1
|
Huang J, Cheng R, Liu X, Chen L, Luo T. Association of cortical macrostructural and microstructural changes with cognitive performance and gene expression in subcortical ischemic vascular disease patients with cognitive impairment. Brain Res Bull 2025; 222:111239. [PMID: 39909351 DOI: 10.1016/j.brainresbull.2025.111239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVE Previous researches have demonstrated that patients with subcortical ischemic vascular disease (SIVD) exhibited brain structure abnormalities. However, the cortical macrostructural and microstructural characteristics and their relationship with cognitive scores and gene expression in SIVD patients remain largely unknown. METHODS This study collected 3D-T1 and diffusion tensor imaging data from 30 SIVD patients with cognitive impairment (SIVD-CI) and 32 normal controls. The between-group comparative analyses of cortical thickness, area, volume, local gyrification index (LGI), and mean diffusivity (MD) were conducted with a general linear model. Moreover, the associations between the significant neuroimaging values and the cognitive scores and gene expression values from Allen Human Brain Atlas database were evaluated using partial least squares regression and partial correlation analysis. RESULTS SIVD-CI patients showed significant decreases in cortical thicknesses across 18 regions, cortical volumes across three regions, and cortical LGI across five regions, as well as significant increases in cortical MD across five regions (P < 0.05). The significantly reduced cortical thicknesses of the right insula, left superior temporal gyrus, left central anterior gyrus, and left caudal anterior cingulate cortex, as well as the significantly reduced cortical LGI in left caudal anterior cingulate cortex, were significantly positively correlated with different cognitive scores (P < 0.05). Furthermore, the abnormal cortical structural indicators were found to be significantly related to nine risk genes (VCAN, APOE, EFEMP1, SALL1, BCAN, KCNK2, EPN2, DENND1B and XKR6) (P < 0.05). CONCLUSIONS The specific cortical structural damage may be related to specific cognitive decline and specific risk genes in SIVD-CI patients.
Collapse
Affiliation(s)
- Jing Huang
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Runtian Cheng
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiaoshuang Liu
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Li Chen
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | - Tianyou Luo
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Nieto-Estevez V, Varma P, Mirsadeghi S, Caballero J, Gamero-Alameda S, Hosseini A, Silvosa MJ, Thodeson DM, Lybrand ZR, Giugliano M, Navara C, Hsieh J. Dual effects of ARX poly-alanine mutations in human cortical and interneuron development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577271. [PMID: 38328230 PMCID: PMC10849640 DOI: 10.1101/2024.01.25.577271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Infantile spasms, with an incidence of 1.6 to 4.5 per 10,000 live births, are a relentless and devastating childhood epilepsy marked by severe seizures but also leads to lifelong intellectual disability. Alarmingly, up to 5% of males with this condition carry a mutation in the Aristaless-related homeobox ( ARX ) gene. Our current lack of human-specific models for developmental epilepsy, coupled with discrepancies between animal studies and human data, underscores the gap in knowledge and urgent need for innovative human models, organoids being one of the best available. Here, we used human neural organoid models, cortical organoids (CO) and ganglionic eminences organoids (GEO) which mimic cortical and interneuron development respectively, to study the consequences of PAE mutations, one of the most prevalent mutation in ARX . ARX PAE produces a decrease expression of ARX in GEOs, and an enhancement in interneuron migration. That accelerated migration is cell autonomously driven, and it can be rescued by inhibiting CXCR4. We also found that PAE mutations result in an early increase in radial glia cells and intermediate progenitor cells, followed by a subsequent loss of cortical neurons at later timepoints. Moreover, ARX expression is upregulated in COs derived from patients at 30 DIV and is associated with alterations in the expression of CDKN1C . Furthermore, ARX PAE assembloids had hyperactivity which were evident at early stages of development. With effective treatments for infantile spasms and developmental epilepsies still elusive, delving into the role of ARX PAE mutations in human brain organoids represents a pivotal step toward uncovering groundbreaking therapeutic strategies.
Collapse
|
3
|
Hampl M, Jandová N, Lusková D, Nováková M, Szotkowská T, Čada Š, Procházka J, Kohoutek J, Buchtová M. Early embryogenesis in CHDFIDD mouse model reveals facial clefts and altered cranial neurogenesis. Dis Model Mech 2024; 17:dmm050261. [PMID: 38511331 PMCID: PMC11212636 DOI: 10.1242/dmm.050261] [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: 04/24/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
CDK13-related disorder, also known as congenital heart defects, dysmorphic facial features and intellectual developmental disorder (CHDFIDD) is associated with mutations in the CDK13 gene encoding transcription-regulating cyclin-dependent kinase 13 (CDK13). Here, we focused on the development of craniofacial structures and analyzed early embryonic stages in CHDFIDD mouse models, with one model comprising a hypomorphic mutation in Cdk13 and exhibiting cleft lip/palate, and another model comprising knockout of Cdk13, featuring a stronger phenotype including midfacial cleft. Cdk13 was found to be physiologically expressed at high levels in the mouse embryonic craniofacial structures, namely in the forebrain, nasal epithelium and maxillary mesenchyme. We also uncovered that Cdk13 deficiency leads to development of hypoplastic branches of the trigeminal nerve including the maxillary branch. Additionally, we detected significant changes in the expression levels of genes involved in neurogenesis (Ache, Dcx, Mef2c, Neurog1, Ntn1, Pou4f1) within the developing palatal shelves. These results, together with changes in the expression pattern of other key face-specific genes (Fgf8, Foxd1, Msx1, Meis2 and Shh) at early stages in Cdk13 mutant embryos, demonstrate a key role of CDK13 in the regulation of craniofacial morphogenesis.
Collapse
Affiliation(s)
- Marek Hampl
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 60200 Brno, Czech Republic
| | - Nela Jandová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 60200 Brno, Czech Republic
| | - Denisa Lusková
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Monika Nováková
- Department of Chemistry and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Tereza Szotkowská
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Štěpán Čada
- Department of Experimental Biology, Faculty of Science, Masaryk University, 60200 Brno, Czech Republic
| | - Jan Procházka
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics, Czech Academy of Sciences, 14220 Prague, Czech Republic
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Czech Academy of Sciences, 14220 Prague, Czech Republic
| | - Jiri Kohoutek
- Department of Experimental Biology, Faculty of Science, Masaryk University, 60200 Brno, Czech Republic
| | - Marcela Buchtová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 60200 Brno, Czech Republic
| |
Collapse
|
4
|
Nalamalapu RR, Yue M, Stone AR, Murphy S, Saha MS. The tweety Gene Family: From Embryo to Disease. Front Mol Neurosci 2021; 14:672511. [PMID: 34262434 PMCID: PMC8273234 DOI: 10.3389/fnmol.2021.672511] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 12/31/2022] Open
Abstract
The tweety genes encode gated chloride channels that are found in animals, plants, and even simple eukaryotes, signifying their deep evolutionary origin. In vertebrates, the tweety gene family is highly conserved and consists of three members—ttyh1, ttyh2, and ttyh3—that are important for the regulation of cell volume. While research has elucidated potential physiological functions of ttyh1 in neural stem cell maintenance, proliferation, and filopodia formation during neural development, the roles of ttyh2 and ttyh3 are less characterized, though their expression patterns during embryonic and fetal development suggest potential roles in the development of a wide range of tissues including a role in the immune system in response to pathogen-associated molecules. Additionally, members of the tweety gene family have been implicated in various pathologies including cancers, particularly pediatric brain tumors, and neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease. Here, we review the current state of research using information from published articles and open-source databases on the tweety gene family with regard to its structure, evolution, expression during development and adulthood, biochemical and cellular functions, and role in human disease. We also identify promising areas for further research to advance our understanding of this important, yet still understudied, family of genes.
