesville, VA 22903;
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
19
|
Abstract
In this review, Goding and Arnheiter present the current understanding of MITF's role and regulation in development and disease and highlight key areas where our knowledge of MITF regulation and function is limited. All transcription factors are equal, but some are more equal than others. In the 25 yr since the gene encoding the microphthalmia-associated transcription factor (MITF) was first isolated, MITF has emerged as a key coordinator of many aspects of melanocyte and melanoma biology. Like all transcription factors, MITF binds to specific DNA sequences and up-regulates or down-regulates its target genes. What marks MITF as being remarkable among its peers is the sheer range of biological processes that it appears to coordinate. These include cell survival, differentiation, proliferation, invasion, senescence, metabolism, and DNA damage repair. In this article we present our current understanding of MITF's role and regulation in development and disease, as well as those of the MITF-related factors TFEB and TFE3, and highlight key areas where our knowledge of MITF regulation and function is limited.
Collapse
Affiliation(s)
- Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Heinz Arnheiter
- National Institute of Neurological Disorders and Stroke, National Institutes of Heath, Bethesda, Maryland 20824, USA
| |
Collapse
|
20
|
Inter-chromosomal coupling between vision and pigmentation genes during genomic divergence. Nat Ecol Evol 2019; 3:657-667. [PMID: 30833758 DOI: 10.1038/s41559-019-0814-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/11/2019] [Indexed: 12/30/2022]
Abstract
Recombination between loci underlying mate choice and ecological traits is a major evolutionary force acting against speciation with gene flow. The evolution of linkage disequilibrium between such loci is therefore a fundamental step in the origin of species. Here, we show that this process can take place in the absence of physical linkage in hamlets-a group of closely related reef fishes from the wider Caribbean that differ essentially in colour pattern and are reproductively isolated through strong visually-based assortative mating. Using full-genome analysis, we identify four narrow genomic intervals that are consistently differentiated among sympatric species in a backdrop of extremely low genomic divergence. These four intervals include genes involved in pigmentation (sox10), axial patterning (hoxc13a), photoreceptor development (casz1) and visual sensitivity (SWS and LWS opsins) that develop islands of long-distance and inter-chromosomal linkage disequilibrium as species diverge. The relatively simple genomic architecture of species differences facilitates the evolution of linkage disequilibrium in the presence of gene flow.
Collapse
|
21
|
Qian L, Qi S, Cao F, Zhang J, Li C, Song M, Wang C. Effects of penthiopyrad on the development and behaviour of zebrafish in early-life stages. CHEMOSPHERE 2019; 214:184-194. [PMID: 30265925 DOI: 10.1016/j.chemosphere.2018.09.117] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 06/08/2023]
Abstract
The agricultural use of succinate dehydrogenase inhibitor (SDHI) fungicides has increased dramatically in the US and Europe. As the SDHI fungicides, boscalid, flutolanil and thifluzamide had been reported to induce a series of toxic effects on zebrafish. However, the toxic effects of penthiopyrad on zebrafish have not been reported yet. This study aimed to assess the acute toxicity of penthiopyrad to zebrafish in early-life stages and investigate behavioural response of larvae and the effects on lipid metabolism and pigmentation under sub-lethal exposure of penthiopyrad. Based on results of the acute toxicity tests of zebrafish embryo and larvae, penthiopyrad had an acute toxicity to early-life stages of zebrafish and induced a series of deformities during development. Based on the results of sub-lethal exposure for 8 days, penthiopyrad resulted in significant decreases in swimming velocity, acceleration speed, distance moved and inactive time of larvae at 0.3, 0.6 and 1.2 mg/L. Penthiopyrad induced the disorders of lipid metabolism via affecting fatty acid synthesis and β-oxidation, in accordance with remarkable changes in the content of triglycerides and cholesterol and the expression of key genes (hmgcrα, pparα1, srebf1, cyp51 and acca1) at 1.2 mg/L. In addition, the disorder of melanin synthesis and distribution was caused by penthiopyrad in larvae in accordance with changes in body colour and related gene expression at 8 dpe.
Collapse
Affiliation(s)
- Le Qian
- College of Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Suzhen Qi
- Risk Assessment Laboratory for Bee Product Quality and Safety of Ministry of Agriculture, Institute of Agricultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, People's Republic of China
| | - Fangjie Cao
- College of Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jie Zhang
- College of Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Changping Li
- Plant Protection Station, Beijing, People's Republic of China
| | - Min Song
- Institute of Agricultural Research, Taian, Shandong, People's Republic of China
| | - Chengju Wang
- College of Sciences, China Agricultural University, Beijing, People's Republic of China.
| |
Collapse
|
22
|
Liu L, Fu M, Pei S, Zhou L, Shang J. R-Fluoxetine Increases Melanin Synthesis Through a 5-HT1A/2A Receptor and p38 MAPK Signaling Pathways. Int J Mol Sci 2018; 20:ijms20010080. [PMID: 30585252 PMCID: PMC6337216 DOI: 10.3390/ijms20010080] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 12/20/2018] [Accepted: 12/20/2018] [Indexed: 12/02/2022] Open
Abstract
Fluoxetine, a member of the class of selective serotonin reuptake inhibitors, is a racemic mixture and has an anxiolytic effect in rodents. Previously, we have shown that fluoxetine can up-regulate melanin synthesis in B16F10 melanoma cells and normal human melanocytes (NMHM). However, the role of r-fluoxetine and s-fluoxetine, in the regulation of melanin synthesis, is still unknown. Here, we show how r-fluoxetine plays a critical role in fluoxetine enhancing melanogenesis, both in vivo and vitro, by up-regulating tyrosinase (TYR) and the microphthalmia-associated transcription factor (MITF) expression, whereas, s-fluoxetine does not show any effect in the vivo and vitro systems. In addition, we found that r-fluoxetine induced melanin synthesis through the serotonin1A receptor (5-HT1A) and serotonin 2A receptor (5-HT2A). Furthermore, r-fluoxetine increased the phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK), without affecting the phosphorylation of extracellularly responsive kinase (ERK1/2) and c-Jun N-terminal kinase (JNK). These data suggest that r-fluoxetine may be used as a drug for skin hypopigmentation disorders.
Collapse
Affiliation(s)
- Li Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 210009, China.
| | - Mengsi Fu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 210009, China.
| | - Siran Pei
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 210009, China.
| | - Liangliang Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 210009, China.
| | - Jing Shang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
23
|
Petratou K, Subkhankulova T, Lister JA, Rocco A, Schwetlick H, Kelsh RN. A systems biology approach uncovers the core gene regulatory network governing iridophore fate choice from the neural crest. PLoS Genet 2018; 14:e1007402. [PMID: 30286071 PMCID: PMC6191144 DOI: 10.1371/journal.pgen.1007402] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 10/16/2018] [Accepted: 08/22/2018] [Indexed: 12/29/2022] Open
Abstract
Multipotent neural crest (NC) progenitors generate an astonishing array of derivatives, including neuronal, skeletal components and pigment cells (chromatophores), but the molecular mechanisms allowing balanced selection of each fate remain unknown. In zebrafish, melanocytes, iridophores and xanthophores, the three chromatophore lineages, are thought to share progenitors and so lend themselves to investigating the complex gene regulatory networks (GRNs) underlying fate segregation of NC progenitors. Although the core GRN governing melanocyte specification has been previously established, those guiding iridophore and xanthophore development remain elusive. Here we focus on the iridophore GRN, where mutant phenotypes identify the transcription factors Sox10, Tfec and Mitfa and the receptor tyrosine kinase, Ltk, as key players. Here we present expression data, as well as loss and gain of function results, guiding the derivation of an initial iridophore specification GRN. Moreover, we use an iterative process of mathematical modelling, supplemented with a Monte Carlo screening algorithm suited to the qualitative nature of the experimental data, to allow for rigorous predictive exploration of the GRN dynamics. Predictions were experimentally evaluated and testable hypotheses were derived to construct an improved version of the GRN, which we showed produced outputs consistent with experimentally observed gene expression dynamics. Our study reveals multiple important regulatory features, notably a sox10-dependent positive feedback loop between tfec and ltk driving iridophore specification; the molecular basis of sox10 maintenance throughout iridophore development; and the cooperation between sox10 and tfec in driving expression of pnp4a, a key differentiation gene. We also assess a candidate repressor of mitfa, a melanocyte-specific target of sox10. Surprisingly, our data challenge the reported role of Foxd3, an established mitfa repressor, in iridophore regulation. Our study builds upon our previous systems biology approach, by incorporating physiologically-relevant parameter values and rigorous evaluation of parameter values within a qualitative data framework, to establish for the first time the core GRN guiding specification of the iridophore lineage.
