1
|
Shen F, Li J, Li D, Zhou H. Cleft palate, congenital heart disease, and developmental delay involving MEIS2 heterozygous mutations found in the patient with attention deficit hyperactivity disorder: a case report. Front Pediatr 2024; 12:1500152. [PMID: 39776641 PMCID: PMC11703840 DOI: 10.3389/fped.2024.1500152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
This case is the first reported patient with a MEIS2 gene mutation who primarily exhibits pronounced inattention as the main manifestation and is diagnosed with ADHD, requiring methylphenidate treatment. It is characterized by unique clinical features that set it apart from previously reported cases with mutations in the MEIS2 gene. Here, we report a female child with a diagnosis of ADHD and comorbidities. She received treatment with methylphenidate, starting at a dose of 18 milligrams per day, which was gradually increased to 45 milligrams per day based on her attention performance, while also undergoing physical and language rehabilitation training. In addition, the parents involved the child in reading and retelling stories at home every day. After 2 years of treatment, the scale results indicated that the child still had a moderate degree of attention deficit. Therefore, she underwent whole exome sequencing (WES) showing that her MEIS2 gene carries a de novo frameshift mutation (c.934_937del, p. Leu312Argfs*11). After comparing the patient's features with those of other patients who also had the MEIS2 mutation, we discovered that the patient's cleft palate, heart abnormalities, and minor facial dysmorphism were all extremely comparable. A broad forehead, elongated and arched eyebrows, and a tent-shaped upper lip were examples of mild facial dysmorphic traits. Subtypes with phenotypes such as cleft palate, cardiac anomalies, or facial malformations were presented in all previously reported cases of MEIS2 mutations. Furthermore, less common characteristics include ADHD, learning difficulties, hearing loss, recurring respiratory infections, asthma, rhinitis, enuresis, and dental cavities. This case further supports the critical role of genetic testing in patients with ADHD who exhibit a suboptimal response to methylphenidate and present with multiple comorbidities. Furthermore, this case report expands the clinical symptom spectrum associated with MEIS2 gene mutations, providing a broader understanding of the condition.
Collapse
Affiliation(s)
- Fang Shen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junyan Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dandan Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Müller T, Reichlmeir M, Hau AC, Wittig I, Schulte D. The neuronal transcription factor MEIS2 is a calpain-2 protease target. J Cell Sci 2024; 137:jcs261482. [PMID: 38305737 PMCID: PMC10941658 DOI: 10.1242/jcs.261482] [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: 07/12/2023] [Accepted: 01/25/2024] [Indexed: 02/03/2024] Open
Abstract
Tight control over transcription factor activity is necessary for a sensible balance between cellular proliferation and differentiation in the embryo and during tissue homeostasis by adult stem cells, but mechanistic details have remained incomplete. The homeodomain transcription factor MEIS2 is an important regulator of neurogenesis in the ventricular-subventricular zone (V-SVZ) adult stem cell niche in mice. We here identify MEIS2 as direct target of the intracellular protease calpain-2 (composed of the catalytic subunit CAPN2 and the regulatory subunit CAPNS1). Phosphorylation at conserved serine and/or threonine residues, or dimerization with PBX1, reduced the sensitivity of MEIS2 towards cleavage by calpain-2. In the adult V-SVZ, calpain-2 activity is high in stem and progenitor cells, but rapidly declines during neuronal differentiation, which is accompanied by increased stability of MEIS2 full-length protein. In accordance with this, blocking calpain-2 activity in stem and progenitor cells, or overexpression of a cleavage-insensitive form of MEIS2, increased the production of neurons, whereas overexpression of a catalytically active CAPN2 reduced it. Collectively, our results support a key role for calpain-2 in controlling the output of adult V-SVZ neural stem and progenitor cells through cleavage of the neuronal fate determinant MEIS2.
Collapse
Affiliation(s)
- Tanja Müller
- Goethe University, Faculty of Medicine, University Hospital Frankfurt, Institute of Neurology (Edinger Institute), 60528 Frankfurt, Germany
- Goethe University, University Hospital Frankfurt, Dr. Senckenberg Institute of Neurooncology and Institute of Neurology (Edinger Institute), Frankfurt Cancer Institute (FCI), University Cancer Center Frankfurt (UCT), MSNZ Junior Group Translational Neurooncology, 60528 Frankfurt, Germany
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Luxembourg Centre of Neuropathology (LCNP), 1445 Luxembourg, Luxembourg
| | - Marina Reichlmeir
- Goethe University, Faculty of Medicine, University Hospital Frankfurt, Institute of Neurology (Edinger Institute), 60528 Frankfurt, Germany
| | - Ann-Christin Hau
- Goethe University, University Hospital Frankfurt, Dr. Senckenberg Institute of Neurooncology and Institute of Neurology (Edinger Institute), Frankfurt Cancer Institute (FCI), University Cancer Center Frankfurt (UCT), MSNZ Junior Group Translational Neurooncology, 60528 Frankfurt, Germany
| | - Ilka Wittig
- Goethe University, Faculty of Medicine, Institute for Cardiovascular Physiology, Functional Proteomics, 60590, Frankfurt, Germany
| | - Dorothea Schulte
- Goethe University, Faculty of Medicine, University Hospital Frankfurt, Institute of Neurology (Edinger Institute), 60528 Frankfurt, Germany
| |
Collapse
|
3
|
Lee Q, Chan WC, Qu X, Sun Y, Abdelkarim H, Le J, Saqib U, Sun MY, Kruse K, Banerjee A, Hitchinson B, Geyer M, Huang F, Guaiquil V, Mutso AA, Sanders M, Rosenblatt MI, Maienschein-Cline M, Lawrence MS, Gaponenko V, Malik AB, Komarova YA. End binding-3 inhibitor activates regenerative program in age-related macular degeneration. Cell Rep Med 2023; 4:101223. [PMID: 37794584 PMCID: PMC10591057 DOI: 10.1016/j.xcrm.2023.101223] [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: 08/22/2020] [Revised: 07/19/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
Wet age-related macular degeneration (AMD), characterized by leaky neovessels emanating from the choroid, is a main cause of blindness. As current treatments for wet AMD require regular intravitreal injections of anti-vascular endothelial growth factor (VEGF) biologics, there is a need for the development of less invasive treatments. Here, we designed an allosteric inhibitor of end binding-3 (EB3) protein, termed EBIN, which reduces the effects of environmental stresses on endothelial cells by limiting pathological calcium signaling. Delivery of EBIN via eye drops in mouse and non-human primate (NHP) models of wet AMD prevents both neovascular leakage and choroidal neovascularization. EBIN reverses the epigenetic changes induced by environmental stresses, allowing an activation of a regenerative program within metabolic-active endothelial cells comprising choroidal neovascularization (CNV) lesions. These results suggest the therapeutic potential of EBIN in preventing the degenerative processes underlying wet AMD.
Collapse
Affiliation(s)
- Quinn Lee
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Wan Ching Chan
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Xinyan Qu
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Ying Sun
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | - Jonathan Le
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Uzma Saqib
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Mitchell Y Sun
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Kevin Kruse
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Avik Banerjee
- Department of Chemistry, The University of Illinois, Chicago, IL 60612, USA
| | - Ben Hitchinson
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Melissa Geyer
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Fei Huang
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Victor Guaiquil
- Department of Ophthalmology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Amelia A Mutso
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | - Mark I Rosenblatt
- Department of Ophthalmology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Asrar B Malik
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yulia A Komarova
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
4
|
Angueyra JM, Kunze VP, Patak LK, Kim H, Kindt K, Li W. Transcription factors underlying photoreceptor diversity. eLife 2023; 12:e81579. [PMID: 36745553 PMCID: PMC9901936 DOI: 10.7554/elife.81579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/22/2023] [Indexed: 02/07/2023] Open
Abstract
During development, retinal progenitors navigate a complex landscape of fate decisions to generate the major cell classes necessary for proper vision. Transcriptional regulation is critical to generate diversity within these major cell classes. Here, we aim to provide the resources and techniques required to identify transcription factors necessary to generate and maintain diversity in photoreceptor subtypes, which are critical for vision. First, we generate a key resource: a high-quality and deep transcriptomic profile of each photoreceptor subtype in adult zebrafish. We make this resource openly accessible, easy to explore, and have integrated it with other currently available photoreceptor transcriptomic datasets. Second, using our transcriptomic profiles, we derive an in-depth map of expression of transcription factors in photoreceptors. Third, we use efficient CRISPR-Cas9 based mutagenesis to screen for null phenotypes in F0 larvae (F0 screening) as a fast, efficient, and versatile technique to assess the involvement of candidate transcription factors in the generation of photoreceptor subtypes. We first show that known phenotypes can be easily replicated using this method: loss of S cones in foxq2 mutants and loss of rods in nr2e3 mutants. We then identify novel functions for the transcription factor Tbx2, demonstrating that it plays distinct roles in controlling the generation of all photoreceptor subtypes within the retina. Our study provides a roadmap to discover additional factors involved in this process. Additionally, we explore four transcription factors of unknown function (Skor1a, Sall1a, Lrrfip1a, and Xbp1), and find no evidence for their involvement in the generation of photoreceptor subtypes. This dataset and screening method will be a valuable way to explore the genes involved in many other essential aspects of photoreceptor biology.
Collapse
Affiliation(s)
- Juan M Angueyra
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Vincent P Kunze
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Laura K Patak
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Hailey Kim
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Wei Li
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
5
|
Safgren SL, Olson RJ, Pinto E Vairo F, Bothun ED, Hanna C, Klee EW, Schimmenti LA. De novo PBX1 variant in a patient with glaucoma, kidney anomalies, and developmental delay: An expansion of the CAKUTHED phenotype. Am J Med Genet A 2022; 188:919-925. [PMID: 34797033 DOI: 10.1002/ajmg.a.62576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/05/2021] [Accepted: 11/02/2021] [Indexed: 01/22/2023]
Abstract
An infant was referred for evaluation of congenital glaucoma and corneal clouding. In addition, he had a pelvic kidney, hypotonia, patent ductus arteriosus, abnormal pinnae, and developmental delay. Exome sequencing identified a previously unpublished de novo single nucleotide insertion in PBX1 c.400dupG (NM_002585.3), predicted to cause a frameshift resulting in a truncated protein with loss of function (p.Ala134Glyfs*65). Identification of this loss of function variant supports the diagnosis of congenital anomalies of the kidney and urinary tract syndrome with or without hearing loss, abnormal ears, or developmental delay (CAKUTHED). Here, we propose glaucoma as an extra-renal manifestation associated with PBX1-related disease due to the relationship of PBX1 with MEIS1, MEIS2, and FOXC1 transcription factors associated with eye development.
Collapse
Affiliation(s)
- Stephanie L Safgren
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rory J Olson
- Center of Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Filippo Pinto E Vairo
- Center of Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Erick D Bothun
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Christian Hanna
- Department of Pediatric Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric W Klee
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, Minnesota, USA
- Center of Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Lisa A Schimmenti
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Otorhinolaryngology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
6
|
Huang M, Xu L, Liu J, Huang P, Tan Y, Chen S. Cell–Cell Communication Alterations via Intercellular Signaling Pathways in Substantia Nigra of Parkinson’s Disease. Front Aging Neurosci 2022; 14:828457. [PMID: 35283752 PMCID: PMC8914319 DOI: 10.3389/fnagi.2022.828457] [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] [Received: 12/03/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative movement disorder characterized with dopaminergic neuron (DaN) loss within the substantia nigra (SN). Despite bulk studies focusing on intracellular mechanisms of PD inside DaNs, few studies have explored the pathogeneses outside DaNs, or between DaNs and other cells. Here, we set out to probe the implication of intercellular communication involving DaNs in the pathogeneses of PD at a systemic level with bioinformatics methods. We harvested three online published single-cell/single-nucleus transcriptomic sequencing (sc/snRNA-seq) datasets of human SN (GSE126838, GSE140231, and GSE157783) from the Gene Expression Omnibus (GEO) database, and integrated them with one of the latest integration algorithms called Harmony. We then applied CellChat, the latest cell–cell communication analytic algorithm, to our integrated dataset. We first found that the overall communication quantity was decreased while the overall communication strength was enhanced in PD sample compared with control sample. We then focused on the intercellular communication where DaNs are involved, and found that the communications between DaNs and other cell types via certain signaling pathways were selectively altered in PD, including some growth factors, neurotrophic factors, chemokines, etc. pathways. Our bioinformatics analysis showed that the alteration in intercellular communications involving DaNs might be a previously underestimated aspect of PD pathogeneses with novel translational potential.
