1
|
de Toledo VHC, Feltrin AS, Barbosa AR, Tahira AC, Brentani H. Sex differences in gene regulatory networks during mid-gestational brain development. Front Hum Neurosci 2022; 16:955607. [PMID: 36061507 PMCID: PMC9428411 DOI: 10.3389/fnhum.2022.955607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Neurodevelopmental disorders differ considerably between males and females, and fetal brain development is one of the most critical periods to determine risk for these disorders. Transcriptomic studies comparing male and female fetal brain have demonstrated that the highest difference in gene expression occurs in sex chromosomes, but several autossomal genes also demonstrate a slight difference that has not been yet explored. In order to investigate biological pathways underlying fetal brain sex differences, we applied medicine network principles using integrative methods such as co-expression networks (CEMiTool) and regulatory networks (netZoo). The pattern of gene expression from genes in the same pathway tend to reflect biologically relevant phenomena. In this study, network analysis of fetal brain expression reveals regulatory differences between males and females. Integrating two different bioinformatics tools, our results suggest that biological processes such as cell cycle, cell differentiation, energy metabolism and extracellular matrix organization are consistently sex-biased. MSET analysis demonstrates that these differences are relevant to neurodevelopmental disorders, including autism.
Collapse
Affiliation(s)
- Victor Hugo Calegari de Toledo
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Psicopatologia e Terapêutica Psiquiátrica (LIM23), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Ana Carolina Tahira
- Laboratório de Expressão Gênica, Departamento de Parasitologia, Instituto Butantan, São Paulo, Brazil
| | - Helena Brentani
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Psicopatologia e Terapêutica Psiquiátrica (LIM23), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
- *Correspondence: Helena Brentani
| |
Collapse
|
2
|
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: 0.5] [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
|
3
|
Kerschner JL, Paranjapye A, NandyMazumdar M, Yin S, Leir SH, Harris A. OTX2 regulates CFTR expression during endoderm differentiation and occupies 3' cis-regulatory elements. Dev Dyn 2021; 250:684-700. [PMID: 33386644 PMCID: PMC11227118 DOI: 10.1002/dvdy.293] [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: 10/05/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Cell-specific and developmental mechanisms contribute to expression of the cystic fibrosis transmembrane conductance regulator (CFTR) gene; however, its developmental regulation is poorly understood. Here we use human induced pluripotent stem cells differentiated into pseudostratified airway epithelial cells to study these mechanisms. RESULTS Changes in gene expression and open chromatin profiles were investigated by RNA-seq and ATAC-seq, and revealed that alterations in CFTR expression are associated with differences in stage-specific open chromatin. Additionally, two novel open chromatin regions, at +19.6 kb and +22.6 kb 3' to the CFTR translational stop signal, were observed in definitive endoderm (DE) cells, prior to an increase in CFTR expression in anterior foregut endoderm (AFE) cells. Chromatin studies in DE and AFE cells revealed enrichment of active enhancer marks and occupancy of OTX2 at these sites in DE cells. Loss of OTX2 in DE cells alters histone modifications across the CFTR locus and results in a 2.5-fold to 5-fold increase in CFTR expression. However, deletion of the +22.6 kb site alone does not affect CFTR expression in DE or AFE cells. CONCLUSIONS These results suggest that a network of interacting cis-regulatory elements recruit OTX2 to the locus to impact CFTR expression during early endoderm differentiation.
Collapse
Affiliation(s)
- Jenny L Kerschner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Monali NandyMazumdar
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Shiyi Yin
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Shih-Hsing Leir
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
4
|
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: 2.4] [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]
|
5
|
Furchtgott LA, Melton S, Menon V, Ramanathan S. Discovering sparse transcription factor codes for cell states and state transitions during development. eLife 2017; 6:e20488. [PMID: 28296636 PMCID: PMC5352226 DOI: 10.7554/elife.20488] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/31/2017] [Indexed: 12/16/2022] Open
Abstract
Computational analysis of gene expression to determine both the sequence of lineage choices made by multipotent cells and to identify the genes influencing these decisions is challenging. Here we discover a pattern in the expression levels of a sparse subset of genes among cell types in B- and T-cell developmental lineages that correlates with developmental topologies. We develop a statistical framework using this pattern to simultaneously infer lineage transitions and the genes that determine these relationships. We use this technique to reconstruct the early hematopoietic and intestinal developmental trees. We extend this framework to analyze single-cell RNA-seq data from early human cortical development, inferring a neocortical-hindbrain split in early progenitor cells and the key genes that could control this lineage decision. Our work allows us to simultaneously infer both the identity and lineage of cell types as well as a small set of key genes whose expression patterns reflect these relationships.