Collapse
Affiliation(s)
- Kleio Petratou
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, Faculty of Science, University of Bath, Bath, United Kingdom
| | - Tatiana Subkhankulova
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, Faculty of Science, University of Bath, Bath, United Kingdom
| | - James A. Lister
- Department of Human and Molecular Genetics and Massey Cancer Center, VCU School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Andrea Rocco
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Hartmut Schwetlick
- Department of Mathematical Sciences, Faculty of Science, University of Bath, Bath, United Kingdom
| | - Robert N. Kelsh
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, Faculty of Science, University of Bath, Bath, United Kingdom
| |
Collapse
|
24
|
Cooper CD, Erickson SD, Yin S, Moravec T, Peh B, Curran K. Protein Kinase A Signaling Inhibits Iridophore Differentiation in Zebrafish. J Dev Biol 2018; 6:jdb6040023. [PMID: 30261583 PMCID: PMC6315511 DOI: 10.3390/jdb6040023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/08/2018] [Accepted: 09/14/2018] [Indexed: 12/31/2022] Open
Abstract
In zebrafish (Danio rerio), iridophores are specified from neural crest cells and represent a tractable system for examining mechanisms of cell fate and differentiation. Using this system, we have investigated the role of cAMP protein kinase A (PKA) signaling in pigment cell differentiation. Activation of PKA with the adenylyl cyclase activator forskolin reduces the number of differentiated iridophores in wildtype larvae, with insignificant changes to melanophore number. Inhibition of PKA with H89 significantly increases iridophore number, supporting a specific role for PKA during iridophore development. To determine the effects of altering PKA activity on iridophore and melanophore gene expression, we examined expression of iridophore marker pnp4a, melanophore marker mitfa, and the mitfa repressor foxd3. Consistent with our cell counts, forskolin significantly decreased pnp4a expression as detected by in situ hybridization and quantification of pnp4a+ cells. Forskolin had the opposite effect on mitfa and foxd3 gene activity, increasing the area of expression. As mitfa/nacre mutants have extra iridophores as compared to wildtype larvae, we examined the function of mitfa during PKA-sensitive iridophore development. Forskolin treatment of mitfa/nacre mutants did significantly reduce the number of iridophores but to a lesser extent than that observed in treated wildtype larvae. Taken together, our data suggests that PKA inhibits iridophore development in a subset of iridophore precursors, potentially via a foxd3-independent pathway.
Collapse
Affiliation(s)
- Cynthia D Cooper
- School of Molecular Biosciences, Washington State University Vancouver, Vancouver, WA 98686, USA.
- College of Arts and Sciences, Washington State University Vancouver, Vancouver, WA 98686, USA.
| | - Steve D Erickson
- College of Arts and Sciences, Washington State University Vancouver, Vancouver, WA 98686, USA.
| | - Scott Yin
- College of Arts and Sciences, Washington State University Vancouver, Vancouver, WA 98686, USA.
| | - Trevor Moravec
- College of Arts and Sciences, Washington State University Vancouver, Vancouver, WA 98686, USA.
| | - Brian Peh
- College of Arts and Sciences, Washington State University Vancouver, Vancouver, WA 98686, USA.
| | - Kevin Curran
- Department of Biology, University of San Diego, San Diego, CA 92110, USA.
| |
Collapse
|
25
|
Zhang YM, Zimmer MA, Guardia T, Callahan SJ, Mondal C, Di Martino J, Takagi T, Fennell M, Garippa R, Campbell NR, Bravo-Cordero JJ, White RM. Distant Insulin Signaling Regulates Vertebrate Pigmentation through the Sheddase Bace2. Dev Cell 2018; 45:580-594.e7. [PMID: 29804876 PMCID: PMC5991976 DOI: 10.1016/j.devcel.2018.04.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 03/07/2018] [Accepted: 04/27/2018] [Indexed: 11/15/2022]
Abstract
Patterning of vertebrate melanophores is essential for mate selection and protection from UV-induced damage. Patterning can be influenced by circulating long-range factors, such as hormones, but it is unclear how their activity is controlled in recipient cells to prevent excesses in cell number and migration. The zebrafish wanderlust mutant harbors a mutation in the sheddase bace2 and exhibits hyperdendritic and hyperproliferative melanophores that localize to aberrant sites. We performed a chemical screen to identify suppressors of the wanderlust phenotype and found that inhibition of insulin/PI3Kγ/mTOR signaling rescues the defect. In normal physiology, Bace2 cleaves the insulin receptor, whereas its loss results in hyperactive insulin/PI3K/mTOR signaling. Insulin B, an isoform enriched in the head, drives the melanophore defect. These results suggest that insulin signaling is negatively regulated by melanophore-specific expression of a sheddase, highlighting how long-distance factors can be regulated in a cell-type-specific manner.
Collapse
Affiliation(s)
- Yan M Zhang
- Weill Cornell Graduate School of Medical Sciences, Cell and Developmental Biology Program, New York, NY 10065, USA; Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA
| | - Milena A Zimmer
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA
| | - Talia Guardia
- University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Scott J Callahan
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA; Memorial Sloan Kettering Cancer Center, Gerstner Graduate School of Biomedical Sciences, New York, NY 10065, USA
| | - Chandrani Mondal
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Julie Di Martino
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Toshimitsu Takagi
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA
| | - Myles Fennell
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA
| | - Ralph Garippa
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA
| | - Nathaniel R Campbell
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & Genetics, New York, NY 10065, USA.
| |
Collapse
|
26
|
Kapp FG, Perlin JR, Hagedorn EJ, Gansner JM, Schwarz DE, O'Connell LA, Johnson NS, Amemiya C, Fisher DE, Wölfle U, Trompouki E, Niemeyer CM, Driever W, Zon LI. Protection from UV light is an evolutionarily conserved feature of the haematopoietic niche. Nature 2018; 558:445-448. [PMID: 29899448 PMCID: PMC6093292 DOI: 10.1038/s41586-018-0213-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 05/15/2018] [Indexed: 11/09/2022]
Abstract
Haematopoietic stem and progenitor cells (HSPCs) require a specific microenvironment, the haematopoietic niche, which regulates HSPC behaviour1,2. The location of this niche varies across species, but the evolutionary pressures that drive HSPCs to different microenvironments remain unknown. The niche is located in the bone marrow in adult mammals, whereas it is found in other locations in non-mammalian vertebrates, for example, in the kidney marrow in teleost fish. Here we show that a melanocyte umbrella above the kidney marrow protects HSPCs against ultraviolet light in zebrafish. Because mutants that lack melanocytes have normal steady-state haematopoiesis under standard laboratory conditions, we hypothesized that melanocytes above the stem cell niche protect HSPCs against ultraviolet-light-induced DNA damage. Indeed, after ultraviolet-light irradiation, unpigmented larvae show higher levels of DNA damage in HSPCs, as indicated by staining of cyclobutane pyrimidine dimers and have reduced numbers of HSPCs, as shown by cmyb (also known as myb) expression. The umbrella of melanocytes associated with the haematopoietic niche is highly evolutionarily conserved in aquatic animals, including the sea lamprey, a basal vertebrate. During the transition from an aquatic to a terrestrial environment, HSPCs relocated into the bone marrow, which is protected from ultraviolet light by the cortical bone around the marrow. Our studies reveal that melanocytes above the haematopoietic niche protect HSPCs from ultraviolet-light-induced DNA damage in aquatic vertebrates and suggest that during the transition to terrestrial life, ultraviolet light was an evolutionary pressure affecting the location of the haematopoietic niche.