Collapse
Affiliation(s)
- Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Xu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuyan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yuyan Tan,
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies, Shanghai Tech University, Shanghai, China
- Shengdi Chen,
| |
Collapse
|
7
|
Lam DD, Nikolic AA, Zhao C, Mirza-Schreiber N, Krężel W, Oexle K, Winkelmann J. Intronic elements associated with insomnia and restless legs syndrome exhibit cell type-specific epigenetic features contributing to MEIS1 regulation. Hum Mol Genet 2021; 31:1733-1746. [PMID: 34888668 DOI: 10.1093/hmg/ddab355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/25/2021] [Accepted: 12/05/2021] [Indexed: 12/13/2022] Open
Abstract
A highly evolutionarily conserved MEIS1 intronic region is strongly associated with restless legs syndrome (RLS) and insomnia. To understand its regulatory function, we dissected the region by analyzing chromatin accessibility, enhancer-promoter contacts, DNA methylation, and eQTLs in different human neural cell types and tissues. We observed specific activity with respect to cell type and developmental maturation, indicating a prominent role for distinct highly conserved intronic elements in forebrain inhibitory neuron differentiation. Two elements were hypomethylated in neural cells with higher MEIS1 expression, suggesting a role of enhancer demethylation in gene regulation. MEIS1 eQTLs showed a striking modular chromosomal distribution, with forebrain eQTLs clustering in intron 8/9. CRISPR interference targeting of individual elements in this region attenuated MEIS1 expression, revealing a complex regulatory interplay of distinct elements. In summary, we found that MEIS1 regulation is organized in a modular pattern. Disease-associated intronic regulatory elements control MEIS1 expression with cell type and maturation stage specificity, particularly in the inhibitory neuron lineage. The precise spatiotemporal activity of these elements likely contributes to the pathogenesis of insomnia and RLS.
Collapse
Affiliation(s)
- Daniel D Lam
- Institute of Neurogenomics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
| | - Ana Antic Nikolic
- Institute of Neurogenomics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
| | - Chen Zhao
- Institute of Neurogenomics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
| | - Nazanin Mirza-Schreiber
- Institute of Neurogenomics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Wojciech Krężel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Konrad Oexle
- Institute of Neurogenomics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany.,Chair of Neurogenetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
8
|
Abstract
The vertebrate eye is derived from the neuroepithelium, surface ectoderm, and extracellular mesenchyme. The neuroepithelium forms an optic cup in which the spatial separation of three domains is established, namely, the region of multipotent retinal progenitor cells (RPCs), the ciliary margin zone (CMZ)-which possesses both a neurogenic and nonneurogenic potential-and the optic disk (OD), the interface between the optic stalk and the neuroretina. Here, we show by genetic ablation in the developing optic cup that Meis1 and Meis2 homeobox genes function redundantly to maintain the retinal progenitor pool while they simultaneously suppress the expression of genes characteristic of CMZ and OD fates. Furthermore, we demonstrate that Meis transcription factors bind regulatory regions of RPC-, CMZ-, and OD-specific genes, thus providing a mechanistic insight into the Meis-dependent gene regulatory network. Our work uncovers the essential role of Meis1 and Meis2 as regulators of cell fate competence, which organize spatial territories in the vertebrate eye.
Collapse
|
9
|
D’Aniello E, Paganos P, Anishchenko E, D’Aniello S, Arnone MI. Comparative Neurobiology of Biogenic Amines in Animal Models in Deuterostomes. Front Ecol Evol 2020. [DOI: 10.3389/fevo.2020.587036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
10
|
Liu H, Barnes J, Pedrosa E, Herman NS, Salas F, Wang P, Zheng D, Lachman HM. Transcriptome analysis of neural progenitor cells derived from Lowe syndrome induced pluripotent stem cells: identification of candidate genes for the neurodevelopmental and eye manifestations. J Neurodev Disord 2020; 12:14. [PMID: 32393163 PMCID: PMC7212686 DOI: 10.1186/s11689-020-09317-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Lowe syndrome (LS) is caused by loss-of-function mutations in the X-linked gene OCRL, which codes for an inositol polyphosphate 5-phosphatase that plays a key role in endosome recycling, clathrin-coated pit formation, and actin polymerization. It is characterized by congenital cataracts, intellectual and developmental disability, and renal proximal tubular dysfunction. Patients are also at high risk for developing glaucoma and seizures. We recently developed induced pluripotent stem cell (iPSC) lines from three patients with LS who have hypomorphic variants affecting the 3' end of the gene, and their neurotypical brothers to serve as controls. METHODS In this study, we used RNA sequencing (RNA-seq) to obtain transcriptome profiles in LS and control neural progenitor cells (NPCs). RESULTS In a comparison of the patient and control NPCs (n = 3), we found 16 differentially expressed genes (DEGs) at the multiple test adjusted p value (padj) < 0.1, with nine at padj < 0.05. Using nominal p value < 0.05, 319 DEGs were detected. The relatively small number of DEGs could be due to the fact that OCRL is not a transcription factor per se, although it could have secondary effects on gene expression through several different mechanisms. Although the number of DEGs passing multiple test correction was small, those that were found are quite consistent with some of the known molecular effects of OCRL protein, and the clinical manifestations of LS. Furthermore, using gene set enrichment analysis (GSEA), we found that genes increased expression in the patient NPCs showed enrichments of several gene ontology (GO) terms (false discovery rate < 0.25): telencephalon development, pallium development, NPC proliferation, and cortex development, which are consistent with a condition characterized by intellectual disabilities and psychiatric manifestations. In addition, a significant enrichment among the nominal DEGs for genes implicated in autism spectrum disorder (ASD) was found (e.g., AFF2, DNER, DPP6, DPP10, RELN, CACNA1C), as well as several that are strong candidate genes for the development of eye problems found in LS, including glaucoma. The most notable example is EFEMP1, a well-known candidate gene for glaucoma and other eye pathologies. CONCLUSION Overall, the RNA-seq findings present several candidate genes that could help explain the underlying basis for the neurodevelopmental and eye problems seen in boys with LS.
Collapse
Affiliation(s)
- Hequn Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jesse Barnes
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nathaniel S. Herman
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Franklin Salas
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
11
|
Markitantova Y, Simirskii V. Inherited Eye Diseases with Retinal Manifestations through the Eyes of Homeobox Genes. Int J Mol Sci 2020; 21:E1602. [PMID: 32111086 PMCID: PMC7084737 DOI: 10.3390/ijms21051602] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Retinal development is under the coordinated control of overlapping networks of signaling pathways and transcription factors. The paper was conceived as a review of the data and ideas that have been formed to date on homeobox genes mutations that lead to the disruption of eye organogenesis and result in inherited eye/retinal diseases. Many of these diseases are part of the same clinical spectrum and have high genetic heterogeneity with already identified associated genes. We summarize the known key regulators of eye development, with a focus on the homeobox genes associated with monogenic eye diseases showing retinal manifestations. Recent advances in the field of genetics and high-throughput next-generation sequencing technologies, including single-cell transcriptome analysis have allowed for deepening of knowledge of the genetic basis of inherited retinal diseases (IRDs), as well as improve their diagnostics. We highlight some promising avenues of research involving molecular-genetic and cell-technology approaches that can be effective for IRDs therapy. The most promising neuroprotective strategies are aimed at mobilizing the endogenous cellular reserve of the retina.
Collapse
|
12
|
Gulyaeva O, Nguyen H, Sambeat A, Heydari K, Sul HS. Sox9-Meis1 Inactivation Is Required for Adipogenesis, Advancing Pref-1 + to PDGFRα + Cells. Cell Rep 2019; 25:1002-1017.e4. [PMID: 30355480 PMCID: PMC6903418 DOI: 10.1016/j.celrep.2018.09.086] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/27/2018] [Accepted: 09/26/2018] [Indexed: 12/28/2022] Open
Abstract
Adipocytes arise from the commitment and differentiation of adipose precursors in white adipose tissue (WAT). In studying adipogenesis, precursor markers, including Pref-1 and PDGFRα, are used to isolate precursors from stromal vascular fractions of WAT, but the relation among the markers is not known. Here, we used the Pref-1 promoter-rtTA system in mice for labeling Pref-1+ cells and for inducible inactivation of the Pref-1 target Sox9. We show the requirement of Sox9 for the maintenance of Pref-1+ proliferative, early precursors. Upon Sox9 inactivation, these Pref-1+ cells become PDGFRα+ cells that express early adipogenic markers. Thus, we show that Pref-1+ cells precede PDGFRα+ cells in the adipogenic pathway and that Sox9 inactivation is required for WAT growth and expansion. Furthermore, we show that in maintaining early adipose precursors, Sox9 activates Meis1, which prevents adipogenic differentiation. Our study also demonstrates the Pref-1 promoter-rtTA system for inducible gene inactivation in early adipose precursor populations. The relationship among Sox9+, Pref-1+, and PDGFRα+ WAT precursors has not been studied. Gulyaeva et al. show that Pref-1+ cells are early adipose precursors and, upon Sox9 inactivation, they become PDGFRα+ cells at a later stage of the adipogenic pathway. In maintaining Pref-1+ adipose precursors, Sox9 activates Meis1, which prevents adipogenic differentiation.
Collapse
Affiliation(s)
- Olga Gulyaeva
- Endocrinology Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hai Nguyen
- Endocrinology Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Audrey Sambeat
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kartoosh Heydari
- Cancer Research Laboratory, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hei Sook Sul
- Endocrinology Program, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
13
|
Schulte D, Geerts D. MEIS transcription factors in development and disease. Development 2019; 146:146/16/dev174706. [PMID: 31416930 DOI: 10.1242/dev.174706] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/28/2019] [Indexed: 12/12/2022]
Abstract
MEIS transcription factors are key regulators of embryonic development and cancer. Research on MEIS genes in the embryo and in stem cell systems has revealed novel and surprising mechanisms by which these proteins control gene expression. This Primer summarizes recent findings about MEIS protein activity and regulation in development, and discusses new insights into the role of MEIS genes in disease, focusing on the pathogenesis of solid cancers.
Collapse
Affiliation(s)
- Dorothea Schulte
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany
| | - Dirk Geerts
- Department of Medical Biology L2-109, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
14
|
Abstract
The use of retinal organoids requires efficient differentiation from induced pluripotent stem cells (iPSCs). In this issue of Cell Reports, Wang et al. (2018) examine how the chromatin landscape after iPSC programming predicts their ability to differentiate into retinal tissue.