Collapse
Affiliation(s)
- Leon A Furchtgott
- FAS Center for Systems Biology, Harvard University, Cambridge, United States
- Biophysics Program, Harvard University, Cambridge, United States
| | - Samuel Melton
- FAS Center for Systems Biology, Harvard University, Cambridge, United States
- Harvard Stem Cell Institute, Harvard University, Cambridge, United States
| | - Vilas Menon
- Allen Institute for Brain Science, Seattle, United States
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Sharad Ramanathan
- FAS Center for Systems Biology, Harvard University, Cambridge, United States
- Harvard Stem Cell Institute, Harvard University, Cambridge, United States
- Allen Institute for Brain Science, Seattle, United States
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
- School of Engineering and Applied Sciences, Harvard University, Cambridge, United States
| |
Collapse
|
6
|
Hirono S, Lee EY, Kuribayashi S, Fukuda T, Saeki N, Minokoshi Y, Iwanaga T, Miki T. Importance of Adult Dmbx1 in Long-Lasting Orexigenic Effect of Agouti-Related Peptide. Endocrinology 2016; 157:245-57. [PMID: 26505115 DOI: 10.1210/en.2015-1560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dmbx1 is a brain-specific homeodomain transcription factor expressed primarily during embryogenesis, and its systemic disruption (Dmbx1(-/-)) in the ICR mouse strain resulted in leanness associated with impaired long-lasting orexigenic effect of agouti-related peptide (AgRP). Because spatial and temporal expression patterns of Dmbx1 change dramatically during embryogenesis, it remains unknown when and where Dmbx1 plays a critical role in energy homeostasis. In the present study, the physiological roles of Dmbx1 were examined by its conditional disruption (Dmbx1(loxP/loxP)) in the C57BL/6 mouse strain. Although Dmbx1 disruption in fetal brain resulted in neonatal lethality, its disruption by synapsin promoter-driven Cre recombinase, which eliminated Dmbx1 expression postnatally, exempted the mice (Syn-Cre;Dmbx1(loxP/loxP) mice) from lethality. Syn-Cre;Dmbx1(loxP/loxP) mice show mild leanness and impaired long-lasting orexigenic action of AgRP, demonstrating the physiological relevance of Dmbx1 in the adult. Visualization of Dmbx1-expressing neurons in adult brain using the mice harboring tamoxifen-inducible Cre recombinase in the Dmbx1 locus (Dmbx1(CreERT2/+) mice) revealed Dmbx1 expression in small numbers of neurons in restricted regions, including the lateral parabrachial nucleus (LPB). Notably, c-Fos expression in LPB was increased at 48 hours after AgRP administration in Dmbx1(loxP/loxP) mice but not in Syn-Cre;Dmbx1(loxP/loxP) mice. These c-Fos-positive neurons in LPB did not coincide with neurons expressing Dmbx1 or melanocortin 4 receptor but did coincide with those expressing calcitonin gene-related peptide. Accordingly, Dmbx1 in the adult LPB is required for the long-lasting orexigenic effect of AgRP via the neural circuitry involving calcitonin gene-related peptide neurons.
Collapse
Affiliation(s)
- Seiichiro Hirono
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| | - Eun Young Lee
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| | - Shunsuke Kuribayashi
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| | - Takahiro Fukuda
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| | - Naokatsu Saeki
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| | - Yasuhiko Minokoshi
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| | - Toshihiko Iwanaga
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| | - Takashi Miki
- Departments of Medical Physiology (S.H., E.Y.L., S.K., T.M.) and Neurological Surgery (S.H., N.S.), Chiba University Graduate School of Medicine, Chuo-ku, Chiba, 260-0856, Japan; Division of Neuropathology (T.F.), Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-0003, Japan; Department of Developmental Physiology (Y.M.), National Institute for Physiological Sciences, Myoudaijicho, Okazaki-City, 444-8585, Japan; and Laboratory of Histology and Cytology (T.I.), Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
7
|
Hoch RV, Lindtner S, Price JD, Rubenstein JLR. OTX2 Transcription Factor Controls Regional Patterning within the Medial Ganglionic Eminence and Regional Identity of the Septum. Cell Rep 2015; 12:482-94. [PMID: 26166575 DOI: 10.1016/j.celrep.2015.06.043] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/27/2015] [Accepted: 06/14/2015] [Indexed: 11/27/2022] Open
Abstract
The Otx2 homeodomain transcription factor is essential for gastrulation and early neural development. We generated Otx2 conditional knockout (cKO) mice to investigate its roles in telencephalon development after neurulation (approximately embryonic day 9.0). We conducted transcriptional profiling and in situ hybridization to identify genes de-regulated in Otx2 cKO ventral forebrain. In parallel, we used chromatin immunoprecipitation sequencing to identify enhancer elements, the OTX2 binding motif, and de-regulated genes that are likely direct targets of OTX2 transcriptional regulation. We found that Otx2 was essential in septum specification, regulation of Fgf signaling in the rostral telencephalon, and medial ganglionic eminence (MGE) patterning, neurogenesis, and oligodendrogenesis. Within the MGE, Otx2 was required for ventral, but not dorsal, identity, thus controlling the production of specific MGE derivatives.