Collapse
Affiliation(s)
- Rithvik R Nalamalapu
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Michelle Yue
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Aaron R Stone
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Samantha Murphy
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Margaret S Saha
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| |
Collapse
|
5
|
Boulos LJ, Ben Hamida S, Bailly J, Maitra M, Ehrlich AT, Gavériaux-Ruff C, Darcq E, Kieffer BL. Mu opioid receptors in the medial habenula contribute to naloxone aversion. Neuropsychopharmacology 2020; 45:247-255. [PMID: 31005059 PMCID: PMC6901535 DOI: 10.1038/s41386-019-0395-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 02/08/2023]
Abstract
The medial habenula (MHb) is considered a brain center regulating aversive states. The mu opioid receptor (MOR) has been traditionally studied at the level of nociceptive and mesolimbic circuits, for key roles in pain relief and reward processing. MOR is also densely expressed in MHb, however, MOR function at this brain site is virtually unknown. Here we tested the hypothesis that MOR in the MHb (MHb-MOR) also regulates aversion processing. We used chnrb4-Cre driver mice to delete the Oprm1 gene in chnrb4-neurons, predominantly expressed in the MHb. Conditional mutant (B4MOR) mice showed habenula-specific reduction of MOR expression, restricted to chnrb4-neurons (50% MHb-MORs). We tested B4MOR mice in behavioral assays to evaluate effects of MOR activation by morphine, and MOR blockade by naloxone. Locomotor, analgesic, rewarding, and motivational effects of morphine were preserved in conditional mutants. In contrast, conditioned place aversion (CPA) elicited by naloxone was reduced in both naïve (high dose) and morphine-dependent (low dose) B4MOR mice. Further, physical signs of withdrawal precipitated by either MOR (naloxone) or nicotinic receptor (mecamylamine) blockade were attenuated. These data suggest that MORs expressed in MHb B4-neurons contribute to aversive effects of naloxone, including negative effect and aversive effects of opioid withdrawal. MORs are inhibitory receptors, therefore we propose that endogenous MOR signaling normally inhibits chnrb4-neurons of the MHb and moderates their known aversive activity, which is unmasked upon receptor blockade. Thus, in addition to facilitating reward at several brain sites, tonic MOR activity may also limit aversion within the MHb circuitry.
Collapse
Affiliation(s)
- L. J. Boulos
- McGill University, Faculty of Medicine, Douglas Research Centre, Montreal, Canada ,0000 0004 0638 2716grid.420255.4Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France ,0000 0001 2157 9291grid.11843.3fUniversité de Strasbourg, Illkirch, France ,0000 0001 2112 9282grid.4444.0Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U 1258 Illkirch, France
| | - S. Ben Hamida
- McGill University, Faculty of Medicine, Douglas Research Centre, Montreal, Canada ,0000 0004 0638 2716grid.420255.4Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France ,0000 0001 2157 9291grid.11843.3fUniversité de Strasbourg, Illkirch, France ,0000 0001 2112 9282grid.4444.0Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U 1258 Illkirch, France
| | - J. Bailly
- McGill University, Faculty of Medicine, Douglas Research Centre, Montreal, Canada
| | - M. Maitra
- McGill University, Faculty of Medicine, Douglas Research Centre, Montreal, Canada
| | - A. T. Ehrlich
- McGill University, Faculty of Medicine, Douglas Research Centre, Montreal, Canada ,0000 0004 0638 2716grid.420255.4Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France ,0000 0001 2157 9291grid.11843.3fUniversité de Strasbourg, Illkirch, France ,0000 0001 2112 9282grid.4444.0Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U 1258 Illkirch, France
| | - C. Gavériaux-Ruff
- 0000 0004 0638 2716grid.420255.4Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France ,0000 0001 2157 9291grid.11843.3fUniversité de Strasbourg, Illkirch, France ,0000 0001 2112 9282grid.4444.0Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U 1258 Illkirch, France
| | - E. Darcq
- McGill University, Faculty of Medicine, Douglas Research Centre, Montreal, Canada
| | - B. L. Kieffer
- McGill University, Faculty of Medicine, Douglas Research Centre, Montreal, Canada ,0000 0004 0638 2716grid.420255.4Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France ,0000 0001 2157 9291grid.11843.3fUniversité de Strasbourg, Illkirch, France ,0000 0001 2112 9282grid.4444.0Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U 1258 Illkirch, France
| |
Collapse
|
6
|
Klann M, Seaver EC. Functional role of pax6 during eye and nervous system development in the annelid Capitella teleta. Dev Biol 2019; 456:86-103. [PMID: 31445008 DOI: 10.1016/j.ydbio.2019.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 12/18/2022]
Abstract
The transcription factor Pax6 is an important regulator of early animal development. Loss of function mutations of pax6 in a range of animals result in a reduction or complete loss of the eye, a reduction of a subset of neurons, and defects in axon growth. There are no studies focusing on the role of pax6 during development of any lophotrochozoan representative, however, expression of pax6 in the developing eye and nervous system in a number of species suggest that pax6 plays a highly conserved role in eye and nervous system formation. We investigated the functional role of pax6 during development of the marine annelid Capitella teleta. Expression of pax6 transcripts in C. teleta larvae is similar to patterns found in other animals, with distinct subdomains in the brain and ventral nerve cord as well as in the larval and juvenile eye. To perturb pax6 function, two different splice-blocking morpholinos and a translation-blocking morpholino were used. Larvae resulting from microinjections with either splice-blocking morpholino show a reduction of the pax6 transcript. Development of both the larval eyes and the central nervous system architecture are highly disrupted following microinjection of each of the three morpholinos. The less severe phenotype observed when only the homeodomain is disrupted suggests that presence of the paired domain is sufficient for partial function of the Pax6 protein. Preliminary downstream target analysis confirms disruption in expression of some components of the retinal gene regulatory network, as well as disruption of genes involved in nervous system development. Results from this study, taken together with studies from other species, reveal an evolutionarily conserved role for pax6 in eye and neural specification and development.
Collapse
Affiliation(s)
- Marleen Klann
- Whitney Laboratory for Marine Bioscience, University of Florida, 9505 Ocean Shore Blvd, St. Augustine, Fl, 32080, USA
| | - Elaine C Seaver
- Whitney Laboratory for Marine Bioscience, University of Florida, 9505 Ocean Shore Blvd, St. Augustine, Fl, 32080, USA.
| |
Collapse
|
7
|
Ljungberg MC, Sadi M, Wang Y, Aronow BJ, Xu Y, Kao RJ, Liu Y, Gaddis N, Ardini-Poleske ME, Umrod T, Ambalavanan N, Nicola T, Kaminski N, Ahangari F, Sontag R, Corley RA, Ansong C, Carson JP. Spatial distribution of marker gene activity in the mouse lung during alveolarization. Data Brief 2019; 22:365-372. [PMID: 30596133 PMCID: PMC6307338 DOI: 10.1016/j.dib.2018.10.150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 11/28/2022] Open
Abstract
This data is a curated collection of visual images of gene expression patterns from the pre- and post-natal mouse lung, accompanied by associated mRNA probe sequences and RNA-Seq expression profiles. Mammalian lungs undergo significant growth and cellular differentiation before and after the transition to breathing air. Documenting normal lung development is an important step in understanding abnormal lung development, as well as the challenges faced during a preterm birth. Images in this dataset indicate the spatial distribution of mRNA transcripts for over 500 different genes that are active during lung development, as initially determined via RNA-Seq. Images were systematically acquired using high-throughput in situ hybridization with non-radioactive digoxigenin-labeled mRNA probes across mouse lungs from developmental time points E16.5, E18.5, P7, and P28. The dataset was produced as part of The Molecular Atlas of Lung Development Program (LungMAP) and is hosted at https://lungmap.net. This manuscript describes the nature of the data and the protocols for generating the dataset.
Collapse
Affiliation(s)
- M. Cecilia Ljungberg
- Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children׳s Hospital, Houston, TX, USA
| | - Mayce Sadi
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, TX, USA
| | | | - Bruce J. Aronow
- Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH, USA
| | - Yan Xu
- Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH, USA
| | - Rong J. Kao
- Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children׳s Hospital, Houston, TX, USA
| | - Ying Liu
- Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children׳s Hospital, Houston, TX, USA
| | | | | | - Tipparat Umrod
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, TX, USA
| | | | - Teodora Nicola
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Ryan Sontag
- Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Charles Ansong
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - James P. Carson
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
8
|
Singh AJ, Chang CN, Ma HY, Ramsey SA, Filtz TM, Kioussi C. FACS-Seq analysis of Pax3-derived cells identifies non-myogenic lineages in the embryonic forelimb. Sci Rep 2018; 8:7670. [PMID: 29769607 PMCID: PMC5956100 DOI: 10.1038/s41598-018-25998-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/01/2018] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle in the forelimb develops during embryonic and fetal development and perinatally. While much is known regarding the molecules involved in forelimb myogenesis, little is known about the specific mechanisms and interactions. Migrating skeletal muscle precursor cells express Pax3 as they migrate into the forelimb from the dermomyotome. To compare gene expression profiles of the same cell population over time, we isolated lineage-traced Pax3+ cells (Pax3EGFP) from forelimbs at different embryonic days. We performed whole transcriptome profiling via RNA-Seq of Pax3+ cells to construct gene networks involved in different stages of embryonic and fetal development. With this, we identified genes involved in the skeletal, muscular, vascular, nervous and immune systems. Expression of genes related to the immune, skeletal and vascular systems showed prominent increases over time, suggesting a non-skeletal myogenic context of Pax3-derived cells. Using co-expression analysis, we observed an immune-related gene subnetwork active during fetal myogenesis, further implying that Pax3-derived cells are not a strictly myogenic lineage, and are involved in patterning and three-dimensional formation of the forelimb through multiple systems.