Collapse
|
15
|
Wang L, Hiler D, Xu B, AlDiri I, Chen X, Zhou X, Griffiths L, Valentine M, Shirinifard A, Sablauer A, Thiagarajan S, Barabas ME, Zhang J, Johnson D, Frase S, Dyer MA. Retinal Cell Type DNA Methylation and Histone Modifications Predict Reprogramming Efficiency and Retinogenesis in 3D Organoid Cultures. Cell Rep 2019. [PMID: 29514090 PMCID: PMC5872828 DOI: 10.1016/j.celrep.2018.01.075] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diverse cell types can be reprogrammed into pluripotent stem cells by ectopic expression of Oct4 (Pou5f1), Klf4, Sox3, and Myc. Many of these induced pluripotent stem cells (iPSCs) retain memory, in terms of DNA methylation and histone modifications (epigenetic memory), of their cellular origins, and this may bias subsequent differentiation. Neurons are difficult to reprogram, and there has not been a systematic side-by-side characterization of reprogramming efficiency or epigenetic memory across different neuronal subtypes. Here, we compare reprogramming efficiency of five different retinal cell types at two different stages of development. Retinal differentiation from each iPSC line was measured using a quantitative standardized scoring system called STEM-RET and compared to the epigenetic memory. Neurons with the lowest reprogramming efficiency produced iPSC lines with the best retinal differentiation and were more likely to retain epigenetic memory of their cellular origins. In addition, we identified biomarkers of iPSCs that are predictive of retinal differentiation.
Collapse
Affiliation(s)
- Lu Wang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Daniel Hiler
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Issam AlDiri
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lyra Griffiths
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marc Valentine
- Cytogenetics Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - András Sablauer
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Suresh Thiagarajan
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marie-Elizabeth Barabas
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiakun Zhang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dianna Johnson
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sharon Frase
- Cell and Tissue Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
16
|
Liang CL, Hsu PY, Ngo CS, Seow WJ, Karnani N, Pan H, Saw SM, Juo SHH. HOXA9 is a novel myopia risk gene. BMC Ophthalmol 2019; 19:28. [PMID: 30674274 PMCID: PMC6343304 DOI: 10.1186/s12886-019-1038-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 01/15/2019] [Indexed: 11/10/2022] Open
Abstract
Purpose A recent meta-analysis revealed PAX6 as a risk gene for myopia. There is a link between PAX6 and HOXA9. Furthermore, HOXA9 has been reported to activate TGF-β that is a risk factor for myopia. We speculate HOXA9 may participate in myopia development. Methods The Singapore GUSTO birth cohort provides data on children’s cycloplegic refraction measured at age of 3 years and their methylation profile based on the umbilical cord DNA. The HOXA9 expression levels were measured in the eyes of mono-ocular form deprivation myopia in mice. The plasmid with the mouse HOXA9 cDNA was constructed and then transfected to mouse primary retinal pigment epithelial (RPE) cells. The expression levels of myopia-related genes and cell proliferation were measured in the HOXA9-overexpressed RPE cells. Results A total of 519 children had data on methylation profile and cycloplegic refraction. The mean spherical equivalent refraction (SE) was 0.90D. Among 8 SE outliers (worse than -2D), 7 children had HOXA9 hypomethylation. The HOXA9 levels in the retina of myopic eyes was 2.65-fold (p = 0.029; paired t-test) higher than the uncovered fellow eyes. When HOXA9 was over-expressed in the RPE cells, TGF-β, MMP2, FGF2 and IGF1R expression levels were dose-dependently increased by HOXA9. However, over-expression of HOXA9 had no significant influence on IGF1 or HGF expression. In addition, HOXA9 also increased RPE proliferation. Conclusion Based on the human, animal and cellular data, the transcription factor HOXA9 may promote the expression of pro-myopia genes and RPE proliferation, which eventually contribute to myopia development.
Collapse
Affiliation(s)
- Chung-Ling Liang
- Department of Ophthalmology, Asia University Hospital, Taichung, Taiwan.,Department of Optometry, College of Medical and Health Science, Asia University, Taichung, Taiwan.,Center for Myopia and Eye Disease, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,Bright-Eyes Clinic, Kaohsiung, Taiwan
| | - Po-Yuan Hsu
- Center for Myopia and Eye Disease, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Cheryl S Ngo
- Department of Ophthalmology, National University Hospital, Singapore, Singapore
| | - Wei Jie Seow
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Hong Pan
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Seang-Mei Saw
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore
| | - Suh-Hang H Juo
- Center for Myopia and Eye Disease, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan. .,The Ophthalmology & Visual Sciences Academic Clinical Program, DUKE-NUS Graduate Medical School, Singapore, Singapore. .,Graduate Institute of Biomedical Sciences, Singapore, Singapore. .,Institute of New Drug Development, Singapore, Singapore. .,Drug Development Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
17
|
Liu Z, Sun C, Yan Y, Li G, Wu G, Liu A, Yang N. Genome-Wide Association Analysis of Age-Dependent Egg Weights in Chickens. Front Genet 2018; 9:128. [PMID: 29755503 PMCID: PMC5932955 DOI: 10.3389/fgene.2018.00128] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/29/2018] [Indexed: 12/22/2022] Open
Abstract
Egg weight (EW) is an economically-important trait and displays a consecutive increase with the hen's age. Because extremely large eggs cause a range of problems in the poultry industry, we performed a genome-wide association study (GWAS) in order to identify genomic variations that are associated with EW. We utilized the Affymetrix 600 K high density SNP array in a population of 1,078 hens at seven time points from day at first egg to 80 weeks age (EW80). Results reveal that a 90 Kb genomic region (169.42 Mb ~ 169.51 Mb) in GGA1 is significantly associated with EW36 and is also potentially associated with egg weight at 28, 56, and 66 week of age. The leading SNP could account for 3.66% of the phenotypic variation, while two promising genes (DLEU7 and MIR15A) can be mapped to this narrow significant region and may affect EW in a pleiotropic manner. In addition, one gene (CECR2 on GGA1) and two genes (MEIS1 and SPRED2 on GGA3), which involved in the processes of embryogenesis and organogenesis, were also considered to be candidates related to first egg weight (FEW) and EW56, respectively. Findings in our study could provide worthy theoretical basis to generate eggs of ideal size based on marker assisted breeding selection.
Collapse
Affiliation(s)
- Zhuang Liu
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yiyuan Yan
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Beijing Engineering Research Center of Layer, Beijing, China
| | - Guangqi Li
- Beijing Engineering Research Center of Layer, Beijing, China
| | - Guiqin Wu
- Beijing Engineering Research Center of Layer, Beijing, China
| | - Aiqiao Liu
- Beijing Engineering Research Center of Layer, Beijing, China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
18
|
Meis1 Coordinates Cerebellar Granule Cell Development by Regulating Pax6 Transcription, BMP Signaling and Atoh1 Degradation. J Neurosci 2018; 38:1277-1294. [PMID: 29317485 DOI: 10.1523/jneurosci.1545-17.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 11/21/2022] Open
Abstract
Cerebellar granule cell precursors (GCPs) and granule cells (GCs) represent good models to study neuronal development. Here, we report that the transcription factor myeloid ectopic viral integration site 1 homolog (Meis1) plays pivotal roles in the regulation of mouse GC development. We found that Meis1 is expressed in GC lineage cells and astrocytes in the cerebellum during development. Targeted disruption of the Meis1 gene specifically in the GC lineage resulted in smaller cerebella with disorganized lobules. Knock-down/knock-out (KO) experiments for Meis1 and in vitro assays showed that Meis1 binds to an upstream sequence of Pax6 to enhance its transcription in GCPs/GCs and also suggested that the Meis1-Pax6 cascade regulates morphology of GCPs/GCs during development. In the conditional KO (cKO) cerebella, many Atoh1-positive GCPs were observed ectopically in the inner external granule layer (EGL) and a similar phenomenon was observed in cultured cerebellar slices treated with a bone morphogenic protein (BMP) inhibitor. Furthermore, expression of Smad proteins and Smad phosphorylation were severely reduced in the cKO cerebella and Meis1-knock-down GCPs cerebella. Reduction of phosphorylated Smad was also observed in cerebellar slices electroporated with a Pax6 knock-down vector. Because it is known that BMP signaling induces Atoh1 degradation in GCPs, these findings suggest that the Meis1-Pax6 pathway increases the expression of Smad proteins to upregulate BMP signaling, leading to degradation of Atoh1 in the inner EGL, which contributes to differentiation from GCPs to GCs. Therefore, this work reveals crucial functions of Meis1 in GC development and gives insights into the general understanding of the molecular machinery underlying neural differentiation from neural progenitors.SIGNIFICANCE STATEMENT We report that myeloid ectopic viral integration site 1 homolog (Meis1) plays pivotal roles in the regulation of mouse granule cell (GC) development. Here, we show Meis1 is expressed in GC precursors (GCPs) and GCs during development. Our knock-down and conditional knock-out (cKO) experiments and in vitro assays revealed that Meis1 is required for proper cerebellar structure formation and for Pax6 transcription in GCPs and GCs. The Meis1-Pax6 cascade regulates the morphology of GCs. In the cKO cerebella, Smad proteins and bone morphogenic protein (BMP) signaling are severely reduced and Atoh1-expressing GCPs are ectopically detected in the inner external granule layer. These findings suggest that Meis1 regulates degradation of Atoh1 via BMP signaling, contributing to GC differentiation in the inner EGL, and should provide understanding into GC development.
Collapse
|
19
|
Fernandes-Silva H, Vaz-Cunha P, Barbosa VB, Silva-Gonçalves C, Correia-Pinto J, Moura RS. Retinoic acid regulates avian lung branching through a molecular network. Cell Mol Life Sci 2017; 74:4599-4619. [PMID: 28735443 PMCID: PMC11107646 DOI: 10.1007/s00018-017-2600-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 07/04/2017] [Accepted: 07/18/2017] [Indexed: 12/14/2022]
Abstract
Retinoic acid (RA) is of major importance during vertebrate embryonic development and its levels need to be strictly regulated otherwise congenital malformations will develop. Through the action of specific nuclear receptors, named RAR/RXR, RA regulates the expression of genes that eventually influence proliferation and tissue patterning. RA has been described as crucial for different stages of mammalian lung morphogenesis, and as part of a complex molecular network that contributes to precise organogenesis; nonetheless, nothing is known about its role in avian lung development. The current report characterizes, for the first time, the expression pattern of RA signaling members (stra6, raldh2, raldh3, cyp26a1, rarα, and rarβ) and potential RA downstream targets (sox2, sox9, meis1, meis2, tgfβ2, and id2) by in situ hybridization. In the attempt of unveiling the role of RA in chick lung branching, in vitro lung explants were performed. Supplementation studies revealed that RA stimulates lung branching in a dose-dependent manner. Moreover, the expression levels of cyp26a1, sox2, sox9, rarβ, meis2, hoxb5, tgfβ2, id2, fgf10, fgfr2, and shh were evaluated after RA treatment to disclose a putative molecular network underlying RA effect. In situ hybridization analysis showed that RA is able to alter cyp26a1, sox9, tgfβ2, and id2 spatial distribution; to increase rarβ, meis2, and hoxb5 expression levels; and has a very modest effect on sox2, fgf10, fgfr2, and shh expression levels. Overall, these findings support a role for RA in the proximal-distal patterning and branching morphogenesis of the avian lung and reveal intricate molecular interactions that ultimately orchestrate branching morphogenesis.
Collapse
Affiliation(s)
- Hugo Fernandes-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Patrícia Vaz-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Violina Baranauskaite Barbosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Carla Silva-Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Jorge Correia-Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
- Department of Pediatric Surgery, Hospital de Braga, 4710-243, Braga, Portugal
| | - Rute Silva Moura
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.
- ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
- Biology Department, School of Sciences, University of Minho, 4710-057, Braga, Portugal.
| |
Collapse
|
20
|
Lai CK, Norddahl GL, Maetzig T, Rosten P, Lohr T, Sanchez Milde L, von Krosigk N, Docking TR, Heuser M, Karsan A, Humphries RK. Meis2 as a critical player in MN1-induced leukemia. Blood Cancer J 2017; 7:e613. [PMID: 28960191 PMCID: PMC5709755 DOI: 10.1038/bcj.2017.86] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 01/05/2023] Open
Abstract
Meningioma 1 (MN1) is an independent prognostic marker for normal karyotype acute myeloid leukemia (AML), with high expression linked to all-trans retinoic acid resistance and poor survival. MN1 is also a potent and sufficient oncogene in murine leukemia models, strongly dependent on the MEIS1/AbdB-like HOX protein complex to transform common myeloid progenitors, block myeloid differentiation, and promote leukemic stem cell self-renewal. To identify key genes and pathways underlying leukemic activity, we functionally assessed MN1 cell phenotypic heterogeneity, revealing leukemic and non-leukemic subsets. Using gene expression profiling of these subsets combined with previously published comparisons of full-length MN1 and mutants with varying leukemogenic activity, we identified candidate genes critical to leukemia. Functional analysis identified Hlf and Hoxa9 as critical to MN1 in vitro proliferation, self-renewal and impaired myeloid differentiation. Although critical to transformation, Meis1 knockdown had little impact on these properties in vitro. However, we identified Meis2 as critical to MN1-induced leukemia, with essential roles in proliferation, self-renewal, impairment of differentiation and disease progression in vitro and in vivo. Here, we provide evidence of phenotypic and functional hierarchy in MN1-induced leukemic cells, characterise contributions of Hlf, Hoxa9 and Meis1 to in vitro leukemic properties, and reveal Meis2 as a novel player in MN1-induced leukemogenesis.
Collapse
Affiliation(s)
- C K Lai
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - G L Norddahl
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - T Maetzig
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - P Rosten
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - T Lohr
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - L Sanchez Milde
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - N von Krosigk
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - T R Docking
- Genome Sciences Centre, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - M Heuser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - A Karsan
- Genome Sciences Centre, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - R K Humphries
- Terry Fox Laboratory, BC Cancer Agency Research Centre, Vancouver, British Columbia, Canada.,Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
Teotia P, Van Hook MJ, Wichman CS, Allingham RR, Hauser MA, Ahmad I. Modeling Glaucoma: Retinal Ganglion Cells Generated from Induced Pluripotent Stem Cells of Patients with SIX6 Risk Allele Show Developmental Abnormalities. Stem Cells 2017; 35:2239-2252. [PMID: 28792678 DOI: 10.1002/stem.2675] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 01/01/2023]
Abstract
Glaucoma represents a group of multifactorial diseases with a unifying pathology of progressive retinal ganglion cell (RGC) degeneration, causing irreversible vision loss. To test the hypothesis that RGCs are intrinsically vulnerable in glaucoma, we have developed an in vitro model using the SIX6 risk allele carrying glaucoma patient-specific induced pluripotent stem cells (iPSCs) for generating functional RGCs. Here, we demonstrate that the efficiency of RGC generation by SIX6 risk allele iPSCs is significantly lower than iPSCs-derived from healthy, age- and sex-matched controls. The decrease in the number of RGC generation is accompanied by repressed developmental expression of RGC regulatory genes. The SIX6 risk allele RGCs display short and simple neurites, reduced expression of guidance molecules, and immature electrophysiological signature. In addition, these cells have higher expression of glaucoma-associated genes, CDKN2A and CDKN2B, suggesting an early onset of the disease phenotype. Consistent with the developmental abnormalities, the SIX6 risk allele RGCs display global dysregulation of genes which map on developmentally relevant biological processes for RGC differentiation and signaling pathways such as mammalian target of rapamycin that integrate diverse functions for differentiation, metabolism, and survival. The results suggest that SIX6 influences different stages of RGC differentiation and their survival; therefore, alteration in SIX6 function due to the risk allele may lead to cellular and molecular abnormalities. These abnormalities, if carried into adulthood, may make RGCs vulnerable in glaucoma. Stem Cells 2017;35:2239-2252.
Collapse
Affiliation(s)
- Pooja Teotia
- Department of Ophthalmology and Visual Sciences, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew J Van Hook
- Department of Ophthalmology and Visual Sciences, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Christopher S Wichman
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - R Rand Allingham
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Michael A Hauser
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Iqbal Ahmad
- Department of Ophthalmology and Visual Sciences, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
22
|
Neto M, Naval-Sánchez M, Potier D, Pereira PS, Geerts D, Aerts S, Casares F. Nuclear receptors connect progenitor transcription factors to cell cycle control. Sci Rep 2017; 7:4845. [PMID: 28687780 PMCID: PMC5501803 DOI: 10.1038/s41598-017-04936-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 05/23/2017] [Indexed: 01/31/2023] Open
Abstract
The specification and growth of organs is controlled simultaneously by networks of transcription factors. While the connection between these transcription factors with fate determinants is increasingly clear, how they establish the link with the cell cycle is far less understood. Here we investigate this link in the developing Drosophila eye, where two transcription factors, the MEIS1 homologue hth and the Zn-finger tsh, synergize to stimulate the proliferation of naïve eye progenitors. Experiments combining transcriptomics, open-chromatin profiling, motif analysis and functional assays indicate that these progenitor transcription factors exert a global regulation of the proliferation program. Rather than directly regulating cell cycle genes, they control proliferation through an intermediary layer of nuclear receptors of the ecdysone/estrogen-signaling pathway. This regulatory subnetwork between hth, tsh and nuclear receptors might be conserved from Drosophila to mammals, as we find a significant co-overexpression of their human homologues in specific cancer types.
Collapse
Affiliation(s)
- Marta Neto
- CABD, Andalusian Centre for Developmental Biology, CSIC-UPO-JA, 41013, Seville, Spain.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | | | - Delphine Potier
- School of Medicine, University of Leuven, box 602 3000, Leuven, Belgium
| | - Paulo S Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Dirk Geerts
- Department of Medical Biology L2-109, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Stein Aerts
- School of Medicine, University of Leuven, box 602 3000, Leuven, Belgium.
| | - Fernando Casares
- CABD, Andalusian Centre for Developmental Biology, CSIC-UPO-JA, 41013, Seville, Spain.
| |
Collapse
|
23
|
Grebbin BM, Hau AC, Groß A, Anders-Maurer M, Schramm J, Koss M, Wille C, Mittelbronn M, Selleri L, Schulte D. Pbx1 is required for adult subventricular zone neurogenesis. Development 2016; 143:2281-91. [PMID: 27226325 DOI: 10.1242/dev.128033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 05/15/2016] [Indexed: 12/22/2022]
Abstract
TALE-homeodomain proteins function as components of heteromeric complexes that contain one member each of the PBC and MEIS/PREP subclasses. We recently showed that MEIS2 cooperates with the neurogenic transcription factor PAX6 in the control of adult subventricular zone (SVZ) neurogenesis in rodents. Expression of the PBC protein PBX1 in the SVZ has been reported, but its functional role(s) has not been investigated. Using a genetic loss-of-function mouse model, we now show that Pbx1 is an early regulator of SVZ neurogenesis. Targeted deletion of Pbx1 by retroviral transduction of Cre recombinase into Pbx2-deficient SVZ stem and progenitor cells carrying floxed alleles of Pbx1 significantly reduced the production of neurons and increased the generation of oligodendrocytes. Loss of Pbx1 expression in neuronally committed neuroblasts in the rostral migratory stream in a Pbx2 null background, by contrast, severely compromised cell survival. By chromatin immunoprecipitation from endogenous tissues or isolated cells, we further detected PBX1 binding to known regulatory regions of the neuron-specific genes Dcx and Th days or even weeks before the respective genes are expressed during the normal program of SVZ neurogenesis, suggesting that PBX1 might act as a priming factor to mark these genes for subsequent activation. Collectively, our results establish that PBX1 regulates adult neural cell fate determination in a manner beyond that of its heterodimerization partner MEIS2.
Collapse
Affiliation(s)
- Britta Moyo Grebbin
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| | - Ann-Christin Hau
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| | - Anja Groß
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| | - Marie Anders-Maurer
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| | - Jasmine Schramm
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| | - Matthew Koss
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Christoph Wille
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| | - Licia Selleri
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA Program in Craniofacial Biology, Institute of Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Departments of Orofacial Sciences and Anatomy, University of California, San Francisco, 513 Parnassus Avenue, HSW 710, San Francisco, CA 94143, USA
| | - Dorothea Schulte
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, German Cancer Consortium (DKTK), Heinrich-Hoffmann Str. 7, Frankfurt D-60528, Germany
| |
Collapse
|
24
|
Transitional Progenitors during Vertebrate Retinogenesis. Mol Neurobiol 2016; 54:3565-3576. [PMID: 27194297 DOI: 10.1007/s12035-016-9899-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 05/03/2016] [Indexed: 12/13/2022]
Abstract
The retina is a delicate neural tissue responsible for light signal capturing, modulating, and passing to mid-brain. The brain then translated the signals into three-dimensional vision. The mature retina is composed of more than 50 subtypes of cells, all of which are developed from a pool of early multipotent retinal progenitors, which pass through sequential statuses of oligopotent, bipotent, and unipotent progenitors, and finally become terminally differentiated retinal cells. A transitional progenitor model is proposed here to describe how intrinsic developmental programs, along with environmental cues, control the step-by-step differentiation during retinogenesis. The model could elegantly explain many current findings as well as predict roles of intrinsic factors during retinal development.
Collapse
|
25
|
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
|
26
|
Beccari L, Marco-Ferreres R, Tabanera N, Manfredi A, Souren M, Wittbrodt B, Conte I, Wittbrodt J, Bovolenta P. A trans-Regulatory Code for the Forebrain Expression of Six3.2 in the Medaka Fish. J Biol Chem 2015; 290:26927-26942. [PMID: 26378230 PMCID: PMC4646366 DOI: 10.1074/jbc.m115.681254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/11/2015] [Indexed: 12/16/2022] Open
Abstract
A well integrated and hierarchically organized gene regulatory network is responsible for the progressive specification of the forebrain. The transcription factor Six3 is one of the central components of this network. As such, Six3 regulates several components of the network, but its upstream regulators are still poorly characterized. Here we have systematically identified such regulators, taking advantage of the detailed functional characterization of the regulatory region of the medaka fish Six3.2 ortholog and of a time/cost-effective trans-regulatory screening, which complemented and overcame the limitations of in silico prediction approaches. The candidates resulting from this search were validated with dose-response luciferase assays and expression pattern criteria. Reconfirmed candidates with a matching expression pattern were also tested with chromatin immunoprecipitation and functional studies. Our results confirm the previously proposed direct regulation of Pax6 and further demonstrate that Msx2 and Pbx1 are bona fide direct regulators of early Six3.2 distribution in distinct domains of the medaka fish forebrain. They also point to other transcription factors, including Tcf3, as additional regulators of different spatial-temporal domains of Six3.2 expression. The activity of these regulators is discussed in the context of the gene regulatory network proposed for the specification of the forebrain.