Collapse
Affiliation(s)
- Friedrich G Kapp
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julie R Perlin
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Elliott J Hagedorn
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - John M Gansner
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel E Schwarz
- US Fish and Wildlife Service, Private John Allen National Fish Hatchery, Tupelo, MS, USA
| | | | - Nicholas S Johnson
- US Geological Survey, Great Lakes Science Center, Hammond Bay Biological Station, Millersburg, MI, USA
| | - Chris Amemiya
- Molecular Cell Biology, University of California, Merced, CA, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ute Wölfle
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Eirini Trompouki
- Department of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Charlotte M Niemeyer
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Driever
- Developmental Biology, Faculty of Biology, Centre for Biological Signalling Studies (BIOSS), Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Leonard I Zon
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Lajis AFB. A Zebrafish Embryo as an Animal Model for the Treatment of Hyperpigmentation in Cosmetic Dermatology Medicine. ACTA ACUST UNITED AC 2018; 54:medicina54030035. [PMID: 30344266 PMCID: PMC6122095 DOI: 10.3390/medicina54030035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 12/18/2022]
Abstract
For years, clinical studies involving human volunteers and several known pre-clinical in vivo models (i.e., mice, guinea pigs) have demonstrated their reliability in evaluating the effectiveness of a number of depigmenting agents. Although these models have great advantages, they also suffer from several drawbacks, especially involving ethical issues regarding experimentation. At present, a new depigmenting model using zebrafish has been proposed and demonstrated. The application of this model for screening and studying the depigmenting activity of many bioactive compounds has been given great attention in genetics, medicinal chemistry and even the cosmetic industry. Depigmenting studies using this model have been recognized as noteworthy approaches to investigating the antimelanogenic activity of bioactive compounds in vivo. This article details the current knowledge of zebrafish pigmentation and its reliability as a model for the screening and development of depigmenting agents. Several methods to quantify the antimelanogenic activity of bioactive compounds in this model, such as phenotype-based screening, melanin content, tyrosinase inhibitory activity, other related proteins and transcription genes, are reviewed. Depigmenting activity of several bioactive compounds which have been reported towards this model are compared in terms of their molecular structure and possible mode of actions. This includes patented materials with regard to the application of zebrafish as a depigmenting model, in order to give an insight of its intellectual value. At the end of this article, some limitations are highlighted and several recommendations are suggested for improvement of future studies.
Collapse
Affiliation(s)
- Ahmad Firdaus B Lajis
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Laboratory of Molecular Medicine, Institute of Bioscience, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Research Center, Faculty of Biotechnology and Biomolecular Sciences, University Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
28
|
Tian X, Pang X, Wang L, Li M, Dong C, Ma X, Wang L, Song D, Feng J, Xu P, Li X. Dynamic regulation of mRNA and miRNA associated with the developmental stages of skin pigmentation in Japanese ornamental carp. Gene 2018; 666:32-43. [PMID: 29684491 DOI: 10.1016/j.gene.2018.04.054] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/24/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022]
Abstract
The Japanese ornamental carp (Cyprinus carpio var. Koi) is famous for multifarious colors and patterns, making it commonly culture and trade across the world. Although functional genes and inheritance of color traits have been commonly studied, seldom attentions were focused on the genetic regulation during the developmental process of pigmentation. To better understand the mechanism of skin color development, we observed the morphogenesis of pigment cells during the post-embryonic stages and analysed the temporal expression pattern of mRNAs/miRNAs profiles in four distinct developmental stages. 59 and 103 differentially expressed genes/miRNAs (DEGs/DEMs) associated with pigmentation and skin were identified, including pax7, mitf, tyr, tyrp1, etc., and the highest DEGs were detected at 11 days post hatching (dph). In addition, the functional characteristics of mRNAs/miRNAs associated with pteridine and carotenoid pathway were also examined. Furthermore, 65 miRNA-mRNA interaction pairs related to pigmentation, pteridines and carotenoids metabolism were detected between different stages. Interestingly, the largest pairs appeared in the transition from 11 dph to 48 dph, which had the similar trend with DEGs further manifesting the importance of 11 dph. This study produced a comprehensive programme of DEGs/DEMs during color development, which will provide resources to understand the regulation mechanism in color formation. The understanding of genetic basis in color formation might promote the production and breeding of the Koi carp.
Collapse
Affiliation(s)
- Xue Tian
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Xiaolei Pang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Liangyan Wang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Mengrong Li
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Chuanju Dong
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Xiao Ma
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Lei Wang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Dongying Song
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China
| | - Jianxin Feng
- Henan Academy of Fishery Science, Zhengzhou, 410100, PR China
| | - Peng Xu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361005, PR China
| | - Xuejun Li
- College of Fisheries, Henan Normal University, Xinxiang, 453007, PR China.
| |
Collapse
|
29
|
Mouti MA, Dee C, Coupland SE, Hurlstone AFL. Minimal contribution of ERK1/2-MAPK signalling towards the maintenance of oncogenic GNAQQ209P-driven uveal melanomas in zebrafish. Oncotarget 2018; 7:39654-39670. [PMID: 27166257 PMCID: PMC5129960 DOI: 10.18632/oncotarget.9207] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/18/2016] [Indexed: 02/03/2023] Open
Abstract
Mutations affecting Gαq proteins are pervasive in uveal melanoma (UM), suggesting they ‘drive’ UM pathogenesis. The ERK1/2-MAPK pathway is critical for cutaneous melanoma development and consequently an important therapeutic target. Defining the contribution of ERK1/2-MAPK signalling to UM development has been hampered by the lack of an informative animal model that spontaneously develops UM. Towards this end, we engineered transgenic zebrafish to express oncogenic GNAQQ209P in the melanocyte lineage. This resulted in hyperplasia of uveal melanocytes, but with no evidence of malignant progression, nor perturbation of skin melanocytes. Combining expression of oncogenic GNAQQ209P with p53 inactivation resulted in earlier onset and even more extensive hyperplasia of uveal melanocytes that progressed to UM. Immunohistochemistry revealed only weak immunoreactivity to phosphorylated (p)ERK1/2 in established uveal tumours—in contrast to strong immunoreactivity in oncogenic RAS-driven skin lesions—but ubiquitous positive staining for nuclear Yes-associated protein (YAP). Moreover, no changes were observed in pERK1/2 levels upon transient knockdown of GNAQ or phospholipase C-beta (PLC-β) inhibition in the majority of human UM cell lines we tested harbouring GNAQ mutations. In summary, our findings demonstrate a weak correlation between oncogenic GNAQQ209P mutation and sustained ERK1/2-MAPK activation, implying that ERK1/2 signalling is unlikely to be instrumental in the maintenance of GNAQQ209P-driven UMs.
Collapse
Affiliation(s)
- Mai Abdel Mouti
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom
| | - Christopher Dee
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom
| | - Sarah E Coupland
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Adam F L Hurlstone
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
30
|
Heffer A, Marquart GD, Aquilina-Beck A, Saleem N, Burgess HA, Dawid IB. Generation and characterization of Kctd15 mutations in zebrafish. PLoS One 2017; 12:e0189162. [PMID: 29216270 PMCID: PMC5720732 DOI: 10.1371/journal.pone.0189162] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023] Open
Abstract
Potassium channel tetramerization domain containing 15 (Kctd15) was previously found to have a role in early neural crest (NC) patterning, specifically delimiting the region where NC markers are expressed via repression of transcription factor AP-2a and inhibition of Wnt signaling. We used transcription activator-like effector nucleases (TALENs) to generate null mutations in zebrafish kctd15a and kctd15b paralogs to study the in vivo role of Kctd15. We found that while deletions producing frame-shift mutations in each paralog showed no apparent phenotype, kctd15a/b double mutant zebrafish are smaller in size and show several phenotypes including some affecting the NC, such as expansion of the early NC domain, increased pigmentation, and craniofacial defects. Both melanophore and xanthophore pigment cell numbers and early markers are up-regulated in the double mutants. While we find no embryonic craniofacial defects, adult mutants have a deformed maxillary segment and missing barbels. By confocal imaging of mutant larval brains we found that the torus lateralis (TLa), a region implicated in gustatory networks in other fish, is absent. Ablation of this brain tissue in wild type larvae mimics some aspects of the mutant growth phenotype. Thus kctd15 mutants show deficits in the development of both neural crest derivatives, and specific regions within the central nervous system, leading to a strong reduction in normal growth rates.