Collapse
Affiliation(s)
- Arun J Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA.,Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Hsiao-Yen Ma
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Stephen A Ramsey
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, 97331, USA.,School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Theresa M Filtz
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA.
| |
Collapse
|
9
|
Carson JP, Rennie MY, Danilchik M, Thornburg K, Rugonyi S. A chicken embryo cardiac outflow tract atlas for registering changes due to abnormal blood flow. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2016:1236-1239. [PMID: 28268548 DOI: 10.1109/embc.2016.7590929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Subdivision-based image registration has previously been applied to co-localize digital information extracted from rigid structures in biological specimens, such as the brain. Here, we describe and demonstrate the creation and application of a two-dimensional subdivision-based atlas representing a dynamic structure: the outflow tract of the developing chicken heart. The atlas is designed to segment three different anatomical layers of the outflow tract, and is demonstrated on the characterization of collagen XIV in both control and induced abnormal flow specimens. Abnormal blood flow in the embryonic developing heart can lead to congenital heart disease. Comparing local cellular and sub-cellular changes that are caused by abnormal flow can assist in understanding the molecular pathways involved in maladaptations of the heart and congenital defects. This study demonstrates the approach and potential for more extensive applications of the subdivision-based atlas for the embryonic chicken heart.
Collapse
|
10
|
Cao XL, Zhang X, Zhang YF, Zhang YZ, Song CG, Liu F, Hu YY, Zheng MH, Han H. Expression and purification of mouse Ttyh1 fragments as antigens to generate Ttyh1-specific monoclonal antibodies. Protein Expr Purif 2016; 130:81-89. [PMID: 27678288 DOI: 10.1016/j.pep.2016.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 02/03/2023]
Abstract
Ttyh1 is a murine homolog of the Drosophila Tweety and is predicted as a five-pass transmembrane protein. The Ttyh1 mRNA is expressed in mouse brain tissues with a restricted pattern and in human glioma cells. Ttyh1 protein may function as a large-conductance chloride channel, however, the role of Ttyh1 in normal neural development and tumorigenesis has been largely unknown, at least partially due to the lack of effective antibodies. Here we report the expression in E. coli and purification of two recombinant Ttyh1 protein fragments corresponding to one of the predicted extracellular domains and the carboxyl terminus of the mouse Ttyh1. With these Ttyh1 protein products, a set of monoclonal antibodies (mAbs) against the mouse Ttyh1 protein was established by using conventional hybridoma techniques. The specificity of the anti-Ttyh1 mAbs was determined based on their activities in Western blotting and immunofluorescent analysis using embryonic brain tissues and cultured mouse neural stem cells (NSCs). We also show that the mouse Ttyh1 protein was expressed in cultured NSCs, most likely in membrane and cytoplasm. In mouse embryonic brains, it appeared that the Ttyh1 protein was specifically expressed in the apical edge of the ventricular zone as puncta-like structures, as determined by using immunofluorescence. Taken together, our study provided a useful tool for further exploration of the biological functions and pathological significance of Ttyh1 in mice.
Collapse
Affiliation(s)
- Xiu-Li Cao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xing Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Fei Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yi-Zhe Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Chang-Geng Song
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Fan Liu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yi-Yang Hu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Min-Hua Zheng
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
11
|
Ypsilanti AR, Rubenstein JLR. Transcriptional and epigenetic mechanisms of early cortical development: An examination of how Pax6 coordinates cortical development. J Comp Neurol 2015; 524:609-29. [PMID: 26304102 DOI: 10.1002/cne.23866] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/14/2015] [Accepted: 07/17/2015] [Indexed: 12/26/2022]
Abstract
The development of the cortex is an elaborate process that integrates a plethora of finely tuned molecular processes ranging from carefully regulated gradients of transcription factors, dynamic changes in the chromatin landscape, or formation of protein complexes to elicit and regulate transcription. Combined with cellular processes such as cell type specification, proliferation, differentiation, and migration, all of these developmental processes result in the establishment of an adult mammalian cortex with its typical lamination and regional patterning. By examining in-depth the role of one transcription factor, Pax6, on the regulation of cortical development, its integration in the regulation of chromatin state, and its regulation by cis-regulatory elements, we aim to demonstrate the importance of integrating each level of regulation in our understanding of cortical development.
Collapse
Affiliation(s)
- Athéna R Ypsilanti
- Department of Psychiatry, Neuroscience Program, and the Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, California
| | - John L R Rubenstein
- Department of Psychiatry, Neuroscience Program, and the Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
12
|
Lewandowski JP, Du F, Zhang S, Powell MB, Falkenstein KN, Ji H, Vokes SA. Spatiotemporal regulation of GLI target genes in the mammalian limb bud. Dev Biol 2015; 406:92-103. [PMID: 26238476 DOI: 10.1016/j.ydbio.2015.07.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/22/2015] [Accepted: 07/28/2015] [Indexed: 11/19/2022]
Abstract
GLI proteins convert Sonic hedgehog (Shh) signaling into a transcriptional output in a tissue-specific fashion. The Shh pathway has been extensively studied in the limb bud, where it helps regulate growth through a SHH-FGF feedback loop. However, the transcriptional response is still poorly understood. We addressed this by determining the gene expression patterns of approximately 200 candidate GLI-target genes and identified three discrete SHH-responsive expression domains. GLI-target genes expressed in the three domains are predominately regulated by derepression of GLI3 but have different temporal requirements for SHH. The GLI binding regions associated with these genes harbor both distinct and common DNA motifs. Given the potential for interaction between the SHH and FGF pathways, we also measured the response of GLI-target genes to inhibition of FGF signaling and found the majority were either unaffected or upregulated. These results provide the first characterization of the spatiotemporal response of a large group of GLI-target genes and lay the foundation for a systems-level understanding of the gene regulatory networks underlying SHH-mediated limb patterning.
Collapse
Affiliation(s)
- Jordan P Lewandowski
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| | - Fang Du
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Room E3638, Baltimore, MD 21205, USA
| | - Shilu Zhang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Room E3638, Baltimore, MD 21205, USA
| | - Marian B Powell
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| | - Kristin N Falkenstein
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Room E3638, Baltimore, MD 21205, USA
| | - Steven A Vokes
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, 2500 Speedway Stop A4800, Austin, TX 78712, USA.
| |
Collapse
|
13
|
Saal HM, Prows CA, Guerreiro I, Donlin M, Knudson L, Sund KL, Chang CF, Brugmann SA, Stottmann RW. A mutation in FRIZZLED2 impairs Wnt signaling and causes autosomal dominant omodysplasia. Hum Mol Genet 2015; 24:3399-409. [PMID: 25759469 DOI: 10.1093/hmg/ddv088] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 03/06/2015] [Indexed: 01/18/2023] Open
Abstract
Autosomal dominant omodysplasia is a rare skeletal dysplasia characterized by short humeri, radial head dislocation, short first metacarpals, facial dysmorphism and genitourinary anomalies. We performed next-generation whole-exome sequencing and comparative analysis of a proband with omodysplasia, her unaffected parents and her affected daughter. We identified a de novo mutation in FRIZZLED2 (FZD2) in the proband and her daughter that was not found in unaffected family members. The FZD2 mutation (c.1644G>A) changes a tryptophan residue at amino acid 548 to a premature stop (p.Trp548*). This altered protein is still produced in vitro, but we show reduced ability of this mutant form of FZD2 to interact with its downstream target DISHEVELLED. Furthermore, expressing the mutant form of FZD2 in vitro is not able to facilitate the cellular response to canonical Wnt signaling like wild-type FZD2. We therefore conclude that the FRIZZLED2 mutation is a de novo, novel cause for autosomal dominant omodysplasia.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ching-Fang Chang
- Division of Developmental Biology and Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA
| | - Samantha A Brugmann
- Division of Developmental Biology and Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA
| | - Rolf W Stottmann
- Division of Human Genetics, Division of Developmental Biology and
| |
Collapse
|
14
|
Mieda M, Ono D, Hasegawa E, Okamoto H, Honma KI, Honma S, Sakurai T. Cellular clocks in AVP neurons of the SCN are critical for interneuronal coupling regulating circadian behavior rhythm. Neuron 2015; 85:1103-16. [PMID: 25741730 DOI: 10.1016/j.neuron.2015.02.005] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/10/2014] [Accepted: 01/19/2015] [Indexed: 01/06/2023]
Abstract
The suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals, is a network structure composed of multiple types of neurons. Here, we report that mice with a Bmal1 deletion specific to arginine vasopressin (AVP)-producing neurons showed marked lengthening in the free-running period and activity time of behavior rhythms. When exposed to an abrupt 8-hr advance of the light/dark cycle, these mice reentrained faster than control mice did. In these mice, the circadian expression of genes involved in intercellular communications, including Avp, Prokineticin 2, and Rgs16, was drastically reduced in the dorsal SCN, where AVP neurons predominate. In slices, dorsal SCN cells showed attenuated PER2::LUC oscillation with highly variable and lengthened periods. Thus, Bmal1-dependent oscillators of AVP neurons may modulate the coupling of the SCN network, eventually coupling morning and evening behavioral rhythms, by regulating expression of multiple factors important for the network property of these neurons.