Collapse
Affiliation(s)
- Leonardo Beccari
- Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolas Cabrera 1, Madrid 28049, Spain,; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolas Cabrera 1, Madrid 28049, Spain; Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda. Dr. Arce 37, Madrid, 28002, Spain,.
| | - Raquel Marco-Ferreres
- Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolas Cabrera 1, Madrid 28049, Spain,; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolas Cabrera 1, Madrid 28049, Spain; Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda. Dr. Arce 37, Madrid, 28002, Spain
| | - Noemi Tabanera
- Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolas Cabrera 1, Madrid 28049, Spain,; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolas Cabrera 1, Madrid 28049, Spain; Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda. Dr. Arce 37, Madrid, 28002, Spain
| | - Anna Manfredi
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda. Dr. Arce 37, Madrid, 28002, Spain
| | - Marcel Souren
- the Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Beate Wittbrodt
- the Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Ivan Conte
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda. Dr. Arce 37, Madrid, 28002, Spain,; the Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, Naples, 80078, Italy
| | - Jochen Wittbrodt
- the Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Paola Bovolenta
- Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolas Cabrera 1, Madrid 28049, Spain,; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolas Cabrera 1, Madrid 28049, Spain; Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda. Dr. Arce 37, Madrid, 28002, Spain,.
| |
Collapse
|
27
|
Machon O, Masek J, Machonova O, Krauss S, Kozmik Z. Meis2 is essential for cranial and cardiac neural crest development. BMC DEVELOPMENTAL BIOLOGY 2015; 15:40. [PMID: 26545946 PMCID: PMC4636814 DOI: 10.1186/s12861-015-0093-6] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/03/2015] [Indexed: 11/28/2022]
Abstract
Background TALE-class homeodomain transcription factors Meis and Pbx play important roles in formation of the embryonic brain, eye, heart, cartilage or hematopoiesis. Loss-of-function studies of Pbx1, 2 and 3 and Meis1 documented specific functions in embryogenesis, however, functional studies of Meis2 in mouse are still missing. We have generated a conditional allele of Meis2 in mice and shown that systemic inactivation of the Meis2 gene results in lethality by the embryonic day 14 that is accompanied with hemorrhaging. Results We show that neural crest cells express Meis2 and Meis2-defficient embryos display defects in tissues that are derived from the neural crest, such as an abnormal heart outflow tract with the persistent truncus arteriosus and abnormal cranial nerves. The importance of Meis2 for neural crest cells is further confirmed by means of conditional inactivation of Meis2 using crest-specific AP2α-IRES-Cre mouse. Conditional mutants display perturbed development of the craniofacial skeleton with severe anomalies in cranial bones and cartilages, heart and cranial nerve abnormalities. Conclusions Meis2-null mice are embryonic lethal. Our results reveal a critical role of Meis2 during cranial and cardiac neural crest cells development in mouse. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0093-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ondrej Machon
- Institute of Molecular Genetics, The Czech Academy of Sciences, 14200, Praha, Czech Republic.
| | - Jan Masek
- Institute of Molecular Genetics, The Czech Academy of Sciences, 14200, Praha, Czech Republic.
| | - Olga Machonova
- Institute of Molecular Genetics, The Czech Academy of Sciences, 14200, Praha, Czech Republic.
| | - Stefan Krauss
- Unit for Cell Signaling, Oslo University Hospital, N-0349, Oslo, Norway.
| | - Zbynek Kozmik
- Institute of Molecular Genetics, The Czech Academy of Sciences, 14200, Praha, Czech Republic.
| |
Collapse
|
28
|
miR-134 Modulates the Proliferation of Human Cardiomyocyte Progenitor Cells by Targeting Meis2. Int J Mol Sci 2015; 16:25199-213. [PMID: 26512644 PMCID: PMC4632798 DOI: 10.3390/ijms161025199] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/16/2015] [Accepted: 09/25/2015] [Indexed: 12/18/2022] Open
Abstract
Cardiomyocyte progenitor cells play essential roles in early heart development, which requires highly controlled cellular organization. microRNAs (miRs) are involved in various cell behaviors by post-transcriptional regulation of target genes. However, the roles of miRNAs in human cardiomyocyte progenitor cells (hCMPCs) remain to be elucidated. Our previous study showed that miR-134 was significantly downregulated in heart tissue suffering from congenital heart disease, underlying the potential role of miR-134 in cardiogenesis. In the present work, we showed that the upregulation of miR-134 reduced the proliferation of hCMPCs, as determined by EdU assay and Ki-67 immunostaining, while the inhibition of miR-134 exhibited an opposite effect. Both up- and downregulation of miR-134 expression altered the transcriptional level of cell-cycle genes. We identified Meis2 as the target of miR-134 in the regulation of hCMPC proliferation through bioinformatic prediction, luciferase reporter assay and western blot. The over-expression of Meis2 mitigated the effect of miR-134 on hCMPC proliferation. Moreover, miR-134 did not change the degree of hCMPC differentiation into cardiomyocytes in our model, suggesting that miR-134 is not required in this process. These findings reveal an essential role for miR-134 in cardiomyocyte progenitor cell biology and provide new insights into the physiology and pathology of cardiogenesis.
Collapse
|
29
|
Uribe RA, Bronner ME. Meis3 is required for neural crest invasion of the gut during zebrafish enteric nervous system development. Mol Biol Cell 2015; 26:3728-40. [PMID: 26354419 PMCID: PMC4626059 DOI: 10.1091/mbc.e15-02-0112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 09/02/2015] [Indexed: 01/02/2023] Open
Abstract
Loss of Meis3 leads to defects in enteric neural crest cell migration, number, and proliferation during colonization of the gut. This leads to colonic aganglionosis, in which the hindgut is devoid of neurons, identifying it as a novel candidate factor in the etiology of Hirschsprung’s disease during enteric nervous system development. During development, vagal neural crest cells fated to contribute to the enteric nervous system migrate ventrally away from the neural tube toward and along the primitive gut. The molecular mechanisms that regulate their early migration en route to and entry into the gut remain elusive. Here we show that the transcription factor meis3 is expressed along vagal neural crest pathways. Meis3 loss of function results in a reduction in migration efficiency, cell number, and the mitotic activity of neural crest cells in the vicinity of the gut but has no effect on neural crest or gut specification. Later, during enteric nervous system differentiation, Meis3-depleted embryos exhibit colonic aganglionosis, a disorder in which the hindgut is devoid of neurons. Accordingly, the expression of Shh pathway components, previously shown to have a role in the etiology of Hirschsprung’s disease, was misregulated within the gut after loss of Meis3. Taken together, these findings support a model in which Meis3 is required for neural crest proliferation, migration into, and colonization of the gut such that its loss leads to severe defects in enteric nervous system development.
Collapse
Affiliation(s)
- Rosa A Uribe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
30
|
Mason MK, Hockman D, Curry L, Cunningham TJ, Duester G, Logan M, Jacobs DS, Illing N. Retinoic acid-independent expression of Meis2 during autopod patterning in the developing bat and mouse limb. EvoDevo 2015; 6:6. [PMID: 25861444 PMCID: PMC4389300 DOI: 10.1186/s13227-015-0001-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/04/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The bat has strikingly divergent forelimbs (long digits supporting wing membranes) and hindlimbs (short, typically free digits) due to the distinct requirements of both aerial and terrestrial locomotion. During embryonic development, the morphology of the bat forelimb deviates dramatically from the mouse and chick, offering an alternative paradigm for identifying genes that play an important role in limb patterning. RESULTS Using transcriptome analysis of developing Natal long-fingered bat (Miniopterus natalensis) fore- and hindlimbs, we demonstrate that the transcription factor Meis2 has a significantly higher expression in bat forelimb autopods compared to hindlimbs. Validation by reverse transcriptase and quantitative polymerase chain reaction (RT-qPCR) and whole mount in situ hybridisation shows that Meis2, conventionally known as a marker of the early proximal limb bud, is upregulated in the bat forelimb autopod from CS16. Meis2 expression is localised to the expanding interdigital webbing and the membranes linking the wing to the hindlimb and tail. In mice, Meis2 is also expressed in the interdigital region prior to tissue regression. This interdigital Meis2 expression is not activated by retinoic acid (RA) signalling as it is present in the retained interdigital tissue of Rdh10 (trex/trex) mice, which lack RA. Additionally, genes encoding RA-synthesising enzymes, Rdh10 and Aldh1a2, and the RA nuclear receptor Rarβ are robustly expressed in bat fore- and hindlimb interdigital tissues indicating that the mechanism that retains interdigital tissue in bats also occurs independently of RA signalling. CONCLUSIONS Mammalian interdigital Meis2 expression, and upregulation in the interdigital webbing of bat wings, suggests an important role for Meis2 in autopod development. Interdigital Meis2 expression is RA-independent, and retention of interdigital webbing in bat wings is not due to the suppression of RA-induced cell death. Rather, RA signalling may play a role in the thinning (rather than complete loss) of the interdigital tissue in the bat forelimb, while Meis2 may interact with other factors during both bat and mouse autopod development to maintain a pool of interdigital cells that contribute to digit patterning and growth.
Collapse
Affiliation(s)
- Mandy K Mason
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, 7701 South Africa
| | - Dorit Hockman
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, 7701 South Africa ; Present address: Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS UK
| | - Lyle Curry
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, 7701 South Africa
| | - Thomas J Cunningham
- Sanford-Burnham Medical Research Institute, Development, Aging, and Regeneration Program, La Jolla, 92037 California USA
| | - Gregg Duester
- Sanford-Burnham Medical Research Institute, Development, Aging, and Regeneration Program, La Jolla, 92037 California USA
| | - Malcolm Logan
- Randall Division, King's College London, London, SE1 1UL UK
| | - David S Jacobs
- Department of Biological Sciences, University of Cape Town, Rondebosch, 7701 South Africa
| | - Nicola Illing
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, 7701 South Africa
| |
Collapse
|
31
|
Manuel MN, Mi D, Mason JO, Price DJ. Regulation of cerebral cortical neurogenesis by the Pax6 transcription factor. Front Cell Neurosci 2015; 9:70. [PMID: 25805971 PMCID: PMC4354436 DOI: 10.3389/fncel.2015.00070] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/18/2015] [Indexed: 12/19/2022] Open
Abstract
Understanding brain development remains a major challenge at the heart of understanding what makes us human. The neocortex, in evolutionary terms the newest part of the cerebral cortex, is the seat of higher cognitive functions. Its normal development requires the production, positioning, and appropriate interconnection of very large numbers of both excitatory and inhibitory neurons. Pax6 is one of a relatively small group of transcription factors that exert high-level control of cortical development, and whose mutation or deletion from developing embryos causes major brain defects and a wide range of neurodevelopmental disorders. Pax6 is very highly conserved between primate and non-primate species, is expressed in a gradient throughout the developing cortex and is essential for normal corticogenesis. Our understanding of Pax6’s functions and the cellular processes that it regulates during mammalian cortical development has significantly advanced in the last decade, owing to the combined application of genetic and biochemical analyses. Here, we review the functional importance of Pax6 in regulating cortical progenitor proliferation, neurogenesis, and formation of cortical layers and highlight important differences between rodents and primates. We also review the pathological effects of PAX6 mutations in human neurodevelopmental disorders. We discuss some aspects of Pax6’s molecular actions including its own complex transcriptional regulation, the distinct molecular functions of its splice variants and some of Pax6’s known direct targets which mediate its actions during cortical development.
Collapse
Affiliation(s)
- Martine N Manuel
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| | - Da Mi
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| | - John O Mason
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| | - David J Price
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| |
Collapse
|
32
|
Marcos S, González-Lázaro M, Beccari L, Carramolino L, Martin-Bermejo MJ, Amarie O, Martín DMS, Torroja C, Bogdanović O, Doohan R, Puk O, de Angelis MH, Graw J, Gomez-Skarmeta JL, Casares F, Torres M, Bovolenta P. Meis1 coordinates a network of genes implicated in eye development and microphthalmia. Development 2015; 142:3009-20. [DOI: 10.1242/dev.122176] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 07/17/2015] [Indexed: 01/08/2023]
Abstract
Microphthalmos is a rare congenital anomaly characterized by reduced eye size and visual deficits of variable degrees. Sporadic and hereditary microphthalmos has been associated to heterozygous mutations in genes fundamental for eye development. Yet, many cases are idiopathic or await the identification of molecular causes. Here we show that haploinsufficiency of Meis1, a transcription factor with an evolutionary conserved expression in the embryonic trunk, brain and sensory organs, including the eye, causes microphthalmic traits and visual impairment, in adult mice. By combining the analysis of Meis1 loss-of-function and conditional Meis1 functional rescue with ChIP-seq and RNA-seq approaches we show that, in contrast to Meis1 preferential association with Hox-Pbx binding sites in the trunk, Meis1 binds to Hox/Pbx-independent sites during optic cup development. In the eye primordium, Meis1 coordinates, in a dose-dependent manner, retinal proliferation and differentiation by regulating genes responsible for human microphthalmia and components the Notch signalling pathway. In addition, Meis1 is required for eye patterning by controlling a set of eye territory-specific transcription factors, so that in Meis1−/− embryos boundaries among the different eye territories are shifted or blurred. We thus propose that Meis1 is at the core of a genetic network implicated in eye patterning/microphthalmia, itself representing an additional candidate for syndromic cases of these ocular malformations.
