1
|
Hernández-Núñez I, Urman A, Zhang X, Jacobs W, Hoffman C, Rebba S, Harding EG, Li Q, Mao F, Cani AK, Chen S, Dawlaty MM, Rao RC, Ruzycki PA, Edwards JR, Clark BS. Active DNA demethylation is upstream of rod-photoreceptor fate determination and required for retinal development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636318. [PMID: 39975078 PMCID: PMC11838574 DOI: 10.1101/2025.02.03.636318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Retinal cell fate specification from multipotent retinal progenitors is governed by dynamic changes in chromatin structure and gene expression. Methylation at cytosines in DNA (5mC) is actively regulated for proper control of gene expression and chromatin architecture. Numerous genes display active DNA demethylation across retinal development; a process that requires oxidation of 5mC to 5-hydroxymethylcytosine (5hmC) and is controlled by the ten-eleven translocation methylcytosine dioxygenase (TET) enzymes. Using an allelic series of conditional TET enzyme mutants, we determine that DNA demethylation is required upstream of NRL and NR2E3 expression for the establishment of rod-photoreceptor fate. Using histological, behavioral, transcriptomic, and base-pair resolution DNA methylation analyses, we establish that inhibition of active DNA demethylation results in global changes in gene expression and methylation patterns that prevent photoreceptor precursors from adopting a rod-photoreceptor fate, instead producing a retina in which all photoreceptors specify as cones. Our results establish the TET enzymes and DNA demethylation as critical regulators of retinal development and cell fate specification, elucidating a novel mechanism required for the specification of rod-photoreceptors.
Collapse
Affiliation(s)
- Ismael Hernández-Núñez
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Alaina Urman
- Center for Pharmacogenetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaodong Zhang
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - William Jacobs
- Center for Pharmacogenetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Christy Hoffman
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Sohini Rebba
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Ellen G Harding
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Qiang Li
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Fengbiao Mao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Andi K Cani
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Shiming Chen
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Meelad M Dawlaty
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Department of Genetics, and Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rajesh C Rao
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department and Center of Computational Medicine and Bioinformatics, Comprehensive Cancer Center, A. Alfred Taubman Medical Research Institute, Center for RNA Biomedicine, Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Division of Ophthalmology, Surgery Section, VA Ann Arbor Health System, Ann Arbor, MI, USA
| | - Philip A Ruzycki
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - John R Edwards
- Center for Pharmacogenetics, Washington University School of Medicine, St. Louis, MO, USA
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S Clark
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
2
|
Peng X, Teng X, Ma Q, Han D. Serum Circulating mRNA Panel for the Early Detection of Gastric Cancer: A Potential Biomarker Test. ChemMedChem 2024; 19:e202400523. [PMID: 39234977 DOI: 10.1002/cmdc.202400523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
Circulating free messenger RNAs (cfmRNAs) in serum have emerged as potential noninvasive biomarkers for cancer diagnosis, including gastric cancer (GC). This study utilized RNA-sequencing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to identify a training set of 100 differentially expressed genes (DEGs) specific to GC patients. Employing a support vector machine (SVM) classification, we narrowed down the candidate gene set to 23, which was further refined to 4 genes-DMBX1, EVX1, MAL, and PIWIL1-after validation through reverse transcription quantitative polymerase chain reaction (RT-qPCR). The diagnostic performance of mRNA panels, particularly the combinations of DMBX1 with EVX1 and EVX1 with PIWIL1, was exceptional, achieving area under the curve (AUC) values of 0.800, sensitivities of 90.0 %, and specificities of 80.0 %. The accuracy of these biomarkers was corroborated through various machine learning algorithms, underscoring their robust diagnostic potential. The findings of this study are poised to significantly influence clinical practice by providing robust tools for early GC detection. As these biomarkers undergo further investigation and validation, they hold promise to become integral to the diagnostic for GC.