Collapse
Affiliation(s)
- Michihiro Mieda
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Daisuke Ono
- Photonic Bioimaging Section, Research Center for Cooperative Projects, Hokkaido University Graduate School of Medicine, N-15, W-7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Emi Hasegawa
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Hitoshi Okamoto
- Laboratory for Developmental Gene Regulation, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ken-Ichi Honma
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, N-15, W-7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Sato Honma
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, N-15, W-7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Takeshi Sakurai
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| |
Collapse
|
15
|
Romand R, Ripp R, Poidevin L, Boeglin M, Geffers L, Dollé P, Poch O. Integrated annotation and analysis of in situ hybridization images using the ImAnno system: application to the ear and sensory organs of the fetal mouse. PLoS One 2015; 10:e0118024. [PMID: 25706271 PMCID: PMC4338146 DOI: 10.1371/journal.pone.0118024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/30/2014] [Indexed: 11/23/2022] Open
Abstract
An in situ hybridization (ISH) study was performed on 2000 murine genes representing around 10% of the protein-coding genes present in the mouse genome using data generated by the EURExpress consortium. This study was carried out in 25 tissues of late gestation embryos (E14.5), with a special emphasis on the developing ear and on five distinct developing sensory organs, including the cochlea, the vestibular receptors, the sensory retina, the olfactory organ, and the vibrissae follicles. The results obtained from an analysis of more than 11,000 micrographs have been integrated in a newly developed knowledgebase, called ImAnno. In addition to managing the multilevel micrograph annotations performed by human experts, ImAnno provides public access to various integrated databases and tools. Thus, it facilitates the analysis of complex ISH gene expression patterns, as well as functional annotation and interaction of gene sets. It also provides direct links to human pathways and diseases. Hierarchical clustering of expression patterns in the 25 tissues revealed three main branches corresponding to tissues with common functions and/or embryonic origins. To illustrate the integrative power of ImAnno, we explored the expression, function and disease traits of the sensory epithelia of the five presumptive sensory organs. The study identified 623 genes (out of 2000) concomitantly expressed in the five embryonic epithelia, among which many (∼12%) were involved in human disorders. Finally, various multilevel interaction networks were characterized, highlighting differential functional enrichments of directly or indirectly interacting genes. These analyses exemplify an under-represention of "sensory" functions in the sensory gene set suggests that E14.5 is a pivotal stage between the developmental stage and the functional phase that will be fully reached only after birth.
Collapse
Affiliation(s)
- Raymond Romand
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS, INSERM, Université de Strasbourg), BP163, 67404 Illkirch Cedex, France
| | - Raymond Ripp
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS, INSERM, Université de Strasbourg), BP163, 67404 Illkirch Cedex, France
- LBGI Bioinformatique et Génomique Intégratives, ICube Laboratory and Strasbourg Federation of Translational Medecine (FMTS), University of Strasbourg and CNRS, Strasbourg, France
| | - Laetitia Poidevin
- LBGI Bioinformatique et Génomique Intégratives, ICube Laboratory and Strasbourg Federation of Translational Medecine (FMTS), University of Strasbourg and CNRS, Strasbourg, France
| | - Marcel Boeglin
- Imaging & Microscopy Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS, INSERM, Université de Strasbourg), BP163, 67404 Illkirch Cedex, France
| | - Lars Geffers
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077 Göttingen, Germany
| | - Pascal Dollé
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS, INSERM, Université de Strasbourg), BP163, 67404 Illkirch Cedex, France
| | - Olivier Poch
- LBGI Bioinformatique et Génomique Intégratives, ICube Laboratory and Strasbourg Federation of Translational Medecine (FMTS), University of Strasbourg and CNRS, Strasbourg, France
| |
Collapse
|
16
|
Yang G, Smibert CA, Kaplan DR, Miller FD. An eIF4E1/4E-T complex determines the genesis of neurons from precursors by translationally repressing a proneurogenic transcription program. Neuron 2014; 84:723-39. [PMID: 25456498 DOI: 10.1016/j.neuron.2014.10.022] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2014] [Indexed: 02/06/2023]
Abstract
Here, we have addressed the mechanisms that determine genesis of the correct numbers of neurons during development, focusing on the embryonic cortex. We identify in neural precursors a repressive complex involving eIF4E1 and its binding partner 4E-T that coordinately represses translation of proteins that determine neurogenesis. This eIF4E1/4E-T complex is present in granules with the processing body proteins Lsm1 and Rck, and disruption of this complex causes premature and enhanced neurogenesis and neural precursor depletion. Analysis of the 4E-T complex shows that it is highly enriched in mRNAs encoding transcription factors and differentiation-related proteins. These include the proneurogenic bHLH mRNAs, which colocalize with 4E-T in granules and whose protein products are aberrantly upregulated following knockdown of eIF4E, 4E-T, or processing body proteins. Thus, neural precursors are transcriptionally primed to generate neurons, but an eIF4E/4E-T complex sequesters and represses translation of proneurogenic proteins to determine appropriate neurogenesis.
Collapse
Affiliation(s)
- Guang Yang
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada
| | - Craig A Smibert
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David R Kaplan
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Freda D Miller
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
17
|
Liu J, Wang X, Li J, Wang H, Wei G, Yan J. Reconstruction of the gene regulatory network involved in the sonic hedgehog pathway with a potential role in early development of the mouse brain. PLoS Comput Biol 2014; 10:e1003884. [PMID: 25299227 PMCID: PMC4191885 DOI: 10.1371/journal.pcbi.1003884] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 08/28/2014] [Indexed: 11/18/2022] Open
Abstract
The Sonic hedgehog (Shh) signaling pathway is crucial for pattern formation in early central nervous system development. By systematically analyzing high-throughput in situ hybridization data of E11.5 mouse brain, we found that Shh and its receptor Ptch1 define two adjacent mutually exclusive gene expression domains: Shh+Ptch1− and Shh−Ptch1+. These two domains are associated respectively with Foxa2 and Gata3, two transcription factors that play key roles in specifying them. Gata3 ChIP-seq experiments and RNA-seq assays on Gata3-knockdown cells revealed that Gata3 up-regulates the genes that are enriched in the Shh−Ptch1+ domain. Important Gata3 targets include Slit2 and Slit3, which are involved in the process of axon guidance, as well as Slc18a1, Th and Qdpr, which are associated with neurotransmitter synthesis and release. By contrast, Foxa2 both up-regulates the genes expressed in the Shh+Ptch1− domain and down-regulates the genes characteristic of the Shh−Ptch1+ domain. From these and other data, we were able to reconstruct a gene regulatory network governing both domains. Our work provides the first genome-wide characterization of the gene regulatory network involved in the Shh pathway that underlies pattern formation in the early mouse brain. Recent large-scale projects of high-throughput in situ hybridization (ISH) have generated a wealth of spatiotemporal information on gene expression patterns in the early mouse brain. We have developed a computational approach that combines publicly available high-throughput ISH data with our own experimental data to investigate gene regulation, involving signal molecules and transcription factors (TFs), during early brain development. The analysis indicates that two key TFs, Foxa2 and Gata3, play antagonizing roles in the formation of two mutually exclusive domains established by the Sonic hedgehog signaling pathway in the developing mouse brain. Further ChIP-seq and RNA-seq experiments support this hypothesis and have identified novel target genes of Gata3, including the axon guidance regulators Slit2 and Slit3 as well as three neurotransmitter-associated genes, Slc18a1, Th and Qdpr. The findings have allowed us to reconstruct the gene regulatory network brought into play by the Sonic hedgehog pathway that mediates early mouse brain development.