Collapse
Affiliation(s)
- Séverine Marcos
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| | - Monica González-Lázaro
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Leonardo Beccari
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| | - Laura Carramolino
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Maria Jesus Martin-Bermejo
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| | - Oana Amarie
- Institute of Developmental Genetics Helmholtz Center Munich; D-85764 Neuherberg, Germany
| | - Daniel Mateos-San Martín
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Carlos Torroja
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Ozren Bogdanović
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-UPO, Carretera de Utrera Km1, E-41013 Sevilla, Spain
- ARC Center of Excellence in Plant Energy Biology, School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, WA 6009, Australia
| | - Roisin Doohan
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Oliver Puk
- Institute of Developmental Genetics Helmholtz Center Munich; D-85764 Neuherberg, Germany
| | | | - Jochen Graw
- Institute of Developmental Genetics Helmholtz Center Munich; D-85764 Neuherberg, Germany
| | - Jose Luis Gomez-Skarmeta
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-UPO, Carretera de Utrera Km1, E-41013 Sevilla, Spain
| | - Fernando Casares
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-UPO, Carretera de Utrera Km1, E-41013 Sevilla, Spain
| | - Miguel Torres
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| |
Collapse
|
33
|
Schulte D. Meis: New friends of Pax. NEUROGENESIS 2014; 1:e976014. [PMID: 27502016 PMCID: PMC4973581 DOI: 10.4161/23262133.2014.976014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/04/2014] [Accepted: 10/08/2014] [Indexed: 12/27/2022]
Abstract
The generation of neuronal diversity in the mammalian brain is a multistep process, beginning with the regional patterning of neural stem- and progenitor cell domains, the commitment of these cells toward a general neuronal fate, followed by the selection of a particular neuronal subtype and the differentiation of postmitotic neurons. Each of these steps as well as the transitions between them require precisely controlled changes in transcriptional programs. Although a large number of transcription factors are known to regulate neurogenesis in the embryonic and adult central nervous system, the sheer number of neuronal cell types in the brain and the complexity of the cellular processes that accompany their production suggest that transcription factors act cooperatively to control individual steps in neurogenesis. In fact, combinatorial regulation by sets of transcription factors has emerged as a versatile mode to control cell fate specification. Here, I discuss our recent finding that members of the MEIS-subfamily of TALE-transcription factors, originally identified as HOX cofactors in non-neural tissues, function in concert with PAX-proteins in the regulation of cell fate specification and neuronal differentiation in the embryonic and adult brain.
Collapse
Affiliation(s)
- Dorothea Schulte
- Institute of Neurology (Edinger Institute); University Hospital Goethe University ; Frankfurt, Germany
| |
Collapse
|
34
|
MEIS2 is essential for neuroblastoma cell survival and proliferation by transcriptional control of M-phase progression. Cell Death Dis 2014; 5:e1417. [PMID: 25210800 PMCID: PMC4540202 DOI: 10.1038/cddis.2014.370] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/14/2014] [Accepted: 07/22/2014] [Indexed: 01/01/2023]
Abstract
MEIS2 has an important role in development and organogenesis, and is implicated in the pathogenesis of human cancer. The molecular basis of MEIS2 action in tumorigenesis is not clear. Here, we show that MEIS2 is highly expressed in human neuroblastoma cell lines and is required for neuroblastoma cell survival and proliferation. Depletion of MEIS2 in neuroblastoma cells leads to M-phase arrest and mitotic catastrophe, whereas ectopic expression of MEIS2 markedly enhances neuroblastoma cell proliferation, anchorage-independent growth, and tumorigenicity. Gene expression profiling reveals an essential role of MEIS2 in maintaining the expression of a large number of late cell-cycle genes, including those required for DNA replication, G2-M checkpoint control and M-phase progression. Importantly, we identify MEIS2 as a transcription activator of the MuvB-BMYB-FOXM1 complex that functions as a master regulator of cell-cycle gene expression. Further, we show that FOXM1 is a direct target gene of MEIS2 and is required for MEIS2 to upregulate mitotic genes. These findings link a developmentally important gene to the control of cell proliferation and suggest that high MEIS2 expression is a molecular mechanism for high expression of mitotic genes that is frequently observed in cancers of poor prognosis.
Collapse
|
35
|
Dai M, Wang Y, Fang L, Irwin DM, Zhu T, Zhang J, Zhang S, Wang Z. Differential expression of Meis2, Mab21l2 and Tbx3 during limb development associated with diversification of limb morphology in mammals. PLoS One 2014; 9:e106100. [PMID: 25166052 PMCID: PMC4148388 DOI: 10.1371/journal.pone.0106100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 07/27/2014] [Indexed: 11/19/2022] Open
Abstract
Bats are the only mammals capable of self-powered flight using wings. Differing from mouse or human limbs, four elongated digits within a broad wing membrane support the bat wing, and the foot of the bat has evolved a long calcar that spread the interfemoral membrane. Our recent mRNA sequencing (mRNA-Seq) study found unique expression patterns for genes at the 5' end of the Hoxd gene cluster and for Tbx3 that are associated with digit elongation and wing membrane growth in bats. In this study, we focused on two additional genes, Meis2 and Mab21l2, identified from the mRNA-Seq data. Using whole-mount in situ hybridization (WISH) we validated the mRNA-Seq results for differences in the expression patterns of Meis2 and Mab21l2 between bat and mouse limbs, and further characterize the timing and location of the expression of these two genes. These analyses suggest that Meis2 may function in wing membrane growth and Mab21l2 may have a role in AP and DV axial patterning. In addition, we found that Tbx3 is uniquely expressed in the unique calcar structure found in the bat hindlimb, suggesting a role for this gene in calcar growth and elongation. Moreover, analysis of the coding sequences for Meis2, Mab21l2 and Tbx3 showed that Meis2 and Mab21l2 have high sequence identity, consistent with the functions of genes being conserved, but that Tbx3 showed accelerated evolution in bats. However, evidence for positive selection in Tbx3 was not found, which would suggest that the function of this gene has not been changed. Together, our findings support the hypothesis that the modulation of the spatiotemporal expression patterns of multiple functional conserved genes control limb morphology and drive morphological change in the diversification of mammalian limbs.
Collapse
Affiliation(s)
- Mengyao Dai
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Yao Wang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Lu Fang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Tengteng Zhu
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Junpeng Zhang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Shuyi Zhang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Zhe Wang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| |
Collapse
|
36
|
Bokolia NP, Mishra M. Hearing molecules, mechanism and transportation: modeled in Drosophila melanogaster. Dev Neurobiol 2014; 75:109-30. [PMID: 25081222 DOI: 10.1002/dneu.22221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/29/2014] [Accepted: 07/29/2014] [Indexed: 01/19/2023]
Abstract
Mechanosensory transduction underlies the perception of touch, sound and acceleration. The mechanical signals exist in the environment are resensed by the specialized mechanosensory cells, which convert the external forces into the electrical signals. Hearing is a magnificent example that relies on the mechanotransduction mediated by the auditory cells, for example the inner-ear hair cells in vertebrates and the Johnston's organ (JO) in fly. Previous studies have shown the fundamental physiological processes in the fly and vertebrate auditory organs are similar, suggesting that there might be a set of similar molecules underlying these processes. The molecular studies of the fly JO have been shown to be remarkably successful in discovering the developmental and functional genes that provided further implications in vertebrates. Several evolutionarily conserved molecules and signaling pathways have been shown to govern the development of the auditory organs in both vertebrates and invertebrates. The current review describes the similarities and differences between the vertebrate and fly auditory organs at developmental, structural, molecular, and transportation levels.
Collapse
Affiliation(s)
- Naveen Prakash Bokolia
- Department of Life Science, National Institute of Technology Rourkela, Rourkela, Orissa, India
| | | |
Collapse
|
37
|
Zagozewski JL, Zhang Q, Pinto VI, Wigle JT, Eisenstat DD. The role of homeobox genes in retinal development and disease. Dev Biol 2014; 393:195-208. [PMID: 25035933 DOI: 10.1016/j.ydbio.2014.07.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 07/02/2014] [Accepted: 07/08/2014] [Indexed: 11/18/2022]
Abstract
Homeobox genes are an evolutionarily conserved class of transcription factors that are critical for development of many organ systems, including the brain and eye. During retinogenesis, homeodomain-containing transcription factors, which are encoded by homeobox genes, play essential roles in the regionalization and patterning of the optic neuroepithelium, specification of retinal progenitors and differentiation of all seven of the retinal cell classes that derive from a common progenitor. Homeodomain transcription factors control retinal cell fate by regulating the expression of target genes required for retinal progenitor cell fate decisions and for terminal differentiation of specific retinal cell types. The essential role of homeobox genes during retinal development is demonstrated by the number of human eye diseases, including colobomas and anophthalmia, which are attributed to homeobox gene mutations. In the following review, we highlight the role of homeodomain transcription factors during retinogenesis and regulation of their gene targets. Understanding the complexities of vertebrate retina development will enhance our ability to drive differentiation of specific retinal cell types towards novel cell-based replacement therapies for retinal degenerative diseases.
Collapse
Affiliation(s)
- Jamie L Zagozewski
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada T6G 2H7
| | - Qi Zhang
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada R3E 0J9
| | - Vanessa I Pinto
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada R3E 0J9
| | - Jeffrey T Wigle
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada R3E 0J9; Institute of Cardiovascular Sciences, St. Boniface Hospital Research Institute, Winnipeg, MB, Canada R2H 2A6
| | - David D Eisenstat
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada T6G 2H7; Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada R3E 0J9; Department of Pediatrics, University of Alberta, Edmonton, AB, Canada T6G 1C9.
| |
Collapse
|
38
|
Okumura K, Saito M, Isogai E, Aoto Y, Hachiya T, Sakakibara Y, Katsuragi Y, Hirose S, Kominami R, Goitsuka R, Nakamura T, Wakabayashi Y. Meis1 regulates epidermal stem cells and is required for skin tumorigenesis. PLoS One 2014; 9:e102111. [PMID: 25013928 PMCID: PMC4094504 DOI: 10.1371/journal.pone.0102111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/14/2014] [Indexed: 12/17/2022] Open
Abstract
Previous studies have shown that Meis1 plays an important role in blood development and vascular homeostasis, and can induce blood cancers, such as leukemia. However, its role in epithelia remains largely unknown. Here, we uncover two roles for Meis1 in the epidermis: as a critical regulator of epidermal homeostasis in normal tissues and as a proto-oncogenic factor in neoplastic tissues. In normal epidermis, we show that Meis1 is predominantly expressed in the bulge region of the hair follicles where multipotent adult stem cells reside, and that the number of these stem cells is reduced when Meis1 is deleted in the epidermal tissue of mice. Mice with epidermal deletion of Meis1 developed significantly fewer DMBA/TPA-induced benign and malignant tumors compared with wild-type mice, suggesting that Meis1 plays a role in both tumor development and malignant progression. This is consistent with the observation that Meis1 expression increases as tumors progress from benign papillomas to malignant carcinomas. Interestingly, we found that Meis1 localization was altered to neoplasia development. Instead of being localized to the stem cell region, Meis1 is localized to more differentiated cells in tumor tissues. These findings suggest that, during the transformation from normal to neoplastic tissues, a functional switch occurs in Meis1.