Collapse
Affiliation(s)
- Xinyu Peng
- School of Life Sciences, Shanghai University, 200444, Shanghai, China
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China
| | - Xiaoyan Teng
- Department of Laboratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People's six Hospital, 200233, Shanghai, China
| | - Qian Ma
- Department of Research and Development, Intellinosis Biotechnologies Co. Ltd., 201112, Shanghai, China
| | - Da Han
- School of Life Sciences, Shanghai University, 200444, Shanghai, China
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, China
| |
Collapse
|
3
|
Schilling K. Revisiting the development of cerebellar inhibitory interneurons in the light of single-cell genetic analyses. Histochem Cell Biol 2024; 161:5-27. [PMID: 37940705 PMCID: PMC10794478 DOI: 10.1007/s00418-023-02251-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 11/10/2023]
Abstract
The present review aims to provide a short update of our understanding of the inhibitory interneurons of the cerebellum. While these cells constitute but a minority of all cerebellar neurons, their functional significance is increasingly being recognized. For one, inhibitory interneurons of the cerebellar cortex are now known to constitute a clearly more diverse group than their traditional grouping as stellate, basket, and Golgi cells suggests, and this diversity is now substantiated by single-cell genetic data. The past decade or so has also provided important information about interneurons in cerebellar nuclei. Significantly, developmental studies have revealed that the specification and formation of cerebellar inhibitory interneurons fundamentally differ from, say, the cortical interneurons, and define a mode of diversification critically dependent on spatiotemporally patterned external signals. Last, but not least, in the past years, dysfunction of cerebellar inhibitory interneurons could also be linked with clinically defined deficits. I hope that this review, however fragmentary, may stimulate interest and help focus research towards understanding the cerebellum.
Collapse
Affiliation(s)
- Karl Schilling
- Anatomisches Institut - Anatomie und Zellbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Nussallee 10, 53115, Bonn, Germany.
| |
Collapse
|
4
|
Huang CX, Wang Z, Cheng J, Zhu Z, Guan NN, Song J. De novo establishment of circuit modules restores locomotion after spinal cord injury in adult zebrafish. Cell Rep 2022; 41:111535. [DOI: 10.1016/j.celrep.2022.111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/12/2022] [Accepted: 09/29/2022] [Indexed: 11/03/2022] Open
|
5
|
Hehr CL, Halabi R, McFarlane S. Spatial regulation of amacrine cell genesis by Semaphorin 3f. Dev Biol 2022; 491:66-81. [PMID: 36058267 DOI: 10.1016/j.ydbio.2022.08.008] [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: 12/01/2021] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE The axonal projections of retinal ganglion cells (RGCs) of the eye are topographically organized so that spatial information from visual images is preserved. This retinotopic organization is established during development by secreted morphogens that pattern domains of transcription factor expression within naso-temporal and dorso-ventral quadrants of the embryonic eye. Poorly understood are the downstream signaling molecules that generate the topographically organized retinal cells and circuits. The secreted signaling molecule Semaphorin 3fa (Sema3fa) belongs to the Sema family of molecules that provide positional information to developing cells. Here, we test a role for Sema3fa in cell genesis of the temporal zebrafish retina. METHODS We compare retinal cell genesis in wild type and sema3fa CRISPR zebrafish mutants by in situ hybridization and immunohistochemistry. RESULTS We find that mRNAs for sema3fa and known receptors, neuropilin2b (nrp2b) and plexina1a (plxna1a), are expressed by progenitors of the temporal, but not nasal zebrafish embryonic retina. In the sema3faca304/ca304 embryo, initially the domains of expression for atoh7 and neurod4, transcription factors necessary for the specification of RGCs and amacrine cells, respectively, are disrupted. Yet, post-embryonically only amacrine cells of the temporal retina are reduced in numbers, with both GABAergic and glycinergic subtypes affected. CONCLUSIONS These data suggest that Sema3fa acts early on embryonic temporal progenitors to control in a spatially-dependent manner the production of amacrine cells, possibly to allow the establishment of neural circuits with domain-specific functions. We propose that spatially restricted extrinsic signals in the neural retina control cell genesis in a domain-dependent manner.
Collapse
Affiliation(s)
- Carrie Lynn Hehr
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Rami Halabi
- 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
|
6
|
Xiang L, Zhang J, Rao FQ, Yang QL, Zeng HY, Huang SH, Xie ZX, Lv JN, Lin D, Chen XJ, Wu KC, Lu F, Huang XF, Chen Q. Depletion of miR-96 Delays, But Does Not Arrest, Photoreceptor Development in Mice. Invest Ophthalmol Vis Sci 2022; 63:24. [PMID: 35481839 PMCID: PMC9055555 DOI: 10.1167/iovs.63.4.24] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Abundant retinal microRNA-183 cluster (miR-183C) has been reported to be a key player in photoreceptor development and functionality in mice. However, whether there is a protagonist in this cluster remains unclear. Here, we used a mutant mouse model to study the role of miR-96, a member of miR-183C, in photoreceptor development and functionality. Methods The mature miR-96 sequence was removed using the CRISPR/Cas9 genome-editing system. Electroretinogram (ERG) and optical coherence tomography (OCT) investigated the changes in structure and function in mouse retinas. Immunostaining determined the localization and morphology of the retinal cells. RNA sequencing was conducted to observe retinal transcription alterations. Results The miR-96 mutant mice exhibited cone developmental delay, as occurs in miR-183/96 double knockout mice. Immunostaining of cone-specific marker genes revealed cone nucleus mislocalization and exiguous Opn1mw/Opn1sw in the mutant (MT) mouse outer segments at postnatal day 10. Interestingly, this phenomenon could be relieved in the adult stages. Transcriptome analysis revealed activation of microtubule-, actin filament–, and cilia-related pathways, further supporting the findings. Based on ERG and OCT results at different ages, the MT mice displayed developmental delay not only in cones but also in rods. In addition, a group of miR-96 potential direct and indirect target genes was summarized for interpretation and further studies of miR-96–related retinal developmental defects. Conclusions Depletion of miR-96 delayed but did not arrest photoreceptor development in mice. This miRNA is indispensable for mouse photoreceptor maturation, especially for cones.