Collapse
Affiliation(s)
- Renée V Hoch
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Susan Lindtner
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James D Price
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John L R Rubenstein
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Walton T, Preston E, Nair G, Zacharias AL, Raj A, Murray JI. The Bicoid class homeodomain factors ceh-36/OTX and unc-30/PITX cooperate in C. elegans embryonic progenitor cells to regulate robust development. PLoS Genet 2015; 11:e1005003. [PMID: 25738873 PMCID: PMC4349592 DOI: 10.1371/journal.pgen.1005003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 01/14/2015] [Indexed: 01/30/2023] Open
Abstract
While many transcriptional regulators of pluripotent and terminally differentiated states have been identified, regulation of intermediate progenitor states is less well understood. Previous high throughput cellular resolution expression studies identified dozens of transcription factors with lineage-specific expression patterns in C. elegans embryos that could regulate progenitor identity. In this study we identified a broad embryonic role for the C. elegans OTX transcription factor ceh-36, which was previously shown to be required for the terminal specification of four neurons. ceh-36 is expressed in progenitors of over 30% of embryonic cells, yet is not required for embryonic viability. Quantitative phenotyping by computational analysis of time-lapse movies of ceh-36 mutant embryos identified cell cycle or cell migration defects in over 100 of these cells, but most defects were low-penetrance, suggesting redundancy. Expression of ceh-36 partially overlaps with that of the PITX transcription factor unc-30. unc-30 single mutants are viable but loss of both ceh-36 and unc-30 causes 100% lethality, and double mutants have significantly higher frequencies of cellular developmental defects in the cells where their expression normally overlaps. These factors are also required for robust expression of the downstream developmental regulator mls-2/HMX. This work provides the first example of genetic redundancy between the related yet evolutionarily distant OTX and PITX families of bicoid class homeodomain factors and demonstrates the power of quantitative developmental phenotyping in C. elegans to identify developmental regulators acting in progenitor cells.
Collapse
Affiliation(s)
- Travis Walton
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elicia Preston
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gautham Nair
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Amanda L. Zacharias
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Arjun Raj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John Isaac Murray
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
9
|
Expression of VEGF-A, Otx homeobox and p53 family genes in proliferative vitreoretinopathy. Mediators Inflamm 2013; 2013:857380. [PMID: 24227910 PMCID: PMC3818919 DOI: 10.1155/2013/857380] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/02/2013] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Proliferative vitreoretinopathy (PVR) is a severe inflammatory complication of retinal detachment. Pathological epiretinal membranes grow on the retina surface leading to contraction, and surgery fails in 5% to 10% of the cases. We evaluated the expression of VEGF-A, Otx1, Otx2, Otx3, and p53 family members from PVR specimens to correlate their role in inducing or preventing the pathology. METHODS Twelve retinal samples were taken from patients affected by PVR during therapeutic retinectomies in vitreoretinal surgery. Gene expression was evaluated using quantitative real-time reverse transcriptase PCR analysis and immunohistochemistry, using four healthy human retinae as control. RESULT Controls showed basal expression of all genes. PVR samples showed little or no expression of Otx1 and variable expression of VEGF-A, Otx2, Otx3, p53, and p63 genes. Significant correlation was found among VEGF-A, Otx2, p53, and p63 and between Otx1 and Otx3. CONCLUSIONS Otx homeobox, p53 family, and VEGF-A genes are expressed in PVR human retina. We individuated two possible pathways (VEGF-A, Otx2, p53, p63 and Otx1 and Otx3) involved in PVR progression that could influence in different manners the course of the pathology. Individuating the genetic pathways of PVR represents a novel approach to PVR therapies.