Collapse
Affiliation(s)
- Jinhua Liu
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuelong Wang
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Juan Li
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haifang Wang
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Gang Wei
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Yan
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
18
|
Rodrigues M, Lengerer B, Ostermann T, Ladurner P. Molecular biology approaches in bioadhesion research. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2014; 5:983-93. [PMID: 25161834 PMCID: PMC4142862 DOI: 10.3762/bjnano.5.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 06/17/2014] [Indexed: 06/03/2023]
Abstract
The use of molecular biology tools in the field of bioadhesion is still in its infancy. For new research groups who are considering taking a molecular approach, the techniques presented here are essential to unravelling the sequence of a gene, its expression and its biological function. Here we provide an outline for addressing adhesion-related genes in diverse organisms. We show how to gradually narrow down the number of candidate transcripts that are involved in adhesion by (1) generating a transcriptome and a differentially expressed cDNA list enriched for adhesion-related transcripts, (2) setting up a BLAST search facility, (3) perform an in situ hybridization screen, and (4) functional analyses of selected genes by using RNA interference knock-down. Furthermore, latest developments in genome-editing are presented as new tools to study gene function. By using this iterative multi-technologies approach, the identification, isolation, expression and function of adhesion-related genes can be studied in most organisms. These tools will improve our understanding of the diversity of molecules used for adhesion in different organisms and these findings will help to develop innovative bio-inspired adhesives.
Collapse
Affiliation(s)
- Marcelo Rodrigues
- University of Innsbruck, Institute of Zoology and Center for Molecular Biosciences Innsbruck, Technikerstraße 25, A-6020 Innsbruck, Austria
| | - Birgit Lengerer
- University of Innsbruck, Institute of Zoology and Center for Molecular Biosciences Innsbruck, Technikerstraße 25, A-6020 Innsbruck, Austria
| | - Thomas Ostermann
- University of Innsbruck, Institute of Zoology and Center for Molecular Biosciences Innsbruck, Technikerstraße 25, A-6020 Innsbruck, Austria
| | - Peter Ladurner
- University of Innsbruck, Institute of Zoology and Center for Molecular Biosciences Innsbruck, Technikerstraße 25, A-6020 Innsbruck, Austria
| |
Collapse
|
19
|
Feng X, Scott A, Wang Y, Wang L, Zhao Y, Doerner S, Satake M, Croniger CM, Wang Z. PTPRT regulates high-fat diet-induced obesity and insulin resistance. PLoS One 2014; 9:e100783. [PMID: 24949727 PMCID: PMC4065109 DOI: 10.1371/journal.pone.0100783] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 05/28/2014] [Indexed: 02/03/2023] Open
Abstract
Obesity is a risk factor for many human diseases. However, the underlying molecular causes of obesity are not well understood. Here, we report that protein tyrosine phosphatase receptor T (PTPRT) knockout mice are resistant to high-fat diet-induced obesity. Those mice avoid many deleterious side effects of high-fat diet-induced obesity, displaying improved peripheral insulin sensitivity, lower blood glucose and insulin levels. Compared to wild type littermates, PTPRT knockout mice show reduced food intake. Consistently, STAT3 phosphorylation is up-regulated in the hypothalamus of PTPRT knockout mice. These studies implicate PTPRT-modulated STAT3 signaling in the regulation of high-fat diet-induced obesity.
Collapse
Affiliation(s)
- Xiujing Feng
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Anthony Scott
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yong Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Lan Wang
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yiqing Zhao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Stephanie Doerner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Masanobu Satake
- Department of Molecular Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Colleen M. Croniger
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- Genomic Medicine Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
20
|
Abstract
Embryonic stem (ES) cells have been shown to recapitulate normal developmental stages. They are therefore a highly useful tool in the study of developmental biology. Profiling of ES cell-derived cells has yielded important information about the characteristics of differentiated cells, and allowed the identification of novel marker genes and pathways of differentiation. In this review, we focus on recent results from profiling studies of mouse embryos, human islets, and human ES cell-derived differentiated cells from several research groups. Global gene expression data from mouse embryos have been used to identify novel genes or pathways involved in the developmental process, and to search for transcription factors that regulate direct reprogramming. We introduce gene expression databases of human pancreas cells (Beta Cell Gene Atlas, EuroDia database), and summarize profiling studies of islet- or human ES cell-derived pancreatic cells, with a focus on gene expression, microRNAs, epigenetics, and protein expression. Then, we describe our gene expression profile analyses and our search for novel endoderm, or pancreatic, progenitor marker genes. We differentiated mouse ES cells into mesendoderm, definitive endoderm (DE), mesoderm, ectoderm, and Pdx1-expressing pancreatic lineages, and performed DNA microarray analyses. Genes specifically expressed in DE, and/or in Pdx1-expressing cells, were extracted and their expression patterns in normal embryonic development were studied by in situ hybridization. Out of 54 genes examined, 27 were expressed in the DE of E8.5 mouse embryos, and 15 genes were expressed in distinct domains in the pancreatic buds of E14.5 mouse embryos. Akr1c19, Aebp2, Pbxip1, and Creb3l1 were all novel, and none has been described as being expressed, either in the DE, or in the pancreas. By introducing the profiling results of ES cell-derived cells, the benefits of using ES cells to study early embryonic development will be discussed.
Collapse
Affiliation(s)
- Nobuaki Shiraki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | - Soichiro Ogaki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | - Shoen Kume
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| |
Collapse
|
21
|
Stoya G, Redies C, Schmid-Hertel N. Inversion of layer-specific cadherin expression profiles and maintenance of cytoarchitectonic areas in the allocortex of the reeler mutant mouse. J Comp Neurol 2014; 522:3106-19. [PMID: 24591110 DOI: 10.1002/cne.23572] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 02/27/2014] [Accepted: 02/27/2014] [Indexed: 11/11/2022]
Abstract
Cadherins are calcium-depending cell adhesion proteins that play critical roles in brain morphogenesis and wiring. They provide an adhesive code for the development of cortical layers, due to their homophilic interactions and their restricted spatiotemporal expression patterns. In the adult organism, cadherins are involved in the maintenance and plasticity of neuronal circuits that play a role in learning. A well-known model for studying corticogenesis is the reeler mouse model. Numerous investigations of neocortical development suggest that, in the reeler mutant mouse, the lack of the protein Reelin results in cell-type and region-dependent changes of the neocortical layers. To investigate in detail how layer formation and regionalization is perturbed in the phylogenetically older archicortex of the adult reeler mutant mouse, we studied the expression of 11 different cadherins (Cdh4, Cdh7, Cdh8, Cdh11, Pcdh1, Pcdh7, Pcdh8, Pcdh9, Pcdh10, Pcdh17, and Pcdh19) and of the transcription factors ER81 and Cux2 by in situ hybridization in the (peri-)archicortex. All cadherins studied show a layer-specific expression in the (peri-)archicortex of the wildtype brain. In the archicortex of the reeler mutant, the cadherin-expressing cell layers are dispersed in the radial dimension, whereas in the periarchicortex the superficial and deep layers are inverted, both in the adult and during development. Possibly, this inversion relates to the histoarchitectural division of the reeler entorhinal cortex into an external and an internal zone. The regionalized, gradient-like expression of the cadherins is preserved in the reeler mutant mouse.
Collapse
Affiliation(s)
- Gudrun Stoya
- Institute of Anatomy, Friedrich Schiller University School of Medicine, Jena University Hospital, 07743, Jena, Germany
| | | | | |
Collapse
|
22
|
Lee K, Mattiske T, Kitamura K, Gecz J, Shoubridge C. Reduced polyalanine-expanded Arx mutant protein in developing mouse subpallium alters Lmo1 transcriptional regulation. Hum Mol Genet 2013; 23:1084-94. [PMID: 24122442 DOI: 10.1093/hmg/ddt503] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Intellectual disability (ID) is a highly prevalent disorder that affects 1-3% of the population. The Aristaless-related homeobox gene (ARX) is a frequently mutated X-linked ID gene and encodes a transcription factor indispensable for proper forebrain, testis and pancreas development. Polyalanine expansions account for over half of all mutations in ARX and clinically give rise to a spectrum of ID and seizures. To understand how the polyalanine expansions cause the clinical phenotype, we studied mouse models of the two most frequent polyalanine expansion mutations (Arx((GCG)7) and Arx(432-455dup24)). Neither model showed evidence of protein aggregates; however, a marked reduction of Arx protein abundance within the developing forebrain was striking. Examining the expression of known Arx target genes, we found a more prominent loss of Lmo1 repression in Arx((GCG7)/Y) compared with Arx(432-455dup24/Y) mice at 12.5 and 14.5 dpc, stages of peak neural proliferation and neurogenesis, respectively. Once neurogenesis concludes both mutant mouse models showed similar loss of Lmo1 repression. We propose that this temporal difference in the loss of Lmo1 repression may be one of the causes accounting for the phenotypic differences identified between the Arx((GCG)7)and Arx(432-455dup24) mouse models. It is yet to be determined what effect these mutations have on ARX protein in affected males in the human setting.