Collapse
Affiliation(s)
- Lue Xiang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Juan Zhang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Feng-Qin Rao
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou, China
| | - Qiao-Li Yang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hui-Yi Zeng
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Sheng-Hai Huang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhen-Xiang Xie
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ji-Neng Lv
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Dan Lin
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xue-Jiao Chen
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Kun-Chao Wu
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fan Lu
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Xiu-Feng Huang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Chen
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| |
Collapse
|
7
|
Huang X, Xiang L, Liu W, Li M, Ren A, Chen Z, Zheng C, Chengcong C, Liu J, Yuan Y. Roles of diencephalon/mesencephalon homeobox 1 in the development and prognosis of hepatocellular carcinoma. Ann Hepatol 2022; 24:100314. [PMID: 33524552 DOI: 10.1016/j.aohep.2021.100314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES The oncogene diencephalon/mesencephalon homeobox 1 (DMBX1) is widely overexpressed in a variety of human cancers. The present study aimed to analyze the expression and clinical importance of DMBX1 in nonneoplastic tissues and tumor tissues from patients with hepatocellular carcinoma (HCC). MATERIALS AND METHODS DMBX1 expression in HCC and adjacent nontumor tissues was analyzed using immunohistochemical staining. Chi-square tests were applied to compare DMBX1 expression between the tumors and the adjacent normal tissues. We explored the correlation of DMBX1 expression with clinicopathological factors and its effect on the prognosis of HCC. Finally, we investigated the role of DMBX1 in HCC via knockdown experiments, which analyzed changes in cell invasion, cell proliferation and epithelial-mesenchymal transition (EMT) biomarkers (E-cadherin, N-cadherin, vimentin). The mRNAs that were coexpressed with DMBX1 in HCC, based on the TCGA cohort (n = 366), were obtained from the cBioPortal database. RESULTS The average score for DMBX1 expression was significantly different (P < 0.001) between HCC and paired adjacent nontumor tissues, and DMBX1 expression correlated with hepatitis B virus (HBV) infection, tumor size, metastasis, and tumor node metastasis (TNM) stage (P < 0.05). A multivariate Cox regression analysis identified significant correlations of DMBX1 expression with tumor metastasis, TNM stage, and tumor capsule. Moreover, Kaplan-Meier survival analysis revealed an association between DMBX1 overexpression and shorter overall survival of patients with HCC (P < 0.05). In HCC cell lines, silencing DMBX1 markedly inhibited migration, proliferation and EMT markers. The mRNAs that were negatively (R ≤ -0.25, n = 1094) or positively (R ≥ 0.25, n = 2906) coexpressed with DMBX1 mRNA were selected for further Gene Ontology enrichment analysis, and the results revealed that the predicted functions of DMBX1 in HCC support the in vitro experimental results. CONCLUSIONS Our data provide evidence that DMBX1 overexpression is associated with HCC metastasis and poor prognosis, suggesting that DMBX1 represents a therapeutic target in HCC.
Collapse
Affiliation(s)
- Xiaoting Huang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Leyang Xiang
- Department of Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Wei Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Mingyi Li
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Anbang Ren
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Zide Chen
- Department of Radiation Oncology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China
| | - Chu Zheng
- Academic Office, Guilin Medical University, Guilin, China
| | - Chen Chengcong
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Jinquan Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| | - Yawei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Retinal Stem Cell 'Retirement Plans': Growth, Regulation and Species Adaptations in the Retinal Ciliary Marginal Zone. Int J Mol Sci 2021; 22:ijms22126528. [PMID: 34207050 PMCID: PMC8234741 DOI: 10.3390/ijms22126528] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
The vertebrate retina develops from a specified group of precursor cells that adopt distinct identities and generate lineages of either the neural retina, retinal pigmented epithelium, or ciliary body. In some species, including teleost fish and amphibians, proliferative cells with stem-cell-like properties capable of continuously supplying new retinal cells post-embryonically have been characterized and extensively studied. This region, termed the ciliary or circumferential marginal zone (CMZ), possibly represents a conserved retinal stem cell niche. In this review, we highlight the research characterizing similar CMZ-like regions, or stem-like cells located at the peripheral margin, across multiple different species. We discuss the proliferative parameters, multipotency and growth mechanisms of these cells to understand how they behave in vivo and how different molecular factors and signalling networks converge at the CMZ niche to regulate their activity. The evidence suggests that the mature retina may have a conserved propensity for homeostatic growth and plasticity and that dysfunction in the regulation of CMZ activity may partially account for dystrophic eye growth diseases such as myopia and hyperopia. A better understanding of the properties of CMZ cells will enable important insight into how an endogenous generative tissue compartment can adapt to altered retinal physiology and potentially even restore vision loss caused by retinal degenerative conditions.