Collapse
Affiliation(s)
- Kazuhiro Okumura
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Chiba, Japan
| | - Megumi Saito
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Chiba, Japan
| | - Eriko Isogai
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Chiba, Japan
| | - Yoshimasa Aoto
- Department of Biosciences and Informatics, Bioinfomatics Laboratory, Keio University, Yokohama, Kanagawa, Japan
| | - Tsuyoshi Hachiya
- Department of Biosciences and Informatics, Bioinfomatics Laboratory, Keio University, Yokohama, Kanagawa, Japan
| | - Yasubumi Sakakibara
- Department of Biosciences and Informatics, Bioinfomatics Laboratory, Keio University, Yokohama, Kanagawa, Japan
| | - Yoshinori Katsuragi
- Department of Molecular Genetics, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Satoshi Hirose
- Department of Molecular Genetics, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Ryo Kominami
- Department of Molecular Genetics, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Ryo Goitsuka
- Division of Development and Aging, Research Institute for Biological Science, Tokyo University of Science, Noda, Chiba, Japan
| | - Takuro Nakamura
- Division of Carcinogenesis, Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo, Japan
| | - Yuichi Wakabayashi
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Chiba, Japan
- * E-mail:
| |
Collapse
|
39
|
Liu H, Murthi P, Qin S, Kusuma GD, Borg AJ, Knöfler M, Haslinger P, Manuelpillai U, Pertile MD, Abumaree M, Kalionis B. A novel combination of homeobox genes is expressed in mesenchymal chorionic stem/stromal cells in first trimester and term pregnancies. Reprod Sci 2014; 21:1382-94. [PMID: 24692208 DOI: 10.1177/1933719114526471] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human chorionic mesenchymal stem/stromal cells (CMSCs) derived from the placenta are similar to adult tissue-derived MSCs. The aim of this study was to investigate the role of these cells in normal placental development. Transcription factors, particularly members of the homeobox gene family, play crucial roles in maintaining stem cell proliferation and lineage specification in embryonic tissues. In adult tissues and organs, stem cells proliferate at low levels in their niche until they receive cues from the microenvironment to differentiate. The homeobox genes that are expressed in the CMSC niche in placental tissues have not been identified. We used the novel strategy of laser capture microdissection to isolate the stromal component of first trimester villi and excluded the cytotrophoblast and syncytiotrophoblast layers that comprise the outer layer of the chorionic villi. Microarray analysis was then used to screen for homeobox genes in the microdissected tissue. Candidate homeobox genes were selected for further RNA analysis. Immunohistochemistry of candidate genes in first trimester placental villous stromal tissue revealed homeobox genes Meis1, myeloid ectropic viral integration site 1 homolog 2 (MEIS2), H2.0-like Drosophila (HLX), transforming growth factor β-induced factor (TGIF), and distal-less homeobox 5 (DLX5) were expressed in the vascular niche where CMSCs have been shown to reside. Expression of MEIS2, HLX, TGIF, and DLX5 was also detected in scattered stromal cells. Real-time polymerase chain reaction and immunocytochemistry verified expression of MEIS2, HLX, TGIF, and DLX5 homeobox genes in first trimester and term CMSCs. These data suggest a combination of regulatory homeobox genes is expressed in CMSCs from early placental development to term, which may be required for stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- Haiying Liu
- Department of Obstetrics and Gynaecology, QiLu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Padma Murthi
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Sharon Qin
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Gina D Kusuma
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Anthony J Borg
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Martin Knöfler
- Department of Obstetrics and Fetal-Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Haslinger
- Department of Obstetrics and Fetal-Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Ursula Manuelpillai
- Centre for Genetic Diseases, Monash Institute of Medical Research, Monash University, Clayton, Victoria
| | - Mark D Pertile
- VCGS, Murdoch Children's Research Institute, Royal Childrens Hospital, Flemington Road, Parkville, Victoria, Australia
| | - Mohamed Abumaree
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences/ King Abdulla International Medical Research Center, Riyadh, Saudi Arabia
| | - Bill Kalionis
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
40
|
Johansson S, Berland S, Gradek GA, Bongers E, de Leeuw N, Pfundt R, Fannemel M, Rødningen O, Brendehaug A, Haukanes BI, Hovland R, Helland G, Houge G. Haploinsufficiency ofMEIS2is associated with orofacial clefting and learning disability. Am J Med Genet A 2014; 164A:1622-6. [DOI: 10.1002/ajmg.a.36498] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 11/14/2013] [Indexed: 11/12/2022]
Affiliation(s)
- Stefan Johansson
- Center for Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
- Department of Clinical Science; University of Bergen; Bergen Norway
| | - Siren Berland
- Center for Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
| | - Gyri Aasland Gradek
- Center for Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
| | - Ernie Bongers
- Department of Human Genetics; Radboud University Medical Centre; Nijmegen The Netherlands
| | - Nicole de Leeuw
- Department of Human Genetics; Radboud University Medical Centre; Nijmegen The Netherlands
| | - Rolph Pfundt
- Department of Human Genetics; Radboud University Medical Centre; Nijmegen The Netherlands
| | - Madeleine Fannemel
- Department of Medical Genetics; Oslo University Hospital; Ullevål Oslo Norway
| | - Olaug Rødningen
- Department of Medical Genetics; Oslo University Hospital; Ullevål Oslo Norway
| | - Atle Brendehaug
- Center for Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
| | - Bjørn Ivar Haukanes
- Center for Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
| | - Randi Hovland
- Center for Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
| | - Gunnar Helland
- Department of Pediatrics; Levanger Hospital; Levanger Norway
| | - Gunnar Houge
- Center for Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
| |
Collapse
|
41
|
Hirayama T, Asano Y, Iida H, Watanabe T, Nakamura T, Goitsuka R. Meis1 is required for the maintenance of postnatal thymic epithelial cells. PLoS One 2014; 9:e89885. [PMID: 24594519 PMCID: PMC3942356 DOI: 10.1371/journal.pone.0089885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 01/24/2014] [Indexed: 12/19/2022] Open
Abstract
Most epithelial tissues retain stem/progenitor cells to maintain homeostasis of the adult tissues; however, the existence of a thymic epithelial cell (TEC) progenitor capable of maintaining homeostasis of the postnatal thymus remains unclear. Here, we show that a cell population expressing high levels of Meis1, a homeodomain transcription factor, is enriched in TECs with an immature cellular phenotype. These TECs selectively express genes involved in embryonic thymic organogenesis and epithelial stem cell maintenance, and also have the potential to proliferate and differentiate into mature TEC populations. Furthermore, postnatal inactivation of Meis1 in TECs caused disorganization of the thymic architecture, which ultimately leads to premature disappearance of the thymus. There was an age-associated reduction in the proportion of the TEC population expressing high levels of Meis1, which may also be related to thymic involution. These findings indicate that Meis1 is potentially involved in the maintenance of postnatal TECs with progenitor activity that is required for homeostasis of the postnatal thymus.
Collapse
Affiliation(s)
- Takehiro Hirayama
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yusuke Asano
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Hajime Iida
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Takeshi Watanabe
- Center for Innovation in Immunoregulative Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takuro Nakamura
- Department of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryo Goitsuka
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
- * E-mail:
| |
Collapse
|
42
|
Ariki R, Morikawa S, Mabuchi Y, Suzuki S, Nakatake M, Yoshioka K, Hidano S, Nakauchi H, Matsuzaki Y, Nakamura T, Goitsuka R. Homeodomain transcription factor Meis1 is a critical regulator of adult bone marrow hematopoiesis. PLoS One 2014; 9:e87646. [PMID: 24498346 PMCID: PMC3911998 DOI: 10.1371/journal.pone.0087646] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 12/26/2013] [Indexed: 12/02/2022] Open
Abstract
Hematopoietic stem cells in the bone marrow have the capacity to both self-renew and to generate all cells of the hematopoietic system. The balance of these two activities is controlled by hematopoietic stem cell-intrinsic regulatory mechanisms as well as extrinsic signals from the microenvironment. Here we demonstrate that Meis1, a TALE family homeodomain transcription factor involved in numerous embryonic developmental processes, is selectively expressed in hematopoietic stem/progenitor cells. Conditional Meis1 knockout in adult hematopoietic cells resulted in a significant reduction in the hematopoietic stem/progenitor cells. Suppression of hematopoiesis by Meis1 deletion appears to be caused by impaired self-renewal activity and reduced cellular quiescence of hematopoietic stem/progenitor cells in a cell autonomous manner, resulting in stem cell exhaustion and defective long-term hematopoiesis. Meis1 deficiency down-regulated a subset of Pbx1-dependent hematopoietic stem cell signature genes, suggesting a functional link between them in the maintenance of hematopoietic stem/progenitor cells. These results show the importance of Meis1 in adult hematopoiesis.
Collapse
Affiliation(s)
- Reina Ariki
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Satoru Morikawa
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yo Mabuchi
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Sadafumi Suzuki
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Mayuka Nakatake
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Kentaro Yoshioka
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Shinya Hidano
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Hiromitsu Nakauchi
- Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Yumi Matsuzaki
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Takuro Nakamura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
- * E-mail: (TN); (RG)
| | - Ryo Goitsuka
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- * E-mail: (TN); (RG)
| |
Collapse
|
43
|
Cerdá-Esteban N, Spagnoli FM. Glimpse into Hox and tale regulation of cell differentiation and reprogramming. Dev Dyn 2013; 243:76-87. [PMID: 24123411 DOI: 10.1002/dvdy.24075] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 09/15/2013] [Accepted: 10/04/2013] [Indexed: 12/20/2022] Open
Abstract
During embryonic development, cells become gradually restricted in their developmental potential and start elaborating lineage-specific transcriptional networks to ultimately acquire a unique differentiated state. Hox genes play a central role in specifying regional identities, thereby providing the cell with critical information on positional value along its differentiation path. The exquisite DNA-binding specificity of the Hox proteins is frequently dependent upon their interaction with members of the TALE family of homeodomain proteins. In addition to their function as Hox-cofactors, TALE homeoproteins control multiple crucial developmental processes through Hox-independent mechanisms. Here, we will review recent findings on the function of both Hox and TALE proteins in cell differentiation, referring mostly to vertebrate species. In addition, we will discuss the direct implications of this knowledge on cell plasticity and cell reprogramming.
Collapse
Affiliation(s)
- Nuria Cerdá-Esteban
- Laboratory of Molecular and Cellular Basis of Embryonic Development, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | |
Collapse
|
44
|
Agoston Z, Heine P, Brill MS, Grebbin BM, Hau AC, Kallenborn-Gerhardt W, Schramm J, Götz M, Schulte D. Meis2 is a Pax6 co-factor in neurogenesis and dopaminergic periglomerular fate specification in the adult olfactory bulb. Development 2013; 141:28-38. [PMID: 24284204 DOI: 10.1242/dev.097295] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Meis homeodomain transcription factors control cell proliferation, cell fate specification and differentiation in development and disease. Previous studies have largely focused on Meis contribution to the development of non-neuronal tissues. By contrast, Meis function in the brain is not well understood. Here, we provide evidence for a dual role of the Meis family protein Meis2 in adult olfactory bulb (OB) neurogenesis. Meis2 is strongly expressed in neuroblasts of the subventricular zone (SVZ) and rostral migratory stream (RMS) and in some of the OB interneurons that are continuously replaced during adult life. Targeted manipulations with retroviral vectors expressing function-blocking forms or with small interfering RNAs demonstrated that Meis activity is cell-autonomously required for the acquisition of a general neuronal fate by SVZ-derived progenitors in vivo and in vitro. Additionally, Meis2 activity in the RMS is important for the generation of dopaminergic periglomerular neurons in the OB. Chromatin immunoprecipitation identified doublecortin and tyrosine hydroxylase as direct Meis targets in newly generated neurons and the OB, respectively. Furthermore, biochemical analyses revealed a previously unrecognized complex of Meis2 with Pax6 and Dlx2, two transcription factors involved in OB neurogenesis. The full pro-neurogenic activity of Pax6 in SVZ derived neural stem and progenitor cells requires the presence of Meis. Collectively, these results show that Meis2 cooperates with Pax6 in generic neurogenesis and dopaminergic fate specification in the adult SVZ-OB system.