Collapse
|
10
|
Ramialison M, Reinhardt R, Henrich T, Wittbrodt B, Kellner T, Lowy CM, Wittbrodt J. Cis-regulatory properties of medaka synexpression groups. Development 2012; 139:917-28. [PMID: 22318626 DOI: 10.1242/dev.071803] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
During embryogenesis, tissue specification is triggered by the expression of a unique combination of developmental genes and their expression in time and space is crucial for successful development. Synexpression groups are batteries of spatiotemporally co-expressed genes that act in shared biological processes through their coordinated expression. Although several synexpression groups have been described in numerous vertebrate species, the regulatory mechanisms that orchestrate their common complex expression pattern remain to be elucidated. Here we performed a pilot screen on 560 genes of the vertebrate model system medaka (Oryzias latipes) to systematically identify synexpression groups and investigate their regulatory properties by searching for common regulatory cues. We find that synexpression groups share DNA motifs that are arranged in various combinations into cis-regulatory modules that drive co-expression. In contrast to previous assumptions that these genes are located randomly in the genome, we discovered that genes belonging to the same synexpression group frequently occur in synexpression clusters in the genome. This work presents a first repertoire of synexpression group common signatures, a resource that will contribute to deciphering developmental gene regulatory networks.
Collapse
Affiliation(s)
- Mirana Ramialison
- University of Heidelberg, Centre for Organismal Studies, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
11
|
Tsachaki M, Sprecher SG. Genetic and developmental mechanisms underlying the formation of theDrosophilacompound eye. Dev Dyn 2011; 241:40-56. [DOI: 10.1002/dvdy.22738] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2011] [Indexed: 01/15/2023] Open
|
12
|
Wong L, Weadick CJ, Kuo C, Chang BSW, Tropepe V. Duplicate dmbx1 genes regulate progenitor cell cycle and differentiation during zebrafish midbrain and retinal development. BMC DEVELOPMENTAL BIOLOGY 2010; 10:100. [PMID: 20860823 PMCID: PMC2954992 DOI: 10.1186/1471-213x-10-100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 09/22/2010] [Indexed: 01/03/2023]
Abstract
Background The Dmbx1 gene is important for the development of the midbrain and hindbrain, and mouse gene targeting experiments reveal that this gene is required for mediating postnatal and adult feeding behaviours. A single Dmbx1 gene exists in terrestrial vertebrate genomes, while teleost genomes have at least two paralogs. We compared the loss of function of the zebrafish dmbx1a and dmbx1b genes in order to gain insight into the molecular mechanism by which dmbx1 regulates neurogenesis, and to begin to understand why these duplicate genes have been retained in the zebrafish genome. Results Using gene knockdown experiments we examined the function of the dmbx1 gene paralogs in zebrafish, dmbx1a and dmbx1b in regulating neurogenesis in the developing retina and midbrain. Dose-dependent loss of dmbx1a and dmbx1b function causes a significant reduction in growth of the midbrain and retina that is evident between 48-72 hpf. We show that this phenotype is not due to patterning defects or persistent cell death, but rather a deficit in progenitor cell cycle exit and differentiation. Analyses of the morphant retina or anterior hindbrain indicate that paralogous function is partially diverged since loss of dmbx1a is more severe than loss of dmbx1b. Molecular evolutionary analyses of the Dmbx1 genes suggest that while this gene family is conservative in its evolution, there was a dramatic change in selective constraint after the duplication event that gave rise to the dmbx1a and dmbx1b gene families in teleost fish, suggestive of positive selection. Interestingly, in contrast to zebrafish dmbx1a, over expression of the mouse Dmbx1 gene does not functionally compensate for the zebrafish dmbx1a knockdown phenotype, while over expression of the dmbx1b gene only partially compensates for the dmbx1a knockdown phenotype. Conclusion Our data suggest that both zebrafish dmbx1a and dmbx1b genes are retained in the fish genome due to their requirement during midbrain and retinal neurogenesis, although their function is partially diverged. At the cellular level, Dmbx1 regulates cell cycle exit and differentiation of progenitor cells. The unexpected observation of putative post-duplication positive selection of teleost Dmbx1 genes, especially dmbx1a, and the differences in functionality between the mouse and zebrafish genes suggests that the teleost Dmbx1 genes may have evolved a diverged function in the regulation of neurogenesis.