Collapse
|
9
|
Tao Y, Cao J, Li M, Hoffmann B, Xu K, Chen J, Lu X, Guo F, Li X, Phillips MJ, Gamm DM, Chen H, Zhang S. PAX6D instructs neural retinal specification from human embryonic stem cell-derived neuroectoderm. EMBO Rep 2020; 21:e50000. [PMID: 32700445 PMCID: PMC7507545 DOI: 10.15252/embr.202050000] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 11/09/2022] Open
Abstract
PAX6 is essential for neural retina (NR) and forebrain development but how PAX6 instructs NR versus forebrain specification remains unknown. We found that the paired-less PAX6, PAX6D, is expressed in NR cells during human eye development and along human embryonic stem cell (hESC) specification to retinal cells. hESCs deficient for PAX6D failed to enter NR specification. Induced expression of PAX6D but not PAX6A in a PAX6-null background restored the NR specification capacity. ChIP-Seq, confirmed by functional assays, revealed a set of retinal genes and non-retinal neural genes that are potential targets of PAX6D, including WNT8B. Inhibition of WNTs or knocking down of WNT8B restored the NR specification capacity of neuroepithelia with PAX6D knockout, whereas activation of WNTs blocked NR specification even when PAX6D was induced. Thus, PAX6D specifies neuroepithelia to NR cells via the regulation of WNT8B.
Collapse
Affiliation(s)
- Yunlong Tao
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Jingyuan Cao
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Mingxing Li
- Department of Rehabilitation of Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | | | - Ke Xu
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Jing Chen
- Department of Rehabilitation of Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xin Lu
- Wuhan No. 1 HospitalWuhanChina
| | - Fangliang Guo
- Neurological Department of Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiang Li
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - M Joseph Phillips
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
- McPherson Eye Research InstituteUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - David M Gamm
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
- McPherson Eye Research InstituteUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of Ophthalmology and Visual SciencesUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Hong Chen
- Department of Rehabilitation of Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Su‐Chun Zhang
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of NeuroscienceDepartment of NeurologySchool of Medicine and Public HealthUniversity of WisconsinMadisonWIUSA
- Program in Neuroscience & Behavioral DisordersDuke‐NUS Medical SchoolSingapore CitySingapore
| |
Collapse
|
10
|
Gibboney S, Orvis J, Kim K, Johnson CJ, Martinez-Feduchi P, Lowe EK, Sharma S, Stolfi A. Effector gene expression underlying neuron subtype-specific traits in the Motor Ganglion of Ciona. Dev Biol 2020; 458:52-63. [PMID: 31639337 PMCID: PMC6987015 DOI: 10.1016/j.ydbio.2019.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 12/31/2022]
Abstract
The central nervous system of the Ciona larva contains only 177 neurons. The precise regulation of neuron subtype-specific morphogenesis and differentiation observed during the formation of this minimal connectome offers a unique opportunity to dissect gene regulatory networks underlying chordate neurodevelopment. Here we compare the transcriptomes of two very distinct neuron types in the hindbrain/spinal cord homolog of Ciona, the Motor Ganglion (MG): the Descending decussating neuron (ddN, proposed homolog of Mauthner Cells in vertebrates) and the MG Interneuron 2 (MGIN2). Both types are invariantly represented by a single bilaterally symmetric left/right pair of cells in every larva. Supernumerary ddNs and MGIN2s were generated in synchronized embryos and isolated by fluorescence-activated cell sorting for transcriptome profiling. Differential gene expression analysis revealed ddN- and MGIN2-specific enrichment of a wide range of genes, including many encoding potential "effectors" of subtype-specific morphological and functional traits. More specifically, we identified the upregulation of centrosome-associated, microtubule-stabilizing/bundling proteins and extracellular guidance cues part of a single intrinsic regulatory program that might underlie the unique polarization of the ddNs, the only descending MG neurons that cross the midline. Consistent with our predictions, CRISPR/Cas9-mediated, tissue-specific elimination of two such candidate effectors, Efcab6-related and Netrin1, impaired ddN polarized axon outgrowth across the midline.