Collapse
Affiliation(s)
- Kristie Lee
- Robinson Institute, Department of Paediatrics, University of Adelaide, Adelaide, South Australia 5005, Australia
| | | | | | | | | |
Collapse
|
23
|
Di Schiavi E, Andrenacci D. Invertebrate models of kallmann syndrome: molecular pathogenesis and new disease genes. Curr Genomics 2013; 14:2-10. [PMID: 23997646 PMCID: PMC3580776 DOI: 10.2174/138920213804999174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 10/08/2012] [Accepted: 10/08/2012] [Indexed: 11/23/2022] Open
Abstract
Kallmann Syndrome is a heritable disorder characterized by congenital anosmia, hypogonadotropic hypogonadism and, less frequently, by other symptoms. The X-linked form of this syndrome is caused by mutations affecting the KAL1 gene that codes for the extracellular protein anosmin-1. Investigation of KAL1 function in mice has been hampered by the fact that the murine ortholog has not been identified. Thus studies performed in other animal models have contributed significantly to an understanding of the function of KAL1. In this review, the main results obtained using the two invertebrate models, the nematode worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster, are illustrated and the contribution provided by them to the elucidation of the molecular pathogenesis of Kallmann Syndrome is discussed in detail. Structure-function dissection studies performed in these two animal models have shown how the different domains of anosmin-1 carry out specific functions, also suggesting a novel intramolecular regulation mechanism among the different domains of the protein. The model that emerges is one in which anosmin-1 plays different roles in different tissues, interacting with different components of the extracellular matrix. We also describe how the genetic approach in C. elegans has allowed the discovery of the genes involved in KAL1-heparan sulfate proteoglycans interactions and the identification of HS6ST1 as a new disease gene.
Collapse
Affiliation(s)
- Elia Di Schiavi
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | | |
Collapse
|
24
|
Puehringer D, Orel N, Lüningschrör P, Subramanian N, Herrmann T, Chao MV, Sendtner M. EGF transactivation of Trk receptors regulates the migration of newborn cortical neurons. Nat Neurosci 2013; 16:407-15. [PMID: 23416450 DOI: 10.1038/nn.3333] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 01/17/2013] [Indexed: 12/13/2022]
Abstract
The development of neuronal networks in the neocortex depends on control mechanisms for mitosis and migration that allow newborn neurons to find their accurate position. Multiple mitogens, neurotrophic factors, guidance molecules and their corresponding receptors are involved in this process, but the mechanisms by which these signals are integrated are only poorly understood. We found that TrkB and TrkC, the receptors for brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), are activated by epidermal growth factor receptor (EGFR) signaling rather than by BDNF or NT-3 in embryonic mouse cortical precursor cells. This transactivation event regulated migration of early neuronal cells to their final position in the developing cortex. Transactivation by EGF led to membrane translocation of TrkB, promoting its signaling responsiveness. Our results provide genetic evidence that TrkB and TrkC activation in early cortical neurons do not depend on BDNF and NT-3, but instead on transactivation by EGFR signaling.
Collapse
Affiliation(s)
- Dirk Puehringer
- Institute for Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Geffers L, Tetzlaff B, Cui X, Yan J, Eichele G. METscout: a pathfinder exploring the landscape of metabolites, enzymes and transporters. Nucleic Acids Res 2013; 41:D1047-54. [PMID: 23019219 PMCID: PMC3531068 DOI: 10.1093/nar/gks886] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 08/31/2012] [Indexed: 11/12/2022] Open
Abstract
METscout (http://metscout.mpg.de) brings together metabolism and gene expression landscapes. It is a MySQL relational database linking biochemical pathway information with 3D patterns of gene expression determined by robotic in situ hybridization in the E14.5 mouse embryo. The sites of expression of ∼1500 metabolic enzymes and of ∼350 solute carriers (SLCs) were included and are accessible as single cell resolution images and in the form of semi-quantitative image abstractions. METscout provides several graphical web-interfaces allowing navigation through complex anatomical and metabolic information. Specifically, the database shows where in the organism each of the many metabolic reactions take place and where SLCs transport metabolites. To link enzymatic reactions and transport, the KEGG metabolic reaction network was extended to include metabolite transport. This network in conjunction with spatial expression pattern of the network genes allows for a tracing of metabolic reactions and transport processes across the entire body of the embryo.
Collapse
Affiliation(s)
- Lars Geffers
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Anwendungs- und Informationssysteme, Gesellschaft für wissenschaftliche Datenverarbeitung mbH Göttingen, Am Faßberg 11, 37077 Göttingen, Germany and Functional Genomics Group, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, 200031 Shanghai, China
| | - Benjamin Tetzlaff
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Anwendungs- und Informationssysteme, Gesellschaft für wissenschaftliche Datenverarbeitung mbH Göttingen, Am Faßberg 11, 37077 Göttingen, Germany and Functional Genomics Group, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, 200031 Shanghai, China
| | - Xiao Cui
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Anwendungs- und Informationssysteme, Gesellschaft für wissenschaftliche Datenverarbeitung mbH Göttingen, Am Faßberg 11, 37077 Göttingen, Germany and Functional Genomics Group, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, 200031 Shanghai, China
| | - Jun Yan
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Anwendungs- und Informationssysteme, Gesellschaft für wissenschaftliche Datenverarbeitung mbH Göttingen, Am Faßberg 11, 37077 Göttingen, Germany and Functional Genomics Group, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, 200031 Shanghai, China
| | - Gregor Eichele
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Anwendungs- und Informationssysteme, Gesellschaft für wissenschaftliche Datenverarbeitung mbH Göttingen, Am Faßberg 11, 37077 Göttingen, Germany and Functional Genomics Group, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, 200031 Shanghai, China
| |
Collapse
|
26
|
Talchai C, Xuan S, Lin HV, Sussel L, Accili D. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell 2012; 150:1223-34. [PMID: 22980982 DOI: 10.1016/j.cell.2012.07.029] [Citation(s) in RCA: 1098] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 03/20/2012] [Accepted: 07/06/2012] [Indexed: 12/12/2022]
Abstract
Diabetes is associated with β cell failure. But it remains unclear whether the latter results from reduced β cell number or function. FoxO1 integrates β cell proliferation with adaptive β cell function. We interrogated the contribution of these two processes to β cell dysfunction, using mice lacking FoxO1 in β cells. FoxO1 ablation caused hyperglycemia with reduced β cell mass following physiologic stress, such as multiparity and aging. Surprisingly, lineage-tracing experiments demonstrated that loss of β cell mass was due to β cell dedifferentiation, not death. Dedifferentiated β cells reverted to progenitor-like cells expressing Neurogenin3, Oct4, Nanog, and L-Myc. A subset of FoxO1-deficient β cells adopted the α cell fate, resulting in hyperglucagonemia. Strikingly, we identify the same sequence of events as a feature of different models of murine diabetes. We propose that dedifferentiation trumps endocrine cell death in the natural history of β cell failure and suggest that treatment of β cell dysfunction should restore differentiation, rather than promoting β cell replication.
Collapse
Affiliation(s)
- Chutima Talchai
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
27
|
Nfonsam LE, Cano C, Mudge J, Schilkey FD, Curtiss J. Analysis of the transcriptomes downstream of Eyeless and the Hedgehog, Decapentaplegic and Notch signaling pathways in Drosophila melanogaster. PLoS One 2012; 7:e44583. [PMID: 22952997 PMCID: PMC3432130 DOI: 10.1371/journal.pone.0044583] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 08/09/2012] [Indexed: 01/22/2023] Open
Abstract
Tissue-specific transcription factors are thought to cooperate with signaling pathways to promote patterned tissue specification, in part by co-regulating transcription. The Drosophila melanogaster Pax6 homolog Eyeless forms a complex, incompletely understood regulatory network with the Hedgehog, Decapentaplegic and Notch signaling pathways to control eye-specific gene expression. We report a combinatorial approach, including mRNAseq and microarray analyses, to identify targets co-regulated by Eyeless and Hedgehog, Decapentaplegic or Notch. Multiple analyses suggest that the transcriptomes resulting from co-misexpression of Eyeless+signaling factors provide a more complete picture of eye development compared to previous efforts involving Eyeless alone: (1) Principal components analysis and two-way hierarchical clustering revealed that the Eyeless+signaling factor transcriptomes are closer to the eye control transcriptome than when Eyeless is misexpressed alone; (2) more genes are upregulated at least three-fold in response to Eyeless+signaling factors compared to Eyeless alone; (3) based on gene ontology analysis, the genes upregulated in response to Eyeless+signaling factors had a greater diversity of functions compared to Eyeless alone. Through a secondary screen that utilized RNA interference, we show that the predicted gene CG4721 has a role in eye development. CG4721 encodes a neprilysin family metalloprotease that is highly up-regulated in response to Eyeless+Notch, confirming the validity of our approach. Given the similarity between D. melanogaster and vertebrate eye development, the large number of novel genes identified as potential targets of Ey+signaling factors will provide novel insights to our understanding of eye development in D. melanogaster and humans.