Collapse
Affiliation(s)
- Loksum Wong
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
13
|
Seino Y, Miki T, Kiyonari H, Abe T, Fujimoto W, Kimura K, Takeuchi A, Takahashi Y, Oiso Y, Iwanaga T, Seino S. Isx participates in the maintenance of vitamin A metabolism by regulation of beta-carotene 15,15'-monooxygenase (Bcmo1) expression. J Biol Chem 2007; 283:4905-11. [PMID: 18093975 DOI: 10.1074/jbc.m707928200] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Isx (intestine specific homeobox) is an intestine-specific transcription factor. To elucidate its physiological function, we generated Isx-deficient mice by knocking in the beta-galactosidase gene (LacZ) in the Isx locus (IsxLacZ/LacZ mice). LacZ staining of heterozygous (IsxLacZ/+) mice revealed that Isx was expressed abundantly in intestinal epithelial cells from duodenum to proximal colon. Quantitative mRNA expression profiling of duodenum and jejunum showed that beta-carotene 15,15'-monooxygenase (EC1.14.99.36 Bcmo1) and the class B type I scavenger receptor, which are involved in vitamin A synthesis and carotenoid uptake, respectively, were drastically increased in IsxLacZ/LacZ mice. Although mild vitamin A deficiency decreased Isx expression in duodenum of wild-type (Isx+/+) mice, severe vitamin A deficiency decreased Isx mRNA expression in both duodenum and jejunum of Isx+/+ mice. On the other hand, vitamin A deficiency increased Bcmo1 expression in both duodenum and jejunum of Isx+/+ mice. However, Bcmo1 expression was not increased in duodenum of IsxLacZ/LacZ mice by mild vitamin A deficiency. These data suggest that Isx participates in the maintenance of vitamin A metabolism by regulating Bcmo1 expression in the intestine.
Collapse
Affiliation(s)
- Yusuke Seino
- Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Fujimoto W, Shiuchi T, Miki T, Minokoshi Y, Takahashi Y, Takeuchi A, Kimura K, Saito M, Iwanaga T, Seino S. Dmbx1 is essential in agouti-related protein action. Proc Natl Acad Sci U S A 2007; 104:15514-9. [PMID: 17873059 PMCID: PMC1976593 DOI: 10.1073/pnas.0707328104] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dmbx1 is a paired-class homeodomain transcription factor. We show here that mice deficient in Dmbx1 exhibit severe leanness associated with hypophagia and hyperactivity and that isolation of a Dmbx1(-/-) mouse from its cohabitants induces self-starvation, sometimes leading to death, features similar to those of anorexia nervosa in humans. Interestingly, overexpression of agouti in Dmbx1(-/-) mice failed to induce aspects of the A(y)/a phenotype, including hyperphagia, obesity, and diabetes mellitus. In Dmbx1(-/-) mice, administration of agouti-related protein increased cumulative food intake for the initial 6 h but significantly decreased it over 24- and 48-h periods. In addition, Dmbx1 was shown to be expressed at embryonic day 15.5 in the lateral parabrachial nucleus, the rostral nucleus of the tractus solitarius, the dorsal motor nucleus of the vagus, and the reticular nucleus in the brainstem, all of which receive melanocortin signaling, indicating involvement of Dmbx1 in the development of the neural network for the signaling. Thus, Dmbx1 is essential for various actions of agouti-related protein and plays a role in normal regulation of energy homeostasis and behavior.
Collapse
Affiliation(s)
- Wakako Fujimoto
- *Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- Laboratory of Histology and Cytology, Graduate School of Medicine and
| | - Tetsuya Shiuchi
- Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Takashi Miki
- *Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yasuhiko Minokoshi
- Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Yoshihisa Takahashi
- *Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Ayako Takeuchi
- *Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Kazuhiro Kimura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-8638, Japan; and
| | - Masayuki Saito
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-8638, Japan; and
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Graduate School of Medicine and
| | - Susumu Seino
- *Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- To whom correspondence should be addressed at:
7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan. E-mail:
| |
Collapse
|
15
|
Chang L, Khoo B, Wong L, Tropepe V. Genomic sequence and spatiotemporal expression comparison of zebrafish mbx1 and its paralog, mbx2. Dev Genes Evol 2006; 216:647-54. [PMID: 16733737 DOI: 10.1007/s00427-006-0082-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Accepted: 05/01/2006] [Indexed: 11/30/2022]
Abstract
The expression of midbrain homeobox-1 (mbx1) defines a discrete region in the vertebrate neural plate that will give rise to the mesencephalon, as well as subregions of the diencephalon and retinal field. Here, we report on the identification and cloning of a second Mbx gene in zebrafish, termed mbx2. Genomic sequence comparison suggests that mbx1 and mbx2 are derived from the duplication of a single putative ancestral gene that is conserved in other vertebrates as a single copy gene. Furthermore, phylogenetic analyses indicate that the mbx genes belong to a novel subgroup of paired-like homeobox genes. Finally, quantitative reverse transcriptase-PCR and whole mount in situ hybridization experiments revealed a pattern of partial spatiotemporal expression divergence between the mbx paralogs that correlates with sequence divergence in noncoding regulatory domains. Our data support a subfunctionalization model that may explain the retention of duplicate mbx genes in teleosts.