Collapse
Affiliation(s)
- Susanne Gibboney
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jameson Orvis
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Kwantae Kim
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Christopher J Johnson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | | | - Elijah K Lowe
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Sarthak Sharma
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
11
|
Wang C, Zou H, Yang H, Wang L, Chu H, Jiao J, Wang Y, Chen A. Long non‑coding RNA plasmacytoma variant translocation 1 gene promotes the development of cervical cancer via the NF‑κB pathway. Mol Med Rep 2019; 20:2433-2440. [PMID: 31322217 DOI: 10.3892/mmr.2019.10479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/20/2019] [Indexed: 11/06/2022] Open
Abstract
The long noncoding RNA plasmacytoma variant translocation 1 gene (LncRNA PVT1) has an important role in tumor occurrence and development, yet the role and underlying molecular mechanisms of this RNA in cervical cancer have not yet been elucidated. In the present study, three cervical cancer cell lines (HeLa, Ca Ski and SiHa) were used to verify how LncRNA PVT1 mediates cervical cancer development, and the H8 cell line was used as a control. A LncRNA PVT1 overexpression vector or small interfering RNAs targeting LncRNA PVT1 were transfected into cervical cancer cells to generate LncRNA PVT1 overexpression and silencing in these cells. LncRNA PVT1 overexpression accelerated the growth of cervical cancer cells by advancing the cell cycle and inhibiting cellular apoptosis; increases in Cyclin D1 (CCND1) mRNA and activated Bcl‑2 protein expression levels also supported this finding. Furthermore, NF‑κB activation and expression was increased by LncRNA PVT1 overexpression. In addition, NF‑κB activation or inhibition induced changes in cell viability, accompanied by changes in CCND1 and Bcl‑2 expression. Increases or decreases in microRNA‑16 (miR‑16) expression (using miR mimics and inhibitors) also corresponded to changes in LncRNA PVT1 expression, in vitro. miR‑16 mimics and inhibitor had opposite effects to those of NF‑κB activity, and miR‑16 was demonstrated to directly interact with the NF‑κB gene as measured using the dual‑luciferase assay. In summary, LncRNA PVT1 inhibits the effect of miR‑16, promoting the cell cycle and inhibiting cellular apoptosis of cervical cancer cells, potentially via the NF‑κB pathway. The data from the present study will contribute to the current knowledge surrounding the theoretical basis of cervical cancer and provide a new perspective for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Chang Wang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590, P.R. China
| | - Hao Zou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Hongjuan Yang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590, P.R. China
| | - Lei Wang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590, P.R. China
| | - Huijun Chu
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590, P.R. China
| | - Jinwen Jiao
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590, P.R. China
| | - Yankui Wang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590, P.R. China
| | - Aiping Chen
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590, P.R. China
| |
Collapse
|
12
|
Luo J, Liu K, Yao Y, Sun Q, Zheng X, Zhu B, Zhang Q, Xu L, Shen Y, Ren B. DMBX1 promotes tumor proliferation and regulates cell cycle progression via repressing OTX2-mediated transcription of p21 in lung adenocarcinoma cell. Cancer Lett 2019; 453:45-56. [DOI: 10.1016/j.canlet.2019.03.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 12/17/2022]
|
13
|
Feltes BC. Architects meets Repairers: The interplay between homeobox genes and DNA repair. DNA Repair (Amst) 2018; 73:34-48. [PMID: 30448208 DOI: 10.1016/j.dnarep.2018.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
Homeobox genes are widely considered the major protagonists of embryonic development and tissue formation. For the past decades, it was established that the deregulation of these genes is intimately related to developmental abnormalities and a broad range of diseases in adults. Since the proper regulation and expression of homeobox genes are necessary for a successful developmental program and tissue function, their relation to DNA repair mechanisms become a necessary discussion. However, important as it is, studies focused on the interplay between homeobox genes and DNA repair are scarce, and there is no critical discussion on the subject. Hence, in this work, I aim to provide the first review of the current knowledge of the interplay between homeobox genes and DNA repair mechanisms, and offer future perspectives on this, yet, young ground for new researches. Critical discussion is conducted, together with a careful assessment of each reviewed topic.