Collapse
Affiliation(s)
- Zsuzsa Agoston
- Institute of Neurology (Edinger Institute), J. W. Goethe University Medical School, D-60528 Frankfurt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Schulte D, Frank D. TALE transcription factors during early development of the vertebrate brain and eye. Dev Dyn 2013; 243:99-116. [DOI: 10.1002/dvdy.24030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 07/11/2013] [Accepted: 07/13/2013] [Indexed: 12/25/2022] Open
Affiliation(s)
- Dorothea Schulte
- Institute of Neurology (Edinger Institute); University Hospital Frankfurt, J.W. Goethe University; Frankfurt Germany
| | - Dale Frank
- Department of Biochemistry; The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology; Haifa Israel
| |
Collapse
|
46
|
Islam MM, Li Y, Luo H, Xiang M, Cai L. Meis1 regulates Foxn4 expression during retinal progenitor cell differentiation. Biol Open 2013; 2:1125-36. [PMID: 24244849 PMCID: PMC3828759 DOI: 10.1242/bio.20132279] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/12/2013] [Indexed: 12/20/2022] Open
Abstract
The transcription factor forkhead box N4 (Foxn4) is a key regulator in a variety of biological processes during development. In particular, Foxn4 plays an essential role in the genesis of horizontal and amacrine neurons from neural progenitors in the vertebrate retina. Although the functions of Foxn4 have been well established, the transcriptional regulation of Foxn4 expression during progenitor cell differentiation remains unclear. Here, we report that an evolutionarily conserved 129 bp noncoding DNA fragment (Foxn4CR4.2 or CR4.2), located ∼26 kb upstream of Foxn4 transcription start site, functions as a cis-element for Foxn4 regulation. CR4.2 directs gene expression in Foxn4-positive cells, primarily in progenitors, differentiating horizontal and amacrine cells. We further determined that the gene regulatory activity of CR4.2 is modulated by Meis1 binding motif, which is bound and activated by Meis1 transcription factor. Deletion of the Meis1 binding motif or knockdown of Meis1 expression abolishes the gene regulatory activity of CR4.2. In addition, knockdown of Meis1 expression diminishes the endogenous Foxn4 expression and affects cell lineage development. Together, we demonstrate that CR4.2 and its interacting Meis1 transcription factor play important roles in regulating Foxn4 expression during early retinogenesis. These findings provide new insights into molecular mechanisms that govern gene regulation in retinal progenitors and specific cell lineage development.
Collapse
Affiliation(s)
- Mohammed M Islam
- Department of Biomedical Engineering, Rutgers University , 599 Taylor Road, Piscataway, NJ 08854 , USA ; Present address: Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
47
|
Lysenko O, Schulte D, Mittelbronn M, Steinle A. BACL is a novel brain-associated, non-NKC-encoded mammalian C-type lectin-like receptor of the CLEC2 family. PLoS One 2013; 8:e65345. [PMID: 23776472 PMCID: PMC3679072 DOI: 10.1371/journal.pone.0065345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 04/20/2013] [Indexed: 11/24/2022] Open
Abstract
Natural Killer Gene Complex (NKC)–encoded C-type lectin-like receptors (CTLRs) are expressed on various immune cells including T cells, NK cells and myeloid cells and thereby contribute to the orchestration of cellular immune responses. Some NKC-encoded CTLRs are grouped into the C-type lectin family 2 (CLEC2 family) and interact with genetically linked CTLRs of the NKRP1 family. While many CLEC2 family members are expressed by hematopoietic cells (e.g. CD69 (CLEC2C)), others such as the keratinocyte-associated KACL (CLEC2A) are specifically expressed by other tissues. Here we provide the first characterization of the orphan gene CLEC2L. In contrast to other CLEC2 family members, CLEC2L is conserved among mammals and located outside of the NKC. We show that CLEC2L-encoded CTLRs are expressed as non-glycosylated, disulfide-linked homodimers at the cell surface. CLEC2L expression is fairly tissue-restricted with a predominant expression in the brain. Thus CLEC2L-encoded CTLRs were designated BACL (brain-associated C-type lectin). Combining in situ hybridization and immunohistochemistry, we show that BACL is expressed by neurons in the CNS, with a pronounced expression by Purkinje cells. Notably, the CLEC2L locus is adjacent to another orphan CTLR gene (KLRG2), but reporter cell assays did neither indicate interaction of BACL with the KLRG2 ectodomain nor with human NK cell lines or lymphocytes. Along these lines, growth of BACL-expressing tumor cell lines in immunocompetent mice did not provide evidence for an immune-related function of BACL. Altogether, the CLEC2L gene encodes a homodimeric cell surface CTLR that stands out among CLEC2 family members by its conservation in mammals, its biochemical properties and the predominant expression in the brain. Future studies will have to reveal insights into the functional relevance of BACL in the context of its neuronal expression.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Brain/metabolism
- Cell Line
- Flow Cytometry
- Humans
- Immunoblotting
- Immunohistochemistry
- Immunoprecipitation
- In Situ Hybridization
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Mice
- Mice, Inbred C57BL
- Receptors, NK Cell Lectin-Like/genetics
- Receptors, NK Cell Lectin-Like/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Olga Lysenko
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Dorothea Schulte
- Institute of Neurology (Edinger Institute), Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
- * E-mail:
| |
Collapse
|
48
|
Barber BA, Liyanage VRB, Zachariah RM, Olson CO, Bailey MAG, Rastegar M. Dynamic expression of MEIS1 homeoprotein in E14.5 forebrain and differentiated forebrain-derived neural stem cells. Ann Anat 2013; 195:431-40. [PMID: 23756022 DOI: 10.1016/j.aanat.2013.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/03/2013] [Accepted: 04/09/2013] [Indexed: 01/31/2023]
Abstract
Central nervous system development is controlled by highly conserved homeoprotein transcription factors including HOX and TALE (Three Amino acid Loop Extension). TALE proteins are primarily known as HOX-cofactors and play key roles in cell proliferation, differentiation and organogenesis. MEIS1 is a TALE member with established expression in the developing central nervous system. MEIS1 is essential for embryonic development and Meis1 knockout mice dies at embryonic day (E) 14.5. However, Meis1/MEIS1 expression in the devolving forebrain, at this critical time-point has not been studied. Here, for the first time we characterize the region-specific expression of MEIS1 in E14.5 mouse forebrain, filling the gap of MEIS1 expression profile between E12.5 and E16.5. Previously, we reported MEIS1 transcriptional regulatory role in neuronal differentiation and established forebrain-derived neural stem cells (NSC) for gene therapy application of neuronal genes. Here, we show the dynamic expression of Meis1/MEIS1 during the differentiation of forebrain-derived NSC toward a glial lineage. Our results show that Meis1/MEIS1 expression is induced during NSC differentiation and is expressed in both differentiated neurons and astrocytes. Confirming these results, we detected MEIS1 expression in primary cultures of in vivo differentiated cortical neurons and astrocytes. We further demonstrate Meis1/MEIS1 expression relative to other TALE family members in the forebrain-derived NSC in the absence of Hox genes. Our data provide evidence that forebrain-derived NSC can be used as an accessible in vitro model to study the expression and function of TALE proteins, supporting their potential role in modulating NSC self-renewal and differentiation.
Collapse
Affiliation(s)
- Benjamin A Barber
- Regenerative Medicine Program, Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada
| | | | | | | | | | | |
Collapse
|
49
|
Shaham O, Gueta K, Mor E, Oren-Giladi P, Grinberg D, Xie Q, Cvekl A, Shomron N, Davis N, Keydar-Prizant M, Raviv S, Pasmanik-Chor M, Bell RE, Levy C, Avellino R, Banfi S, Conte I, Ashery-Padan R. Pax6 regulates gene expression in the vertebrate lens through miR-204. PLoS Genet 2013; 9:e1003357. [PMID: 23516376 PMCID: PMC3597499 DOI: 10.1371/journal.pgen.1003357] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 01/17/2013] [Indexed: 12/22/2022] Open
Abstract
During development, tissue-specific transcription factors regulate both protein-coding and non-coding genes to control differentiation. Recent studies have established a dual role for the transcription factor Pax6 as both an activator and repressor of gene expression in the eye, central nervous system, and pancreas. However, the molecular mechanism underlying the inhibitory activity of Pax6 is not fully understood. Here, we reveal that Trpm3 and the intronic microRNA gene miR-204 are co-regulated by Pax6 during eye development. miR-204 is probably the best known microRNA to function as a negative modulator of gene expression during eye development in vertebrates. Analysis of genes altered in mouse Pax6 mutants during lens development revealed significant over-representation of miR-204 targets among the genes up-regulated in the Pax6 mutant lens. A number of new targets of miR-204 were revealed, among them Sox11, a member of the SoxC family of pro-neuronal transcription factors, and an important regulator of eye development. Expression of Trpm/miR-204 and a few of its targets are also Pax6-dependent in medaka fish eyes. Collectively, this study identifies a novel evolutionarily conserved mechanism by which Pax6 controls the down-regulation of multiple genes through direct up-regulation of miR-204. The transcription factor Pax6 is reiteratively employed in space and time for the establishment of progenitor pools and the differentiation of neuronal and non-neuronal lineages of the CNS, pancreas, and eye. Execution of these diverse developmental programs depends on simultaneous activation and repression of gene networks functioning downstream of Pax6. MicroRNAs function as inhibitors of gene expression. Many microRNA genes are transcribed through common promoters of host genes. In this study, using wide-scale analysis of changes in gene expression following Pax6 deletion in the lens, we discover that Pax6 regulates the gene Trpm3 and its hosted microRNA, miR-204. We then show that miR-204 suppresses several target genes in the lens, notably the neuronal gene Sox11. Lastly, by conducting parallel experiments in the medaka fish, we show that Pax6 control of miR-204 and its target genes is evolutionarily conserved between mammals and fish, stressing the biological importance of this pathway. Pax6 regulation of miR-204 explains part of the complex, divergent inhibitory activity of Pax6 in ocular progenitor cells, which is required to establish and maintain the identity and function of ocular tissues.
Collapse
Affiliation(s)
- Ohad Shaham
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Karen Gueta
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eyal Mor
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pazit Oren-Giladi
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dina Grinberg
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Qing Xie
- Department of Genetics and Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Ales Cvekl
- Department of Genetics and Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Noa Davis
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maya Keydar-Prizant
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shaul Raviv
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rachel E. Bell
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Carmit Levy
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Sandro Banfi
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Naples, Italy
- * E-mail: (IC); (RA-P)
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (IC); (RA-P)
| |
Collapse
|
50
|
Gregory-Evans CY, Wallace VA, Gregory-Evans K. Gene networks: dissecting pathways in retinal development and disease. Prog Retin Eye Res 2012; 33:40-66. [PMID: 23128416 DOI: 10.1016/j.preteyeres.2012.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 10/18/2012] [Accepted: 10/19/2012] [Indexed: 01/21/2023]
Abstract
During retinal neurogenesis, diverse cellular subtypes originate from multipotent neural progenitors in a spatiotemporal order leading to a highly specialized laminar structure combined with a distinct mosaic architecture. This is driven by the combinatorial action of transcription factors and signaling molecules which specify cell fate and differentiation. The emerging approach of gene network analysis has allowed a better understanding of the functional relationships between genes expressed in the developing retina. For instance, these gene networks have identified transcriptional hubs that have revealed potential targets and pathways for the development of therapeutic options for retinal diseases. Much of the current knowledge has been informed by targeted gene deletion experiments and gain-of-functional analysis. In this review we will provide an update on retinal development gene networks and address the wider implications for future disease therapeutics.
Collapse
Affiliation(s)
- Cheryl Y Gregory-Evans
- Department of Ophthalmology, University of British Columbia, Vancouver, BC V5Z 3N9, Canada.
| | | | | |
Collapse
|