Collapse
Affiliation(s)
- Landry E. Nfonsam
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Carlos Cano
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Joann Mudge
- National Center for Genome Resources, Santa Fe, New Mexico, United States of America
| | - Faye D. Schilkey
- National Center for Genome Resources, Santa Fe, New Mexico, United States of America
| | - Jennifer Curtiss
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| |
Collapse
|
28
|
Geffers L, Herrmann B, Eichele G. Web-based digital gene expression atlases for the mouse. Mamm Genome 2012; 23:525-38. [PMID: 22936000 DOI: 10.1007/s00335-012-9413-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/04/2012] [Indexed: 11/26/2022]
Abstract
Over the past 15 years the publicly available mouse gene expression data determined by in situ hybridization have dramatically increased in scope and spatiotemporal resolution. As a consequence of resources and tools available in the post-genomic era, full transcriptomes in the mouse brain and in the mouse embryo can be studied. Here we introduce and discuss seven current databases (MAMEP, EMBRYS, GenePaint, EURExpress, EuReGene, BGEM, and GENSAT) that grant access to large collections of expression data in mouse. We review the experimental focus, coverage, data assessment, and annotation for each of these databases and the implementation of analytic tools and links to other relevant databases. We provide a user-oriented summary of how to interrogate each database.
Collapse
Affiliation(s)
- Lars Geffers
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany.
| | | | | |
Collapse
|
29
|
Eberwine J, Lovatt D, Buckley P, Dueck H, Francis C, Kim TK, Lee J, Lee M, Miyashiro K, Morris J, Peritz T, Schochet T, Spaethling J, Sul JY, Kim J. Quantitative biology of single neurons. J R Soc Interface 2012; 9:3165-83. [PMID: 22915636 PMCID: PMC3481569 DOI: 10.1098/rsif.2012.0417] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The building blocks of complex biological systems are single cells. Fundamental insights gained from single-cell analysis promise to provide the framework for understanding normal biological systems development as well as the limits on systems/cellular ability to respond to disease. The interplay of cells to create functional systems is not well understood. Until recently, the study of single cells has concentrated primarily on morphological and physiological characterization. With the application of new highly sensitive molecular and genomic technologies, the quantitative biochemistry of single cells is now accessible.
Collapse
Affiliation(s)
- James Eberwine
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, 36th and Hamilton Walk, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ramialison M, Reinhardt R, Henrich T, Wittbrodt B, Kellner T, Lowy CM, Wittbrodt J. Cis-regulatory properties of medaka synexpression groups. Development 2012; 139:917-28. [PMID: 22318626 DOI: 10.1242/dev.071803] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
During embryogenesis, tissue specification is triggered by the expression of a unique combination of developmental genes and their expression in time and space is crucial for successful development. Synexpression groups are batteries of spatiotemporally co-expressed genes that act in shared biological processes through their coordinated expression. Although several synexpression groups have been described in numerous vertebrate species, the regulatory mechanisms that orchestrate their common complex expression pattern remain to be elucidated. Here we performed a pilot screen on 560 genes of the vertebrate model system medaka (Oryzias latipes) to systematically identify synexpression groups and investigate their regulatory properties by searching for common regulatory cues. We find that synexpression groups share DNA motifs that are arranged in various combinations into cis-regulatory modules that drive co-expression. In contrast to previous assumptions that these genes are located randomly in the genome, we discovered that genes belonging to the same synexpression group frequently occur in synexpression clusters in the genome. This work presents a first repertoire of synexpression group common signatures, a resource that will contribute to deciphering developmental gene regulatory networks.
Collapse
Affiliation(s)
- Mirana Ramialison
- University of Heidelberg, Centre for Organismal Studies, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
31
|
Shaham O, Menuchin Y, Farhy C, Ashery-Padan R. Pax6: a multi-level regulator of ocular development. Prog Retin Eye Res 2012; 31:351-76. [PMID: 22561546 DOI: 10.1016/j.preteyeres.2012.04.002] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 04/19/2012] [Accepted: 04/24/2012] [Indexed: 02/08/2023]
Abstract
Eye development has been a paradigm for the study of organogenesis, from the demonstration of lens induction through epithelial tissue morphogenesis, to neuronal specification and differentiation. The transcription factor Pax6 has been shown to play a key role in each of these processes. Pax6 is required for initiation of developmental pathways, patterning of epithelial tissues, activation of tissue-specific genes and interaction with other regulatory pathways. Herein we examine the data accumulated over the last few decades from extensive analyses of biochemical modules and genetic manipulation of the Pax6 gene. Specifically, we describe the regulation of Pax6's expression pattern, the protein's DNA-binding properties, and its specific roles and mechanisms of action at all stages of lens and retinal development. Pax6 functions at multiple levels to integrate extracellular information and execute cell-intrinsic differentiation programs that culminate in the specification and differentiation of a distinct ocular lineage.
Collapse
Affiliation(s)
- Ohad Shaham
- Sackler Faculty of Medicine, Department of Human Molecular Genetics and Biochemistry, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
32
|
Kiselev Y, Eriksen TE, Forsdahl S, Nguyen LHT, Mikkola I. 3T3 cell lines stably expressing Pax6 or Pax6(5a)--a new tool used for identification of common and isoform specific target genes. PLoS One 2012; 7:e31915. [PMID: 22384097 PMCID: PMC3285655 DOI: 10.1371/journal.pone.0031915] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 01/19/2012] [Indexed: 12/03/2022] Open
Abstract
Pax6 and Pax6(5a) are two isoforms of the evolutionary conserved Pax6 gene often co-expressed in specific stochiometric relationship in the brain and the eye during development. The Pax6(5a) protein differs from Pax6 by having a 14 amino acid insert in the paired domain, causing the two proteins to have different DNA binding specificities. Difference in functions during development is proven by the fact that mutations in the 14 amino acid insertion for Pax6(5a) give a slightly different eye phenotype than the one described for Pax6. Whereas quite many Pax6 target genes have been published during the last years, few Pax6(5a) specific target genes have been reported on. However, target genes identified by Pax6 knockout studies can probably be Pax6(5a) targets as well, since this isoform also will be affected by the knockout. In order to identify new Pax6 target genes, and to try to distinguish between genes regulated by Pax6 and Pax6(5a), we generated FlpIn-3T3 cell lines stably expressing Pax6 or Pax6(5a). RNA was harvested from these cell lines and used in gene expression microarrays where we identified a number of genes differentially regulated by Pax6 and Pax6(5a). A majority of these were associated with the extracellular region. By qPCR we verified that Ncam1, Ngef, Sphk1, Dkk3 and Crtap are Pax6(5a) specific target genes, while Tgfbi, Vegfa, EphB2, Klk8 and Edn1 were confirmed as Pax6 specific target genes. Nbl1, Ngfb and seven genes encoding different glycosyl transferases appeared to be regulated by both. Direct binding to the promoters of Crtap, Ctgf, Edn1, Dkk3, Pdgfb and Ngef was verified by ChIP. Furthermore, a change in morphology of the stably transfected Pax6 and Pax6(5a) cells was observed, and the Pax6 expressing cells were shown to have increased proliferation and migration capacities.
Collapse
Affiliation(s)
| | | | | | | | - Ingvild Mikkola
- Research Group of Pharmacology, Department of Pharmacy, University of Tromsø, Tromsø, Norway
- * E-mail:
| |
Collapse
|
33
|
Whelan G, Kreidl E, Wutz G, Egner A, Peters JM, Eichele G. Cohesin acetyltransferase Esco2 is a cell viability factor and is required for cohesion in pericentric heterochromatin. EMBO J 2012; 31:71-82. [PMID: 22101327 PMCID: PMC3252581 DOI: 10.1038/emboj.2011.381] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 09/22/2011] [Indexed: 01/25/2023] Open
Abstract
Sister chromatid cohesion, mediated by cohesin and regulated by Sororin, is essential for chromosome segregation. In mammalian cells, cohesion establishment and Sororin recruitment to chromatin-bound cohesin depends on the acetyltransferases Esco1 and Esco2. Mutations in Esco2 cause Roberts syndrome, a developmental disease in which mitotic chromosomes have a 'railroad' track morphology. Here, we show that Esco2 deficiency leads to termination of mouse development at pre- and post-implantation stages, indicating that Esco2 functions non-redundantly with Esco1. Esco2 is transiently expressed during S-phase when it localizes to pericentric heterochromatin (PCH). In interphase, Esco2 depletion leads to a reduction in cohesin acetylation and Sororin recruitment to chromatin. In early mitosis, Esco2 deficiency causes changes in the chromosomal localization of cohesin and its protector Sgo1. Our results suggest that Esco2 is needed for cohesin acetylation in PCH and that this modification is required for the proper distribution of cohesin on mitotic chromosomes and for centromeric cohesion.