Collapse
Affiliation(s)
- Bruno César Feltes
- Institute of Informatics, Department of Theoretical Informatics, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
14
|
Dai M, Zhang Q, Zheng Z, Wang J. Retinal ganglion cell-conditioned medium and surrounding pressure alters gene expression and differentiation of rat retinal progenitor cells. Mol Med Rep 2018; 17:7177-7183. [PMID: 29568879 PMCID: PMC5928676 DOI: 10.3892/mmr.2018.8738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 12/04/2017] [Indexed: 12/13/2022] Open
Abstract
Loss of retinal ganglion cells is implicated in glaucoma and high intraocular pressure. Factors that affect the differentiation of retinal progenitor cells into retinal ganglion cells remain unclear. The present study aimed to investigate the effects of retinal ganglion cell‑conditioned medium on gene expression and differentiation in retinal progenitor cells, and the effects of surrounding pressure on the survival and differentiation of retinal progenitor cells. Retinal progenitor cells and retinal ganglion cells were isolated from rats. Immunofluorescence staining of Nestin and Thy1 was performed to identify rat retinal progenitor cells and retinal ganglion cells, respectively. Retinal progenitor cells and ganglion cells were cultured for 48 h under surrounding pressure of 0, 20, 40, 60 and 80 mmHg. Cellular apoptosis was detected using a caspase‑3 assay kit. In addition, the culture supernatant of rat retinal ganglion cells was collected. Retinal progenitor cells were cultured in the presence or absence of retinal ganglion‑conditioned medium for 72 h under normal pressure. Gene expression of Nestin, paired box protein 6 (PAX6), Thy1 and brain‑specific homeobox/POU domain protein 3 (Brn‑3) in retinal progenitor cells was detected by reverse transcription‑quantitative polymerase chain reaction. Retinal progenitor cells were cultured in retinal ganglion‑conditioned medium for 72 h under surrounding pressure of 0 and 40 mmHg, respectively, and flow cytometry was utilized to evaluate the effects of pressure on the differentiation of retinal progenitor cells into retinal ganglion cells. The results demonstrated that isolated retinal progenitor cells were Nestin‑positive and retinal ganglion cells were Thy1‑positive, suggesting successful isolation. The activity of caspase‑3 increased in retinal progenitor cells and retinal ganglion cells in a pressure‑dependent manner. When the surrounding pressure reached 40, 60 and 80 mmHg, the activity of caspase‑3 in retinal progenitor cells and ganglion cells increased significantly compared with cells that were not under pressure. Compared with retinal progenitor cells cultured without ganglion‑conditioned medium, those cultured with ganglion‑conditioned medium had significantly decreased expression levels of Nestin and PAX6, and increased expression levels of Thy1 and Brn3. Compared with 0 mmHg pressure, retinal progenitor cells cultured in ganglion‑conditioned medium under 40 mmHg pressure had increased percentages of Thy1‑positive cells. In conclusion, the apoptosis of rat retinal progenitor cells and retinal ganglion cells was pressure‑dependent. Retinal ganglion cell‑conditioned medium increased the differentiation of retinal progenitor cells into retinal ganglion‑like cells, and the differentiation increased as surrounding pressure increased. Current study provides insights that may contribute to the efforts of developing a treatment for glaucoma.
Collapse
Affiliation(s)
- Min Dai
- Department of Ophthalmology, Second People's Hospital of Yunnan Province, Kunming, Yunan 650021, P.R. China
| | - Qing Zhang
- Department of Ophthalmology, Second People's Hospital of Yunnan Province, Kunming, Yunan 650021, P.R. China
| | - Zhikun Zheng
- Department of Ophthalmology, Second People's Hospital of Yunnan Province, Kunming, Yunan 650021, P.R. China
| | - Jianzhou Wang
- Department of Ophthalmology, Yan'an Hospital, Kunming, Yunan 650051, P.R. China
| |
Collapse
|
15
|
Traylor-Knowles N, Rose NH, Sheets EA, Palumbi SR. Early Transcriptional Responses during Heat Stress in the Coral Acropora hyacinthus. THE BIOLOGICAL BULLETIN 2017; 232:91-100. [PMID: 28654330 DOI: 10.1086/692717] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Corals respond to heat pulses that cause bleaching with massive transcriptional change, but the immediate responses to stress that lead up to these shifts have never been detailed. Understanding these early signals could be important for identifying the regulatory mechanisms responsible for bleaching and how these mechanisms vary between more and less resilient corals. Using RNA sequencing (RNAseq) and sampling every 30 minutes during a short-term heat shock, we found that components of the transcriptome were significantly upregulated within 90 min and after a temperature increase of +2 °C. The developmental transcription factor, Krüppel-like factor 7, was highly expressed within 60 min, and stress-related transcription factors such as Elk-3 were highly expressed starting at 240 min. The sets of genes enriched for early transcriptional response to heat stress included heat shock proteins, small GTPases, and proteasome genes. Retrovirus-related Pol polyproteins from transposons were significantly expressed throughout the whole experiment. Lastly, we propose a model for early transcriptional regulation of protein degradation and cell adhesion response that may ultimately lead to the bleaching and stress response.