Collapse
Affiliation(s)
- Alison Heffer
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States of America
| | - Gregory D. Marquart
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States of America
| | - Allisan Aquilina-Beck
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States of America
| | - Nabil Saleem
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States of America
| | - Harold A. Burgess
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States of America
| | - Igor B. Dawid
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail:
| |
Collapse
|
31
|
Kratochwil CF, Sefton MM, Liang Y, Meyer A. Tol2 transposon-mediated transgenesis in the Midas cichlid (Amphilophus citrinellus) - towards understanding gene function and regulatory evolution in an ecological model system for rapid phenotypic diversification. BMC DEVELOPMENTAL BIOLOGY 2017; 17:15. [PMID: 29169323 PMCID: PMC5701313 DOI: 10.1186/s12861-017-0157-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/14/2017] [Indexed: 11/12/2022]
Abstract
Background The Midas cichlid species complex (Amphilophus spp.) is widely known among evolutionary biologists as a model system for sympatric speciation and adaptive phenotypic divergence within extremely short periods of time (a few hundred generations). The repeated parallel evolution of adaptive phenotypes in this radiation, combined with their near genetic identity, makes them an excellent model for studying phenotypic diversification. While many ecological and evolutionary studies have been performed on Midas cichlids, the molecular basis of specific phenotypes, particularly adaptations, and their underlying coding and cis-regulatory changes have not yet been studied thoroughly. Results For the first time in any New World cichlid, we use Tol2 transposon-mediated transgenesis in the Midas cichlid (Amphilophus citrinellus). By adapting existing microinjection protocols, we established an effective protocol for transgenesis in Midas cichlids. Embryos were injected with a Tol2 plasmid construct that drives enhanced green fluorescent protein (eGFP) expression under the control of the ubiquitin promoter. The transgene was successfully integrated into the germline, driving strong ubiquitous expression of eGFP in the first transgenic Midas cichlid line. Additionally, we show transient expression of two further transgenic constructs, ubiquitin::tdTomato and mitfa::eGFP. Transgenesis in Midas cichlids will facilitate further investigation of the genetic basis of species-specific traits, many of which are adaptations. Conclusion Transgenesis is a versatile tool not only for studying regulatory elements such as promoters and enhancers, but also for testing gene function through overexpression of allelic gene variants. As such, it is an important first step in establishing the Midas cichlid as a powerful model for studying adaptive coding and non-coding changes in an ecological and evolutionary context.
Collapse
Affiliation(s)
- Claudius F Kratochwil
- Chair in Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Constance, Germany. .,Zukunftskolleg, University of Konstanz, Constance, Germany. .,International Max Planck Research School for Organismal Biology (IMPRS), Max Planck Institute for Ornithology, Radolfzell, Germany.
| | - Maggie M Sefton
- Chair in Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Constance, Germany.,International Max Planck Research School for Organismal Biology (IMPRS), Max Planck Institute for Ornithology, Radolfzell, Germany
| | - Yipeng Liang
- Chair in Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Constance, Germany
| | - Axel Meyer
- Chair in Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Constance, Germany. .,International Max Planck Research School for Organismal Biology (IMPRS), Max Planck Institute for Ornithology, Radolfzell, Germany.
| |
Collapse
|
32
|
He X, Lindsay-Mosher N, Li Y, Molinaro AM, Pellettieri J, Pearson BJ. FOX and ETS family transcription factors regulate the pigment cell lineage in planarians. Development 2017; 144:4540-4551. [PMID: 29158443 DOI: 10.1242/dev.156349] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/03/2017] [Indexed: 12/16/2022]
Abstract
Many pigment cells acquire unique structural properties and gene expression profiles during animal development. The underlying differentiation pathways have been well characterized in cells formed during embryogenesis, such as the neural crest-derived melanocyte. However, much less is known about the developmental origins of pigment cells produced in adult organisms during tissue homeostasis and repair. Here we report a lineage analysis of ommochrome- and porphyrin-producing cells in the brown, freshwater planarian Schmidtea mediterranea Using an RNA-sequencing approach, we identified two classes of markers expressed in sequential fashion when new pigment cells are generated during regeneration or in response to pigment cell ablation. We also report roles for FOXF-1 and ETS-1 transcription factors, as well as for an FGFR-like molecule, in the specification and maintenance of this cell type. Together, our results provide insights into mechanisms of adult pigment cell development in the strikingly colorful Platyhelminthes phylum.
Collapse
Affiliation(s)
- Xinwen He
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, Ontario M5G0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G0A4, Canada
| | - Nicole Lindsay-Mosher
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, Ontario M5G0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G0A4, Canada
| | - Yan Li
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, Ontario M5G0A4, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario M5G0A4, Canada
| | - Alyssa M Molinaro
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, Ontario M5G0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G0A4, Canada
| | | | - Bret J Pearson
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, Ontario M5G0A4, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G0A4, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario M5G0A4, Canada
| |
Collapse
|
33
|
Zhang Y, Liu J, Fu W, Xu W, Zhang H, Chen S, Liu W, Peng L, Xiao Y. Comparative Transcriptome and DNA methylation analyses of the molecular mechanisms underlying skin color variations in Crucian carp (Carassius carassius L.). BMC Genet 2017; 18:95. [PMID: 29121864 PMCID: PMC5680753 DOI: 10.1186/s12863-017-0564-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 10/31/2017] [Indexed: 11/25/2022] Open
Abstract
Background Crucian carp is a popular ornamental strain in Asia with variants in body color. To further explore the genetic mechanisms underlying gray and red body color formation in crucian carp, the skin transcriptomes and partial DNA methylation sites were obtained from red crucian carp (RCC) and white crucian carp (WCC). Here, we show significant differences in mRNA expression and DNA methylation sites between skin tissues of RCC and WCC. Results Totals of 3434 and 3683 unigenes had significantly lower and higher expression in WCC, respectively, compared with unigenes expressed in RCC. Some potential genes for body color development were further identified by quantitative polymerase chain reaction, such as mitfa, tyr, tyrp1, and dct, which were down-regulated, and foxd3, hpda, ptps, and gch1, which were up-regulated. A KEGG pathway analysis indicated that the differentially expressed genes were mainly related to mitogen activated protein kinase (MAPK), Wnt, cell cycle, and endocytosis signaling pathways, as well as variations in melanogenesis in crucian carp. In addition, some differentially expressed DNA methylation site genes were related to pigmentation, including mitfa, tyr, dct, foxd3, and hpda. The differentially expressed DNA methylation sites were mainly involved in signaling pathways, including MAPK, cAMP, endocytosis, melanogenesis, and Hippo. Conclusions Our study provides the results of comparative transcriptome and DNA methylation analyses between RCC and WCC skin tissues and reveals that the molecular mechanism of body color variation in crucian carp is strongly related to disruptions in gene expression and DNA methylation during pigmentation. Electronic supplementary material The online version of this article (10.1186/s12863-017-0564-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yongqin Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Jinhui Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Wen Fu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Wenting Xu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Huiqin Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Shujuan Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Wenbin Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Liangyue Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Yamei Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China. .,School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.
| |
Collapse
|
34
|
Zhang Y, Liu J, Peng L, Ren L, Zhang H, Zou L, Liu W, Xiao Y. Comparative transcriptome analysis of molecular mechanism underlying gray-to-red body color formation in red crucian carp (Carassius auratus, red var.). FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:1387-1398. [PMID: 28676950 DOI: 10.1007/s10695-017-0379-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/24/2017] [Indexed: 06/07/2023]
Abstract
Red crucian carp (Carassius auratus red var.) is an ornamental fish with vivid red/orange color. It has been found that the adult body color of this strain forms a gray-to-red change. In this study, skin transcriptomes of red crucian carp are first obtained for three different stages of body color development, named by gray-color (GC), color-variation (CV), and red-color (RC) stages, respectively. From the skins of GC, CV, and RC, 103,229; 108,208; and 120,184 transcripts have been identified, respectively. Bioinformatics analysis reveals that 2483, 2967, and 4473 unigenes are differentially expressed between CV and GC, RC and CV, and RC and GC, respectively. A part of the differentially expressed genes (DEGs) are involved in the signaling pathway of pigment synthesis, such as the melanogenesis genes (Mitfa, Pax3a, Foxd3, Mc1r, Asip); tyrosine metabolism genes (Tyr, Dct, Tyrp1, Silva, Tat, Hpda); and pteridine metabolism genes (Gch, Xdh, Ptps, Tc). According to the data of transcriptome and quantitative PCR, the expression of Mitfa and its regulated genes which include the genes of Tyr, Tyrp1, Dct, Tfe3a, and Baxα, decreases with gray-to-red change. It is suggested that Mitfa and some genes, being related to melanin synthesis or melanophore development, are closely related to the gray-to-red body color transformation in the red crucian carp. Furthermore, the DEGs of cell apoptosis and autophagy pathway, such as Tfe3a, Baxα, Hsp70, Beclin1, Lc3, Atg9a, and Atg4a, might be involved in the melanocytes fade away of juvenile fish. These results shed light on the regulation mechanism of gray-to-red body color transformation in red crucian carp, and are helpful to the selective breeding of ornamental fish strains.