Collapse
Affiliation(s)
- Lou Chang
- Department of Cell & Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, M5S 3G5, Canada
| | | | | | | |
Collapse
|
16
|
Castro LFC, Rasmussen SLK, Holland PWH, Holland ND, Holland LZ. A Gbx homeobox gene in amphioxus: insights into ancestry of the ANTP class and evolution of the midbrain/hindbrain boundary. Dev Biol 2006; 295:40-51. [PMID: 16687133 DOI: 10.1016/j.ydbio.2006.03.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 02/28/2006] [Accepted: 03/01/2006] [Indexed: 11/24/2022]
Abstract
In the vertebrate central nervous system (CNS), mutual antagonism between posteriorly expressed Gbx2 and anteriorly expressed Otx2 positions the midbrain/hindbrain boundary (MHB), but does not induce MHB organizer genes such as En, Pax2/5/8 and Wnt1. In the CNS of the cephalochordate amphioxus, Otx is also expressed anteriorly, but En, Pax2/5/8 and Wnt1 are not expressed near the caudal limit of Otx, raising questions about the existence of an MHB organizer in amphioxus. To investigate the evolutionary origins of the MHB, we cloned the single amphioxus Gbx gene. Fluorescence in situ hybridization showed that, as in vertebrates, amphioxus Gbx and the Hox cluster are on the same chromosome. From analysis of linked genes, we argue that during evolution a single ancestral Gbx gene duplicated fourfold in vertebrates, with subsequent loss of two duplicates. Amphioxus Gbx is expressed in all germ layers in the posterior 75% of the embryo, and in the CNS, the Gbx and Otx domains abut at the boundary between the cerebral vesicle (forebrain/midbrain) and the hindbrain. Thus, the genetic machinery to position the MHB was present in the protochordate ancestors of the vertebrates, but is insufficient for induction of organizer genes. Comparison with hemichordates suggests that anterior Otx and posterior Gbx domains were probably overlapping in the ancestral deuterostome and came to abut at the MHB early in the chordate lineage before MHB organizer properties evolved.
Collapse
Affiliation(s)
- L Filipe C Castro
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | | | | | | | | |
Collapse
|
17
|
Holland PWH, Takahashi T. The evolution of homeobox genes: Implications for the study of brain development. Brain Res Bull 2006; 66:484-90. [PMID: 16144637 DOI: 10.1016/j.brainresbull.2005.06.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The homeobox gene superfamily includes many genes implicated in brain development in vertebrates; for example, the Otx, Emx, Dmbx, Gbx, En and Hox gene families. We describe the evolutionary history of the homeobox gene superfamily, as inferred from molecular phylogenetics and chromosomal mapping. Studies of amphioxus, a close relative of vertebrates, have proven particularly informative because it has a genome uncomplicated by recent lineage-specific gene duplications and because in situ hybridisation techniques exist for mapping gene positions and gene expression patterns. We describe an ancient subdivision into gene classes (ANTP, PRD, LIM, POU, SIN, TALE), each containing multiple gene families. The original ANTP class gene duplicated to give distinct NK-like and Hox/ParaHox-related genes, both of which underwent tandem duplication, before the expanding Hox gene cluster duplicated to give Hox and ParaHox clusters. A chromosomal breakage event probably occurred to separate the NK-like and extended Hox genes. Finally, there was additional and extensive gene duplication and gene loss in the vertebrate lineage. We argue that understanding evolutionary history is important for establishing consistent gene nomenclature, and for comparing gene expression patterns and gene functions between species and between gene families.
Collapse
|
18
|
Kimura K, Miki T, Shibasaki T, Zhang Y, Ogawa M, Saisho H, Okuno M, Iwanaga T, Seino S. Functional analysis of transcriptional repressor Otx3/Dmbx1. FEBS Lett 2005; 579:2926-32. [PMID: 15890343 DOI: 10.1016/j.febslet.2005.04.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2004] [Revised: 03/29/2005] [Accepted: 04/14/2005] [Indexed: 11/17/2022]
Abstract
Otx3/Dmbx1 is a member of paired class homeodomain transcription factors. In this study, we found that Otx3/Dmbx1 represses the Otx2-mediated transactivation by forming heterodimer with Otx2 on the P3C (TAATCCGATTA) sequence in vitro. The 156 amino acid region (residues 1-156) of Otx3/Dmbx1 is required for its repressor activity, and interacts directly with Otx2. Co-localization of Otx3/Dmbx1 and Otx2 in brain sections was confirmed by in situ hybridization. These data suggest that Otx3/Dmbx1 represses Otx2-mediated transcription in the developing brain. We also identified the consensus binding sequence [TAATCCGATTA and TAATCC(N2-4)TAATCC] of Otx3/Dmbx1.