Collapse
|
16
|
Bao L, Wang L, Wei G, Wang Y, Wuyun G, Bo A. Role of microRNA-4458 in patients with non-small-cell lung cancer. Oncol Lett 2016; 12:3958-3966. [PMID: 27895756 DOI: 10.3892/ol.2016.5176] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/01/2016] [Indexed: 01/17/2023] Open
Abstract
Incidence and progression of non-small-cell lung cancer (NSCLC) is a multi-factor, multi-step process. The present study investigated the association between the expression level of microRNA (miR)-4458 in NSCLC and paracarcinoma liver tissues and survival rates, and studied the biological functions of miR-4458 at the cellular and protein level. NSCLC and paracarcinoma tissues were sequenced using a miR expression chip. The association between miR-4458 expression and tumor-node-metastasis staging, total survival rate and relapse-free survival rate was analyzed. miR-4458 was subjected to target gene prediction. The target protein of cyclin D1 (CCND1) was verified with western blot analysis, immunohistochemistry and a luciferase reporter assay. The relative level of miR-4458 in paracarcinoma tissues of 9 NSCLC patients decreased from 2.38 to 0.65 (P<0.001). Total five-year survival rates of the high-expression miR-4458 group (29.21%) significantly exceeded that of the low-expression group (14.37%) (P=0.025). The viability of human lung carcinoma A549 and H460 cells transfected with miR-4458 decreased significantly compared with cells transfected with a normal control (blank control plasmid) within 72 h (P<0.001). The percentage of A549 and H460 cells transfected with a miR-4458 mimic at the cell cycle stage G0/G1 was 69.94±8.05 and 68.15±7.75%, respectively. The percentages increased significantly compared with the control group (46.06±6.93 for A549 cells; 45.22±7.24 for H640 cells; P<0.001). CCND1 mRNA was downregulated significantly in H460 cells 72 h subsequent to the addition of miR-4458 mimics (P<0.001). The activity of mutant-CCND1 altered slightly, while the fluorescence intensity of the wild-type-CCND1 group decreased significantly following the addition of miR-4458 mimics. In conclusion, miR-4458 was expressed at low levels in lung cancer tissues, and it arrested cells in vitro at stage G0/G1 and inhibited cell proliferation. Therefore, miR-4458 may participate in the onset of lung cancer as a suppressor gene by inhibiting CCND1.
Collapse
Affiliation(s)
- Lidao Bao
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, P.R. China
| | - Linlin Wang
- Department of Respiration, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Guomin Wei
- Department of Respiration, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Yuehong Wang
- College of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010110, P.R. China
| | - Gerile Wuyun
- College of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010110, P.R. China
| | - Agula Bo
- College of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010110, P.R. China
| |
Collapse
|
17
|
Olsen JB, Wong L, Deimling S, Miles A, Guo H, Li Y, Zhang Z, Greenblatt JF, Emili A, Tropepe V. G9a and ZNF644 Physically Associate to Suppress Progenitor Gene Expression during Neurogenesis. Stem Cell Reports 2016; 7:454-470. [PMID: 27546533 PMCID: PMC5031922 DOI: 10.1016/j.stemcr.2016.06.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 01/05/2023] Open
Abstract
Proliferating progenitor cells undergo changes in competence to give rise to post-mitotic progeny of specialized function. These cell-fate transitions typically involve dynamic regulation of gene expression by histone methyltransferase (HMT) complexes. However, the composition, roles, and regulation of these assemblies in regulating cell-fate decisions in vivo are poorly understood. Using unbiased affinity purification and mass spectrometry, we identified the uncharacterized C2H2-like zinc finger protein ZNF644 as a G9a/GLP-interacting protein and co-regulator of histone methylation. In zebrafish, functional characterization of ZNF644 orthologs, znf644a and znf644b, revealed complementary roles in regulating G9a/H3K9me2-mediated gene silencing during neurogenesis. The non-overlapping requirements for znf644a and znf644b during retinal differentiation demarcate critical aspects of retinal differentiation programs regulated by differential G9a-ZNF644 associations, such as transitioning proliferating progenitor cells toward differentiation. Collectively, our data point to ZNF644 as a critical co-regulator of G9a/H3K9me2-mediated gene silencing during neuronal differentiation.