Collapse
Affiliation(s)
- Yongqin Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Jinhui Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Liangyue Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Li Ren
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Huiqin Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Lijun Zou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Wenbin Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.
| | - Yamei Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, 410081, China.
- School of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.
| |
Collapse
|
35
|
Ciarlo C, Kaufman CK, Kinikoglu B, Michael J, Yang S, D′Amato C, Blokzijl-Franke S, den Hertog J, Schlaeger TM, Zhou Y, Liao E, Zon LI. A chemical screen in zebrafish embryonic cells establishes that Akt activation is required for neural crest development. eLife 2017; 6:e29145. [PMID: 28832322 PMCID: PMC5599238 DOI: 10.7554/elife.29145] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/08/2017] [Indexed: 01/09/2023] Open
Abstract
The neural crest is a dynamic progenitor cell population that arises at the border of neural and non-neural ectoderm. The inductive roles of FGF, Wnt, and BMP at the neural plate border are well established, but the signals required for subsequent neural crest development remain poorly characterized. Here, we conducted a screen in primary zebrafish embryo cultures for chemicals that disrupt neural crest development, as read out by crestin:EGFP expression. We found that the natural product caffeic acid phenethyl ester (CAPE) disrupts neural crest gene expression, migration, and melanocytic differentiation by reducing Sox10 activity. CAPE inhibits FGF-stimulated PI3K/Akt signaling, and neural crest defects in CAPE-treated embryos are suppressed by constitutively active Akt1. Inhibition of Akt activity by constitutively active PTEN similarly decreases crestin expression and Sox10 activity. Our study has identified Akt as a novel intracellular pathway required for neural crest differentiation.
Collapse
Affiliation(s)
- Christie Ciarlo
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
- Harvard Medical SchoolBostonUnited States
| | - Charles K Kaufman
- Division of Oncology, Department of MedicineWashington University School of MedicineSt. LouisUnited States
- Department of Developmental BiologyWashington University School of MedicineSt. LouisUnited States
| | - Beste Kinikoglu
- Center for Regenerative MedicineMassachusetts General HospitalBostonUnited States
- Division of Plastic and Reconstructive SurgeryMassachusetts General HospitalBostonUnited States
| | - Jonathan Michael
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Song Yang
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Christopher D′Amato
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Sasja Blokzijl-Franke
- Hubrecht Institute, Koninklijke Nederlandse Akademie van WetenschappenUniversity Medical Center UtrechtUtrechtNetherlands
| | - Jeroen den Hertog
- Hubrecht Institute, Koninklijke Nederlandse Akademie van WetenschappenUniversity Medical Center UtrechtUtrechtNetherlands
| | - Thorsten M Schlaeger
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Yi Zhou
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Eric Liao
- Harvard Medical SchoolBostonUnited States
- Center for Regenerative MedicineMassachusetts General HospitalBostonUnited States
- Division of Plastic and Reconstructive SurgeryMassachusetts General HospitalBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| | - Leonard I Zon
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
- Harvard Medical SchoolBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| |
Collapse
|
36
|
Williams AL, Bohnsack BL. Multi-Photon Time Lapse Imaging to Visualize Development in Real-time: Visualization of Migrating Neural Crest Cells in Zebrafish Embryos. J Vis Exp 2017. [PMID: 28829423 DOI: 10.3791/56214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Congenital eye and craniofacial anomalies reflect disruptions in the neural crest, a transient population of migratory stem cells that give rise to numerous cell types throughout the body. Understanding the biology of the neural crest has been limited, reflecting a lack of genetically tractable models that can be studied in vivo and in real-time. Zebrafish is a particularly important developmental model for studying migratory cell populations, such as the neural crest. To examine neural crest migration into the developing eye, a combination of the advanced optical techniques of laser scanning microscopy with long wavelength multi-photon fluorescence excitation was implemented to capture high-resolution, three-dimensional, real-time videos of the developing eye in transgenic zebrafish embryos, namely Tg(sox10:EGFP) and Tg(foxd3:GFP), as sox10 and foxd3 have been shown in numerous animal models to regulate early neural crest differentiation and likely represent markers for neural crest cells. Multi-photon time-lapse imaging was used to discern the behavior and migratory patterns of two neural crest cell populations contributing to early eye development. This protocol provides information for generating time-lapse videos during zebrafish neural crest migration, as an example, and can be further applied to visualize the early development of many structures in the zebrafish and other model organisms.
Collapse
Affiliation(s)
- Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan;
| |
Collapse
|
37
|
Eason J, Williams AL, Chawla B, Apsey C, Bohnsack BL. Differences in neural crest sensitivity to ethanol account for the infrequency of anterior segment defects in the eye compared with craniofacial anomalies in a zebrafish model of fetal alcohol syndrome. Birth Defects Res 2017; 109:1212-1227. [PMID: 28681995 DOI: 10.1002/bdr2.1069] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/21/2017] [Accepted: 05/22/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Ethanol (ETOH) exposure during pregnancy is associated with craniofacial and neurologic abnormalities, but infrequently disrupts the anterior segment of the eye. In these studies, we used zebrafish to investigate differences in the teratogenic effect of ETOH on craniofacial, periocular, and ocular neural crest. METHODS Zebrafish eye and neural crest development was analyzed by means of live imaging, TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assay, immunostaining, detection of reactive oxygen species, and in situ hybridization. RESULTS Our studies demonstrated that foxd3-positive neural crest cells in the periocular mesenchyme and developing eye were less sensitive to ETOH than sox10-positive craniofacial neural crest cells that form the pharyngeal arches and jaw. ETOH increased apoptosis in the retina, but did not affect survival of periocular and ocular neural crest cells. ETOH also did not increase reactive oxygen species within the eye. In contrast, ETOH increased ventral neural crest apoptosis and reactive oxygen species production in the facial mesenchyme. In the eye and craniofacial region, sod2 showed high levels of expression in the anterior segment and in the setting of Sod2 knockdown, low levels of ETOH decreased migration of foxd3-positive neural crest cells into the developing eye. However, ETOH had minimal effect on the periocular and ocular expression of transcription factors (pitx2 and foxc1) that regulate anterior segment development. CONCLUSION Neural crest cells contributing to the anterior segment of the eye exhibit increased ability to withstand ETOH-induced oxidative stress and apoptosis. These studies explain the rarity of anterior segment dysgenesis despite the frequent craniofacial abnormalities in fetal alcohol syndrome. Birth Defects Research 109:1212-1227, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jessica Eason
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Bahaar Chawla
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Christian Apsey
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
38
|
Cechmanek PB, McFarlane S. Retinal pigment epithelium expansion around the neural retina occurs in two separate phases with distinct mechanisms. Dev Dyn 2017; 246:598-609. [PMID: 28556369 DOI: 10.1002/dvdy.24525] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/17/2017] [Accepted: 05/10/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The retinal pigment epithelium (RPE) is a specialized monolayer of epithelial cells that forms a tight barrier surrounding the neural retina. RPE cells are indispensable for mature photoreceptor renewal and survival, yet how the initial RPE cell population expands around the neural retina during eye development is poorly understood. RESULTS Here we characterize the differentiation, proliferation, and movements of RPE progenitors in the Zebrafish embryo over the period of optic cup morphogenesis. RPE progenitors are present in the dorsomedial eye vesicle shortly after eye vesicle evagination. We define two separate phases that allow for full RPE expansion. The first phase involves a previously uncharacterized antero-wards expansion of the RPE progenitor domain in the inner eye vesicle leaflet, driven largely by an increase in cell number. During this phase, RPE progenitors start to express differentiation markers. In the second phase, the progenitor domain stretches in the dorsoventral and posterior axes, involving cell movements and shape changes, and coinciding with optic cup morphogenesis. Significantly, cell division is not required for RPE expansion. CONCLUSIONS RPE development to produce the monolayer epithelium that covers the back of the neural retina occurs in two distinct phases driven by distinct mechanisms. Developmental Dynamics 246:598-609, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paula Bernice Cechmanek
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sarah McFarlane
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
39
|
Williams AL, Eason J, Chawla B, Bohnsack BL. Cyp1b1 Regulates Ocular Fissure Closure Through a Retinoic Acid-Independent Pathway. Invest Ophthalmol Vis Sci 2017; 58:1084-1097. [PMID: 28192799 PMCID: PMC5308778 DOI: 10.1167/iovs.16-20235] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Mutations in the CYP1B1 gene are the most commonly identified genetic causes of primary infantile-onset glaucoma. Despite this disease association, the role of CYP1B1 in eye development and its in vivo substrate remain unknown. In the present study, we used zebrafish to elucidate the mechanism by which cyp1b1 regulates eye development. Methods Zebrafish eye and neural crest development were analyzed using live imaging of transgenic zebrafish embryos, in situ hybridization, immunostaining, TUNEL assay, and methylacrylate sections. Cyp1b1 and retinoic acid (RA) levels were genetically (morpholino oligonucleotide antisense and mRNA) and pharmacologically manipulated to examine gene function. Results Using zebrafish, we observed that cyp1b1 was expressed in a specific spatiotemporal pattern in the ocular fissures of the developing zebrafish retina and regulated fissure patency. Decreased Cyp1b1 resulted in the premature breakdown of laminin in the ventral fissure and altered subsequent neural crest migration into the anterior segment. In contrast, cyp1b1 overexpression inhibited cell survival in the ventral ocular fissure and prevented fissure closure via an RA-independent pathway. Cyp1b1 overexpression also inhibited the ocular expression of vsx2, pax6a, and pax6b and increased the extraocular expression of shha. Importantly, embryos injected with human wild-type but not mutant CYP1B1 mRNA also showed colobomas, demonstrating the evolutionary and functional conservation of gene function between species. Conclusions Cyp1b1 regulation of ocular fissure closure indirectly affects neural crest migration and development through an RA-independent pathway. These studies provide insight into the role of Cyp1b1 in eye development and further elucidate the pathogenesis of primary infantile-onset glaucoma.