Collapse
Affiliation(s)
- Keita Kimura
- Division of Cellular and Molecular Medicine, Kobe University, Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Takahashi T. The evolutionary origins of vertebrate midbrain and MHB: insights from mouse, amphioxus and ascidian Dmbx homeobox genes. Brain Res Bull 2005; 66:510-7. [PMID: 16144640 DOI: 10.1016/j.brainresbull.2005.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Indexed: 12/25/2022]
Abstract
Comparative studies on developmental gene expression suggest that the ancestral chordate central nervous system comprised anterior, midbrain-hindbrain boundary (MHB) and posterior regions. The most anterior region consists of both forebrain and midbrain in vertebrates. It remains, however, unresolved when or how the vertebrate midbrain was established from this anterior zone. I previously reported a mouse PRD-class homeobox gene, Dmbx1, expressed in the presumptive midbrain at early developmental stages, and in the hindbrain at later stages, with exclusion from the MHB. To investigate the evolution of midbrain development, I have cloned Dmbx genes from amphioxus and from Ciona, representing the two most closely related lineages to the vertebrates, and examined embryonic Dmbx expression in these species. In amphioxus, no Dmbx expression is observed in the neural tube, supporting previous arguments that the MHB equivalent region has been secondarily lost in evolution. In Ciona, the CiDmbx gene is detected in neural cells posterior to Pax-2/5/8-positive cells (MHB homologue), but not in any cells anterior to them. These results support the lack of a midbrain homologue in Ciona, and suggest that midbrain development is a vertebrate innovation. Here, I report the full sequences of these genes and discuss the evolution of midbrain development in relation to the tripartite neural ground plan and the origin of the MHB organizer.
Collapse
|
20
|
Hislop NR, de Jong D, Hayward DC, Ball EE, Miller DJ. Tandem organization of independently duplicated homeobox genes in the basal cnidarian Acropora millepora. Dev Genes Evol 2005; 215:268-73. [PMID: 15702325 DOI: 10.1007/s00427-005-0468-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2004] [Accepted: 12/31/2004] [Indexed: 10/25/2022]
Abstract
A number of examples of independently duplicated regulatory genes have been identified in cnidarians, but the extent of this phenomenon and organization of these duplicated genes are unknown. Here we describe the identification of three pairs of independently duplicated homeobox genes in the anthozoan cnidarian, Acropora millepora. In each case, the pairs of paralogous genes are tightly linked, but the extent of sequence divergence implies that these do not reflect recent duplication events. The phenomenon is likely to be more general, as the examples reported here represent most of the limited number of Acropora homeobox genes for which genomic data are yet available.
Collapse
Affiliation(s)
- Nikki R Hislop
- Comparative Genomics Centre, Molecular Sciences Building 21, James Cook University, Townsville, Queensland 4811, Australia
| | | | | | | | | |
Collapse
|
21
|
Takahashi T, Holland PWH. Amphioxus and ascidian Dmbx homeobox genes give clues to the vertebrate origins of midbrain development. Development 2004; 131:3285-94. [PMID: 15201221 DOI: 10.1242/dev.01201] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The ancestral chordate neural tube had a tripartite structure, comprising anterior, midbrain-hindbrain boundary (MHB) and posterior regions. The most anterior region encompasses both forebrain and midbrain in vertebrates. It is not clear when or how the distinction between these two functionally and developmentally distinct regions arose in evolution. Recently, we reported a mouse PRD-class homeobox gene, Dmbx1, expressed in the presumptive midbrain at early developmental stages, and the hindbrain at later stages,with exclusion from the MHB. This gene provides a route to investigate the evolution of midbrain development. We report the cloning, genomic structure,phylogeny and embryonic expression of Dmbx genes from amphioxus and from Ciona, representing the two most closely related lineages to the vertebrates. Our analyses show that Dmbx genes form a distinct, ancient,homeobox gene family, with highly conserved sequence and genomic organisation,albeit more divergent in Ciona. In amphioxus, no Dmbx expression is observed in the neural tube, supporting previous arguments that the MHB equivalent region has been secondarily modified in evolution. In Ciona, the CiDmbx gene is detected in neural cells caudal to Pax2/5/8-positive cells (MHB homologue), in the Hox-positive region, but,interestingly, not in any cells rostral to them. These results suggest that a midbrain homologue is missing in Ciona, and argue that midbrain development is a novelty that evolved specifically on the vertebrate lineage. We discuss the evolution of midbrain development in relation to the ancestry of the tripartite neural ground plan and the origin of the MHB organiser.