Collapse
Affiliation(s)
- Jonathan B Olsen
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Medical Science Building, Toronto, ON M5S 3E1, Canada
| | - Loksum Wong
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Steven Deimling
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Amanda Miles
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Hongbo Guo
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Yue Li
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Medical Science Building, Toronto, ON M5S 3E1, Canada; Department of Computer Science, University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Zhaolei Zhang
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Medical Science Building, Toronto, ON M5S 3E1, Canada; Department of Computer Science, University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Jack F Greenblatt
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Medical Science Building, Toronto, ON M5S 3E1, Canada
| | - Andrew Emili
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Medical Science Building, Toronto, ON M5S 3E1, Canada.
| | - Vincent Tropepe
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada; Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON M5S 3B2, Canada.
| |
Collapse
|
18
|
Miles A, Tropepe V. Coordinating progenitor cell cycle exit and differentiation in the developing vertebrate retina. NEUROGENESIS 2016; 3:e1161697. [PMID: 27604453 PMCID: PMC4974023 DOI: 10.1080/23262133.2016.1161697] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/09/2016] [Accepted: 02/29/2016] [Indexed: 02/06/2023]
Abstract
The proper development of the vertebrate retina relies heavily on producing the correct number and type of differentiated retinal cell types. To achieve this, proliferating retinal progenitor cells (RPCs) must exit the cell cycle at an appropriate time and correctly express a subset of differentiation markers that help specify retinal cell fate. Homeobox genes, which encode a family of transcription factors, have been accredited to both these processes, implicated in the transcriptional regulation of important cell cycle components, such as cyclins and cyclin-dependent kinases, and proneural genes. This dual regulation of homeobox genes allows these factors to help co-ordinate the transition from the proliferating RPC to postmitotic, differentiated cell. However, understanding the exact molecular targets of these factors remains a challenging task. This commentary highlights the current knowledge we have about how these factors regulate cell cycle progression and differentiation, with particular emphasis on a recent discovery from our lab demonstrating an antagonistic relationship between Vsx2 and Dmbx1 to control RPC proliferation. Future studies should aim to further understand the direct transcriptional targets of these genes, additional co-factors/interacting proteins and the possible recruitment of epigenetic machinery by these homeobox genes.
Collapse
Affiliation(s)
- Amanda Miles
- Department of Cell & Systems Biology, University of Toronto , Toronto, Ontario, Canada
| | - Vincent Tropepe
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada; Department of Ophthalmology & Vision Sciences; Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Valdivia LE, Lamb DB, Horner W, Wierzbicki C, Tafessu A, Williams AM, Gestri G, Krasnow AM, Vleeshouwer-Neumann TS, Givens M, Young RM, Lawrence LM, Stickney HL, Hawkins TA, Schwarz QP, Cavodeassi F, Wilson SW, Cerveny KL. Antagonism between Gdf6a and retinoic acid pathways controls timing of retinal neurogenesis and growth of the eye in zebrafish. Development 2016; 143:1087-98. [PMID: 26893342 PMCID: PMC4852494 DOI: 10.1242/dev.130922] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/08/2016] [Indexed: 12/27/2022]
Abstract
Maintaining neurogenesis in growing tissues requires a tight balance between progenitor cell proliferation and differentiation. In the zebrafish retina, neuronal differentiation proceeds in two stages with embryonic retinal progenitor cells (RPCs) of the central retina accounting for the first rounds of differentiation, and stem cells from the ciliary marginal zone (CMZ) being responsible for late neurogenesis and growth of the eye. In this study, we analyse two mutants with small eyes that display defects during both early and late phases of retinal neurogenesis. These mutants carry lesions in gdf6a, a gene encoding a BMP family member previously implicated in dorsoventral patterning of the eye. We show that gdf6a mutant eyes exhibit expanded retinoic acid (RA) signalling and demonstrate that exogenous activation of this pathway in wild-type eyes inhibits retinal growth, generating small eyes with a reduced CMZ and fewer proliferating progenitors, similar to gdf6a mutants. We provide evidence that RA regulates the timing of RPC differentiation by promoting cell cycle exit. Furthermore, reducing RA signalling in gdf6a mutants re-establishes appropriate timing of embryonic retinal neurogenesis and restores putative stem and progenitor cell populations in the CMZ. Together, our results support a model in which dorsally expressed gdf6a limits RA pathway activity to control the transition from proliferation to differentiation in the growing eye. Summary: In the vertebrate eye, dorsally expressed Gdf6a limits RA pathway activity to control the transition from proliferation to differentiation, thereby regulating eye size.
Collapse
Affiliation(s)
- Leonardo E Valdivia
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Dayna B Lamb
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Wilson Horner
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Claudia Wierzbicki
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Amanuel Tafessu
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Audrey M Williams
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Gaia Gestri
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Anna M Krasnow
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | | | - McKenzie Givens
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Rodrigo M Young
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Lisa M Lawrence
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Heather L Stickney
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Thomas A Hawkins
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Quenten P Schwarz
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Florencia Cavodeassi
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Kara L Cerveny
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| |
Collapse
|