Collapse
Affiliation(s)
- Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Jessica Eason
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Bahaar Chawla
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
40
|
The master role of microphthalmia-associated transcription factor in melanocyte and melanoma biology. J Transl Med 2017; 97:649-656. [PMID: 28263292 DOI: 10.1038/labinvest.2017.9] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/07/2017] [Accepted: 01/10/2017] [Indexed: 12/20/2022] Open
Abstract
Certain transcription factors have vital roles in lineage development, including specification of cell types and control of differentiation. Microphthalmia-associated transcription factor (MITF) is a key transcription factor for melanocyte development and differentiation. MITF regulates expression of numerous pigmentation genes to promote melanocyte differentiation, as well as fundamental genes for maintaining cell homeostasis, including genes encoding proteins involved in apoptosis (eg, BCL2) and the cell cycle (eg, CDK2). Loss-of-function mutations of MITF cause Waardenburg syndrome type IIA, whose phenotypes include depigmentation due to melanocyte loss, whereas amplification or specific mutation of MITF can be an oncogenic event that is seen in a subset of familial or sporadic melanomas. In this article, we review basic features of MITF biological function and highlight key unresolved questions regarding this remarkable transcription factor.
Collapse
|
41
|
Xu L, Xu QH, Zhou XY, Yin LY, Guan PP, Zhang T, Liu JX. Mechanisms of silver_nanoparticles induced hypopigmentation in embryonic zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 184:49-60. [PMID: 28104549 DOI: 10.1016/j.aquatox.2017.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/07/2017] [Accepted: 01/09/2017] [Indexed: 06/06/2023]
Abstract
Silver_nanoparticles (AgNPs) have been reported to inhibit specification of erythroid cells and to induce spinal cord deformities and cardiac arrhythmia in vertebrates, but have not been implicated in development of neural crest (NC) and pigment cells in an in vivo model yet. In current study, down-regulated expressions of NC genes pax7 and foxd3, melanophore genes mitfa and dct, and xanthophore gene gch2 in AgNPs-exposed embryos were revealed by microarray, qRT-PCR and whole-mount in situ hybridization (WISH). Then, the down-regulated expressions of melanophore genes mitfa and dct but not xanthophore gene gch2 in AgNPs-exposed embryos were found to be recovered by melanogenesis agonists palmitic acid and dibutyryl cyclic AMP (dbcAMP). Finally, Ag+ chelating and AgNPs coating compound l-cysteine was found to neutralize AgNPs-induced hypopigmentation in AgNPs-exposed embryos, and to recover the down-regulated expressions of both dct and gch2 to nearly normal level in embryos, suggesting that AgNPs-releasing Ag+ might mediate their biological effects on zebrafish pigmentation mostly. This study was firstly to unveil that AgNPs might specifically act up-stream of mitfa and pax7 genes to suppress specification and differentiation of melanophore and xanthophore lineages respectively by their releasing Ag+ during vertebrate embryogenesis.
Collapse
Affiliation(s)
- Lian Xu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Qin-Han Xu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Xin-Ying Zhou
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Li-Yan Yin
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources, Hainan University, HaiKou, 570228, China.
| | - Peng-Peng Guan
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Ting Zhang
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China; Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Hunan, Changde, 415000, China.
| |
Collapse
|
42
|
Seberg HE, Van Otterloo E, Loftus SK, Liu H, Bonde G, Sompallae R, Gildea DE, Santana JF, Manak JR, Pavan WJ, Williams T, Cornell RA. TFAP2 paralogs regulate melanocyte differentiation in parallel with MITF. PLoS Genet 2017; 13:e1006636. [PMID: 28249010 PMCID: PMC5352137 DOI: 10.1371/journal.pgen.1006636] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/15/2017] [Accepted: 02/14/2017] [Indexed: 12/20/2022] Open
Abstract
Mutations in the gene encoding transcription factor TFAP2A result in pigmentation anomalies in model organisms and premature hair graying in humans. However, the pleiotropic functions of TFAP2A and its redundantly-acting paralogs have made the precise contribution of TFAP2-type activity to melanocyte differentiation unclear. Defining this contribution may help to explain why TFAP2A expression is reduced in advanced-stage melanoma compared to benign nevi. To identify genes with TFAP2A-dependent expression in melanocytes, we profile zebrafish tissue and mouse melanocytes deficient in Tfap2a, and find that expression of a small subset of genes underlying pigmentation phenotypes is TFAP2A-dependent, including Dct, Mc1r, Mlph, and Pmel. We then conduct TFAP2A ChIP-seq in mouse and human melanocytes and find that a much larger subset of pigmentation genes is associated with active regulatory elements bound by TFAP2A. These elements are also frequently bound by MITF, which is considered the "master regulator" of melanocyte development. For example, the promoter of TRPM1 is bound by both TFAP2A and MITF, and we show that the activity of a minimal TRPM1 promoter is lost upon deletion of the TFAP2A binding sites. However, the expression of Trpm1 is not TFAP2A-dependent, implying that additional TFAP2 paralogs function redundantly to drive melanocyte differentiation, which is consistent with previous results from zebrafish. Paralogs Tfap2a and Tfap2b are both expressed in mouse melanocytes, and we show that mouse embryos with Wnt1-Cre-mediated deletion of Tfap2a and Tfap2b in the neural crest almost completely lack melanocytes but retain neural crest-derived sensory ganglia. These results suggest that TFAP2 paralogs, like MITF, are also necessary for induction of the melanocyte lineage. Finally, we observe a genetic interaction between tfap2a and mitfa in zebrafish, but find that artificially elevating expression of tfap2a does not increase levels of melanin in mitfa hypomorphic or loss-of-function mutants. Collectively, these results show that TFAP2 paralogs, operating alongside lineage-specific transcription factors such as MITF, directly regulate effectors of terminal differentiation in melanocytes. In addition, they suggest that TFAP2A activity, like MITF activity, has the potential to modulate the phenotype of melanoma cells.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites/genetics
- Cell Differentiation/genetics
- Cell Line
- Cell Line, Tumor
- Cells, Cultured
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Gene Expression Profiling/methods
- Gene Expression Regulation, Developmental
- Humans
- Melanocytes/metabolism
- Mice, Knockout
- Microphthalmia-Associated Transcription Factor/genetics
- Microphthalmia-Associated Transcription Factor/metabolism
- Microscopy, Confocal
- Mutation
- Pigmentation/genetics
- RNA Interference
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Homology, Nucleic Acid
- Transcription Factor AP-2/genetics
- Transcription Factor AP-2/metabolism
- Zebrafish
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Hannah E. Seberg
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America
| | - Eric Van Otterloo
- SDM-Craniofacial Biology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Stacie K. Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Huan Liu
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Greg Bonde
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Ramakrishna Sompallae
- Bioinformatics Division, Iowa Institute of Human Genetics, University of Iowa, Iowa City, Iowa, United States of America
| | - Derek E. Gildea
- Bioinformatics and Scientific Programming Core, Computational and Statistical Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Juan F. Santana
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - J. Robert Manak
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - William J. Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Trevor Williams
- SDM-Craniofacial Biology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Robert A. Cornell
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
43
|
Zhang Y, Wang Z, Xiao H, Liu X, Zhu G, Yu D, Han G, Chen G, Hou C, Ma N, Shen B, Li Y, Wang T, Wang R. Foxd3 suppresses interleukin-10 expression in B cells. Immunology 2017; 150:478-488. [PMID: 27995618 DOI: 10.1111/imm.12701] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 12/22/2022] Open
Abstract