Collapse
Affiliation(s)
- Tokiharu Takahashi
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | | |
Collapse
|
22
|
Levantini E, Giorgetti A, Cerisoli F, Traggiai E, Guidi A, Martin R, Acampora D, Aplan PD, Keller G, Simeone A, Iscove NN, Hoang T, Magli MC. Unsuspected role of the brain morphogenetic gene Otx1 in hematopoiesis. Proc Natl Acad Sci U S A 2003; 100:10299-303. [PMID: 12934017 PMCID: PMC193555 DOI: 10.1073/pnas.1734071100] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Indexed: 11/18/2022] Open
Abstract
Otx1 belongs to the paired class of homeobox genes and plays a pivotal role in brain development. Here, we show that Otx1 is expressed in hematopoietic pluripotent and erythroid progenitor cells. Moreover, bone marrow cells from mice lacking Otx1 exhibit a cell-autonomous impairment of the erythroid compartment. In agreement with these results, molecular analysis revealed decreased levels of erythroid genes that include the SCL and GATA-1 transcription factors. Accordingly, a gain of function of SCL rescues the erythroid deficiency in Otx1-/- mice. Taken together, our findings indicate a function for Otx1 in the regulation of blood cell production.
Collapse
Affiliation(s)
- Elena Levantini
- Institute of Biomedical Technologies, Consiglio Nazionale delle Ricerche, 56124 Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Tahayato A, Sonneville R, Pichaud F, Wernet MF, Papatsenko D, Beaufils P, Cook T, Desplan C. Otd/Crx, a dual regulator for the specification of ommatidia subtypes in the Drosophila retina. Dev Cell 2003; 5:391-402. [PMID: 12967559 DOI: 10.1016/s1534-5807(03)00239-9] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Comparison between the inputs of photoreceptors with different spectral sensitivities is required for color vision. In Drosophila, this is achieved in each ommatidium by the inner photoreceptors R7 and R8. Two classes of ommatidia are distributed stochastically in the retina: 30% contain UV-Rh3 in R7 and blue-Rh5 in R8, while the remaining 70% contain UV-Rh4 in R7 and green-Rh6 in R8. We show here that the distinction between the rhodopsins expressed in the two classes of ommatidia depends on a series of highly conserved homeodomain binding sites present in the rhodopsin promoters. The homeoprotein Orthodenticle acts through these sites to activate rh3 and rh5 in their specific ommatidial subclass and through the same sites to prevent rh6 expression in outer photoreceptors. Therefore, Otd is a key player in the terminal differentiation of subtypes of photoreceptors by regulating rhodopsin expression, a function reminiscent of the role of one of its mammalian homologs, Crx, in eye development.
Collapse
Affiliation(s)
- Ali Tahayato
- Department of Biology, New York University, 1009 Silver Building, 100 Washington Square East, New York, NY 10003, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Viczian AS, Vignali R, Zuber ME, Barsacchi G, Harris WA. XOtx5b and XOtx2 regulate photoreceptor and bipolar fates in the Xenopus retina. Development 2003; 130:1281-94. [PMID: 12588845 DOI: 10.1242/dev.00343] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Photoreceptor and bipolar cells are molecularly related cell types in the vertebrate retina. XOtx5b is expressed in both photoreceptors and bipolars, while a closely related member of the same family of transcription factors, XOtx2, is expressed in bipolar cells only. Lipofection of retinal precursors with XOtx5b biases them toward photoreceptor fates whereas a similar experiment with XOtx2 promotes bipolar cell fates. Domain swap experiments show that the ability to specify different cell fates is largely contained in the divergent sequence C-terminal to the homeodomain, while the more homologous N-terminal and homeodomain regions of both genes, when fused to VP16 activators, promote only photoreceptor fates. XOtx5b is closely related to Crx and like Crx it drives expression from an opsin reporter in vivo. XOtx2 suppresses this XOtx5b-driven reporter activity providing a possible explanation for why bipolars do not express opsin. Similarly, co-lipofection of XOtx2 with XOtx5b overrides the latter's ability to promote photoreceptor fates and the combination drives bipolar fates. The results suggest that the shared and divergent parts of these homologous genes may be involved in specifying the shared and distinct characters of related cell types in the vertebrate retina.
Collapse
Affiliation(s)
- Andrea S Viczian
- Department of Anatomy, University of Cambridge, Downing Street, Cambridge, UK
| | | | | | | | | |
Collapse
|