Interleukin-10-positive (IL-10+ ) regulatory B (Breg) cells play an important role in restraining excessive inflammatory responses by secreting IL-10. However, it is still unclear what key transcription factors determine Breg cell differentiation. Hence, we explore what transcription factor plays a key role in the expression of IL-10, a pivotal cytokine in Breg cells. We used two types of web-based prediction software to predict transcription factors binding the IL-10 promoter and found that IL-10 promoter had many binding sites for Foxd3. Chromatin immunoprecipitation PCR assay demonstrated that Foxd3 directly binds the predicted binding sites around the start codon upstream by -1400 bp. Further, we found that Foxd3 suppressed the activation of IL-10 promoter by using an IL-10 promoter report system. Finally, knocking out Foxd3 effectively promotes Breg cell production by up-regulating IL-10 expression. Conversely, up-regulated Foxd3 expression was negatively associated with IL-10+ Breg cells in lupus-prone MRL/lpr mice. Hence, our data suggest that Foxd3 suppresses the production of IL-10+ Breg cells by directly binding the IL-10 promoter. This study demonstrates the mechanism for Breg cell production and its application to the treatment of autoimmune diseases by regulating Foxd3 expression.
Collapse
Affiliation(s)
- Yu Zhang
- College of Pharmacy, Henan University, Kaifeng, China.,Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Zhiding Wang
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China.,Department of Biomedicine, Institute of Frontier Medical Sciences, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - He Xiao
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Xiaoling Liu
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China.,Department of Nephrology, The 307th Hospital of Chinese People's Liberation Army, Beijing, China
| | - Gaizhi Zhu
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China.,Laboratory of Cellular and Molecular Immunology, Henan University, Kaifeng, Henan, China
| | - Dandan Yu
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Gencheng Han
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Guojiang Chen
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Chunmei Hou
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Ning Ma
- Department of Rheumatology, First Hospital of Jilin University, Changchun, China
| | - Beifen Shen
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Yan Li
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Tianxiao Wang
- College of Pharmacy, Henan University, Kaifeng, China
| | - Renxi Wang
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
44
|
Kelsh RN, Sosa KC, Owen JP, Yates CA. Zebrafish adult pigment stem cells are multipotent and form pigment cells by a progressive fate restriction process: Clonal analysis identifies shared origin of all pigment cell types. Bioessays 2016; 39. [PMID: 28009049 DOI: 10.1002/bies.201600234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Skin pigment pattern formation is a paradigmatic example of pattern formation. In zebrafish, the adult body stripes are generated by coordinated rearrangement of three distinct pigment cell-types, black melanocytes, shiny iridophores and yellow xanthophores. A stem cell origin of melanocytes and iridophores has been proposed although the potency of those stem cells has remained unclear. Xanthophores, however, seemed to originate predominantly from proliferation of embryonic xanthophores. Now, data from Singh et al. shows that all three cell-types derive from shared stem cells, and that these cells generate peripheral neural cell-types too. Furthermore, clonal compositions are best explained by a progressive fate restriction model generating the individual cell-types. The numbers of adult pigment stem cells associated with the dorsal root ganglia remain low, but progenitor numbers increase significantly during larval development up to metamorphosis, likely via production of partially restricted progenitors on the spinal nerves.
Collapse
Affiliation(s)
- Robert N Kelsh
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, UK
| | - Karen C Sosa
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, UK
| | - Jennifer P Owen
- Department of Mathematical Sciences and Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, UK
| | - Christian A Yates
- Department of Mathematical Sciences, University of Bath, Claverton Down, Bath, UK
| |
Collapse
|
45
|
Guillot R, Muriach B, Rocha A, Rotllant J, Kelsh RN, Cerdá-Reverter JM. Thyroid Hormones Regulate Zebrafish Melanogenesis in a Gender-Specific Manner. PLoS One 2016; 11:e0166152. [PMID: 27832141 PMCID: PMC5104317 DOI: 10.1371/journal.pone.0166152] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/24/2016] [Indexed: 11/18/2022] Open
Abstract
Zebrafish embryos are treated with anti-thyroidal compounds, such as phenylthiourea, to inhibit melanogenesis. However, the mechanism whereby the thyroidal system controls melanin synthesis has not been assessed in detail. In this work, we tested the effect of the administration of diets supplemented with T3 (500μg/g food) on the pigment pattern of adult zebrafish. Oral T3 induced a pronounced skin paling in both adult female and male zebrafish that was reversible upon cessation of treatment. The number of visible melanophores was significantly reduced in treated fish. Accordingly, treatment down-regulated expression of tyrosinase-related protein 1 in both sexes. We also found sexually dimorphic regulation of some melanogenic genes, such as Dct/Tyrp2 that was dramatically up-regulated in females after T3 treatment. Thus, we demonstrated that melanogenesis is reversibly inhibited by thyroid hormones in adult zebrafish and make the discovery of gender-specific differences in the response of melanogenic gene expression. Thus, fish gender is now shown to be an important variable that should be controlled in future studies of fish melanogenesis.
Collapse
Affiliation(s)
- Raúl Guillot
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre de la Sal, Consejo Superior de Investigaciones Científicas, (IATS-CSIC), Ribera de Cabanes, Castellón, Spain, 12595
| | - Borja Muriach
- Facultad Ciencias de la Salud, Universidad CEU Cardenal Herrera, Castellón, Spain, 12006
| | - Ana Rocha
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre de la Sal, Consejo Superior de Investigaciones Científicas, (IATS-CSIC), Ribera de Cabanes, Castellón, Spain, 12595
| | - Josep Rotllant
- Aquatic Molecular Pathobiology Group, Instituto de Investigaciones Marinas, Consejo Superior de Investigaciones Científicas, (IIM-CSIC), Vigo, Spain, 36208
| | - Robert N. Kelsh
- Centre for Regenerative Medicine and Developmental Biology Programme, Department of Biology and Biochemistry, University of Bath, Bath, England BA2 7AY
| | - José Miguel Cerdá-Reverter
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre de la Sal, Consejo Superior de Investigaciones Científicas, (IATS-CSIC), Ribera de Cabanes, Castellón, Spain, 12595
- * E-mail:
| |
Collapse
|
46
|
Experimental Dissection of Metalloproteinase Inhibition-Mediated and Toxic Effects of Phenanthroline on Zebrafish Development. Int J Mol Sci 2016; 17:ijms17091503. [PMID: 27618022 PMCID: PMC5037780 DOI: 10.3390/ijms17091503] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/25/2016] [Accepted: 09/02/2016] [Indexed: 12/27/2022] Open
Abstract
Metalloproteinases are zinc-dependent endopeptidases that function as primary effectors of tissue remodeling, cell-signaling, and many other roles. Their regulation is ferociously complex, and is exquisitely sensitive to their molecular milieu, making in vivo studies challenging. Phenanthroline (PhN) is an inexpensive, broad-spectrum inhibitor of metalloproteinases that functions by chelating the catalytic zinc ion, however its use in vivo has been limited due to suspected off-target effects. PhN is very similar in structure to phenanthrene (PhE), a well-studied poly aromatic hydrocarbon (PAH) known to cause toxicity in aquatic animals by activating the aryl hydrocarbon receptor (AhR). We show that zebrafish are more sensitive to PhN than PhE, and that PhN causes a superset of the effects caused by PhE. Morpholino knock-down of the AhR rescues the effects of PhN that are shared with PhE, suggesting these are due to PAH toxicity. The effects of PhN that are not shared with PhE (specifically disruption of neural crest development and angiogenesis) involve processes known to depend on metalloproteinase activity. Furthermore these PhN-specific effects are not rescued by AhR knock-down, suggesting that these are bona fide effects of metalloproteinase inhibition, and that PhN can be used as a broad spectrum metalloproteinase inhibitor for studies with zebrafish in vivo.
Collapse
|
47
|
Asad Z, Pandey A, Babu A, Sun Y, Shevade K, Kapoor S, Ullah I, Ranjan S, Scaria V, Bajpai R, Sachidanandan C. Rescue of neural crest-derived phenotypes in a zebrafish CHARGE model by Sox10 downregulation. Hum Mol Genet 2016; 25:3539-3554. [PMID: 27418670 PMCID: PMC5179949 DOI: 10.1093/hmg/ddw198] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 05/27/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022] Open
Abstract
CHD7 mutations are implicated in a majority of cases of the congenital disorder, CHARGE syndrome. CHARGE, an autosomal dominant syndrome, is known to affect multiple tissues including eye, heart, ear, craniofacial nerves and skeleton and genital organs. Using a morpholino-antisense-oligonucleotide-based zebrafish model for CHARGE syndrome, we uncover a complex spectrum of abnormalities in the neural crest and the crest-derived cell types. We report for the first time, defects in myelinating Schwann cells, enteric neurons and pigment cells in a CHARGE model. We also observe defects in the specification of peripheral neurons and the craniofacial skeleton as previously reported. Chd7 morphants have impaired migration of neural crest cells and deregulation of sox10 expression from the early stages. Knocking down Sox10 in the zebrafish CHARGE model rescued the defects in Schwann cells and craniofacial cartilage. Our zebrafish CHARGE model thus reveals important regulatory roles for Chd7 at multiple points of neural crest development viz., migration, fate choice and differentiation and we suggest that sox10 deregulation is an important driver of the neural crest-derived aspects of Chd7 dependent CHARGE syndrome.
Collapse
Affiliation(s)
- Zainab Asad
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Aditi Pandey
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Aswini Babu
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Yuhan Sun
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kaivalya Shevade
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shruti Kapoor
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Ikram Ullah
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Shashi Ranjan
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Vinod Scaria
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Ruchi Bajpai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chetana Sachidanandan
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| |
Collapse
|
48
|
Berry JP, Roy U, Jaja-Chimedza A, Sanchez K, Matysik J, Alia A. High-Resolution Magic Angle Spinning Nuclear Magnetic Resonance of Intact Zebrafish Embryos Detects Metabolic Changes Following Exposure to Teratogenic Polymethoxyalkenes from Algae. Zebrafish 2016; 13:456-65. [PMID: 27348393 DOI: 10.1089/zeb.2016.1280] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Techniques based on nuclear magnetic resonance (NMR) for imaging and chemical analyses of in vivo, or otherwise intact, biological systems are rapidly emerging and finding diverse applications within a wide range of fields. Very recently, several NMR-based techniques have been developed for the zebrafish as a model animal system. In the current study, the novel application of high-resolution magic angle spinning (HR-MAS) NMR is presented as a means of metabolic profiling of intact zebrafish embryos. Toward investigating the utility of HR-MAS NMR as a toxicological tool, these studies specifically examined metabolic changes of embryos exposed to polymethoxy-1-alkenes (PMAs)-a recently identified family of teratogenic compounds from freshwater algae-as emerging environmental contaminants. One-dimensional and two-dimensional HR-MAS NMR analyses were able to effectively identify and quantify diverse metabolites in early-stage (≤36 h postfertilization) embryos. Subsequent comparison of the metabolic profiles between PMA-exposed and control embryos identified several statistically significant metabolic changes associated with subacute exposure to the teratogen, including (1) elevated inositol as a recognized component of signaling pathways involved in embryo development; (2) increases in several metabolites, including inositol, phosphoryl choline, fatty acids, and cholesterol, which are associated with lipid composition of cell membranes; (3) concomitant increase in glucose and decrease in lactate; and (4) decreases in several biochemically related metabolites associated with central nervous system development and function, including γ-aminobutyric acid, glycine, glutamate, and glutamine. A potentially unifying model/hypothesis of PMA teratogenicity based on the data is presented. These findings, taken together, demonstrate that HR-MAS NMR is a promising tool for metabolic profiling in the zebrafish embryo, including toxicological applications.
Collapse
Affiliation(s)
- John P Berry
- 1 Department of Chemistry and Biochemistry, Florida International University , North Miami, Florida
| | - Upasana Roy
- 2 Institute of Medical Physics and Biophysics, University of Leipzig , Leipzig, Germany .,3 Institut für Analytische Chemie, University of Leipzig , Leipzig, Germany
| | - Asha Jaja-Chimedza
- 1 Department of Chemistry and Biochemistry, Florida International University , North Miami, Florida
| | - Kristel Sanchez
- 1 Department of Chemistry and Biochemistry, Florida International University , North Miami, Florida
| | - Joerg Matysik
- 3 Institut für Analytische Chemie, University of Leipzig , Leipzig, Germany
| | - A Alia
- 2 Institute of Medical Physics and Biophysics, University of Leipzig , Leipzig, Germany .,4 Leiden Institute of Chemistry , Leiden, the Netherlands
| |
Collapse
|
49
|
Chawla B, Schley E, Williams AL, Bohnsack BL. Retinoic Acid and Pitx2 Regulate Early Neural Crest Survival and Migration in Craniofacial and Ocular Development. ACTA ACUST UNITED AC 2016; 107:126-35. [PMID: 27175943 DOI: 10.1002/bdrb.21177] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/21/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Bahaar Chawla
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Elisa Schley
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
50
|
Chen Y, Yang F, Zheng H, Zhou J, Zhu G, Hu P, Wu W. Clinical and genetic investigation of families with type II Waardenburg syndrome. Mol Med Rep 2016; 13:1983-8. [PMID: 26781036 PMCID: PMC4768954 DOI: 10.3892/mmr.2016.4774] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 12/11/2015] [Indexed: 11/21/2022] Open
Abstract
The present study aimed to investigate the molecular pathology of Waardenburg syndrome type II in three families, in order to provide genetic diagnosis and hereditary counseling for family members. Relevant clinical examinations were conducted on the probands of the three pedigrees. Peripheral blood samples of the probands and related family members were collected and genomic DNA was extracted. The coding sequences of paired box 3 (PAX3), microphthalmia-associated transcription factor (MITF), sex-determining region Y-box 10 (SOX10) and snail family zinc finger 2 (SNAI2) were analyzed by polymerase chain reaction and DNA sequencing. The heterozygous mutation, c.649_651delAGA in exon 7 of the MITF gene was detected in the proband and all patients of pedigree 1; however, no pathological mutation of the relevant genes (MITF, SNAI2, SOX10 or PAX3) was detected in pedigrees 2 and 3. The heterozygous mutation c.649_651delAGA in exon 7 of the MITF gene is therefore considered the disease-causing mutation in pedigree 1. However, there are novel disease-causing genes in Waardenburg syndrome type II, which require further research.
Collapse
Affiliation(s)
- Yong Chen
- Key Laboratory of Genetics and Birth Health of Hunan, The Family Planning Institute of Hunan, Changsha, Hunan 410126, P.R. China
| | - Fuwei Yang
- Department of Otolaryngology, Meizhou People's Hospital, Meizhou, Guangdong 514031, P.R. China
| | - Hexin Zheng
- Key Laboratory of Genetics and Birth Health of Hunan, The Family Planning Institute of Hunan, Changsha, Hunan 410126, P.R. China
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Ganghua Zhu
- Department of Otolaryngology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Peng Hu
- Department of Otolaryngology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Weijing Wu
- Department of Otolaryngology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|