Collapse
Affiliation(s)
- Gabriela Whelan
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Emanuel Kreidl
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Gordana Wutz
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Alexander Egner
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | | | - Gregor Eichele
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| |
Collapse
|
34
|
Xie Q, Cvekl A. The orchestration of mammalian tissue morphogenesis through a series of coherent feed-forward loops. J Biol Chem 2011; 286:43259-71. [PMID: 21998302 DOI: 10.1074/jbc.m111.264580] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Tissue morphogenesis requires intricate temporal and spatial control of gene expression that is executed through specific gene regulatory networks (GRNs). GRNs are comprised from individual subcircuits of different levels of complexity. An important question is to elucidate the mutual relationship between those genes encoding DNA-binding factors that trigger the subcircuit with those that play major "later" roles during terminal differentiation via expression of specific genes that constitute the phenotype of individual tissues. The ocular lens is a classical model system to study tissue morphogenesis. Pax6 is essential for both lens placode formation and subsequent stages of lens morphogenesis, whereas c-Maf controls terminal differentiation of lens fibers, including regulation of crystallins, key lens structural proteins required for its transparency and refraction. Here, we show that Pax6 directly regulates c-Maf expression during lens development. A 1.3-kb c-Maf promoter with a 1.6-kb upstream enhancer (CR1) recapitulated the endogenous c-Maf expression pattern in lens and retinal pigmented epithelium. ChIP assays revealed binding of Pax6 and c-Maf to multiple regions of the c-Maf locus in lens chromatin. To predict functional Pax6-binding sites, nine novel variants of Pax6 DNA-binding motifs were identified and characterized. Two of these motifs predicted a pair of Pax6-binding sites in the CR1. Mutagenesis of these Pax6-binding sites inactivated transgenic expression in the lens but not in retinal pigmented epithelium. These data establish a novel regulatory role for Pax6 during lens development, link together the Pax6/c-Maf/crystallin regulatory network, and suggest a novel type of GRN subcircuit that controls a major part of embryonic lens development.
Collapse
Affiliation(s)
- Qing Xie
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
35
|
Husse J, Zhou X, Shostak A, Oster H, Eichele G. Synaptotagmin10-Cre, a driver to disrupt clock genes in the SCN. J Biol Rhythms 2011; 26:379-89. [PMID: 21921292 DOI: 10.1177/0748730411415363] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Surgical lesion of the suprachiasmatic nuclei (SCN) profoundly affects the circadian timing system. A complication of SCN ablations is the concomitant scission of SCN afferents and efferents. Genetic disruption of the molecular clockwork in the SCN provides a complementary, less invasive experimental approach. The authors report the generation and functional analysis of a new Cre recombinase driver mouse that evokes homologous recombination with high efficiency in the SCN. They inserted the Cre recombinase cDNA into the Synaptotagmin10 (Syt10) locus, a gene strongly expressed in the SCN. Heterozygous Synaptotagmin10-Cre (Syt10(Cre)) mice have no obvious circadian locomotor phenotype, and homozygous animals show slightly reduced light-induced phase delays. Crosses of Syt10(Cre) mice with β-galactosidase reporter animals revealed strong Cre activity in the vast majority of SCN cells. Cre activity is not detected in nonneuronal tissues with the exception of the testis. The authors demonstrate that conditionally deleting the clock gene Bmal1 using the Syt10(Cre) driver renders animals arrhythmic.
Collapse
Affiliation(s)
- Jana Husse
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | | | | | | | | |
Collapse
|
36
|
Coutinho P, Pavlou S, Bhatia S, Chalmers KJ, Kleinjan DA, van Heyningen V. Discovery and assessment of conserved Pax6 target genes and enhancers. Genome Res 2011; 21:1349-59. [PMID: 21617155 DOI: 10.1101/gr.124115.111] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The characterization of transcriptional networks (TNs) is essential for understanding complex biological phenomena such as development, disease, and evolution. In this study, we have designed and implemented a procedure that combines in silico target screens with zebrafish and mouse validation, in order to identify cis-elements and genes directly regulated by Pax6. We chose Pax6 as the paradigm because of its crucial roles in organogenesis and human disease. We identified over 600 putative Pax6 binding sites and more than 200 predicted direct target genes, conserved in evolution from zebrafish to human and to mouse. This was accomplished using hidden Markov models (HMMs) generated from experimentally validated Pax6 binding sites. A small sample of genes, expressed in the neural lineage, was chosen from the predictions for RNA in situ validation using zebrafish and mouse models. Validation of DNA binding to some predicted cis-elements was also carried out using chromatin immunoprecipitation (ChIP) and zebrafish reporter transgenic studies. The results show that this combined procedure is a highly efficient tool to investigate the architecture of TNs and constitutes a useful complementary resource to ChIP and expression data sets because of its inherent spatiotemporal independence. We have identified several novel direct targets, including some putative disease genes, among them Foxp2; these will allow further dissection of Pax6 function in development and disease.
Collapse
Affiliation(s)
- Pedro Coutinho
- Medical Research Council (MRC) Human Genetics Unit, Western General Hospital, Edinburgh EH4 2XU, UK.
| | | | | | | | | | | |
Collapse
|
37
|
Lachke SA, Maas RL. Building the developmental oculome: systems biology in vertebrate eye development and disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 2:305-323. [PMID: 20836031 DOI: 10.1002/wsbm.59] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The vertebrate eye is a sophisticated multicomponent organ that has been actively studied for over a century, resulting in the identification of the major embryonic and molecular events involved in its complex developmental program. Data gathered so far provides sufficient information to construct a rudimentary network of the various signaling molecules, transcription factors, and their targets for several key stages of this process. With the advent of genomic technologies, there has been a rapid expansion in our ability to collect and process biological information, and the use of systems-level approaches to study specific aspects of vertebrate eye development has already commenced. This is beginning to result in the definition of the dynamic developmental networks that operate in ocular tissues, and the interactions of such networks between coordinately developing ocular tissues. Such an integrative understanding of the eye by a comprehensive systems-level analysis can be termed the 'oculome', and that of serial developmental stages of the eye as it transits from its initiation to a fully formed functional organ represents the 'developmental oculome'. Construction of the developmental oculome will allow novel mechanistic insights that are essential for organ regeneration-based therapeutic applications, and the generation of computational models for eye disease states to predict the effects of drugs. This review discusses our present understanding of two of the individual components of the developing vertebrate eye--the lens and retina--at both the molecular and systems levels, and outlines the directions and tools required for construction of the developmental oculome.
Collapse
Affiliation(s)
- Salil A Lachke
- Division of Genetics, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Richard L Maas
- Division of Genetics, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
38
|
Diez-Roux G, Banfi S, Sultan M, Geffers L, Anand S, Rozado D, Magen A, Canidio E, Pagani M, Peluso I, Lin-Marq N, Koch M, Bilio M, Cantiello I, Verde R, De Masi C, Bianchi SA, Cicchini J, Perroud E, Mehmeti S, Dagand E, Schrinner S, Nürnberger A, Schmidt K, Metz K, Zwingmann C, Brieske N, Springer C, Hernandez AM, Herzog S, Grabbe F, Sieverding C, Fischer B, Schrader K, Brockmeyer M, Dettmer S, Helbig C, Alunni V, Battaini MA, Mura C, Henrichsen CN, Garcia-Lopez R, Echevarria D, Puelles E, Garcia-Calero E, Kruse S, Uhr M, Kauck C, Feng G, Milyaev N, Ong CK, Kumar L, Lam M, Semple CA, Gyenesei A, Mundlos S, Radelof U, Lehrach H, Sarmientos P, Reymond A, Davidson DR, Dollé P, Antonarakis SE, Yaspo ML, Martinez S, Baldock RA, Eichele G, Ballabio A. A high-resolution anatomical atlas of the transcriptome in the mouse embryo. PLoS Biol 2011; 9:e1000582. [PMID: 21267068 PMCID: PMC3022534 DOI: 10.1371/journal.pbio.1000582] [Citation(s) in RCA: 486] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 12/06/2010] [Indexed: 11/23/2022] Open
Abstract
The manuscript describes the “digital transcriptome atlas” of the developing mouse embryo, a powerful resource to determine co-expression of genes, to identify cell populations and lineages and to identify functional associations between genes relevant to development and disease. Ascertaining when and where genes are expressed is of crucial importance to understanding or predicting the physiological role of genes and proteins and how they interact to form the complex networks that underlie organ development and function. It is, therefore, crucial to determine on a genome-wide level, the spatio-temporal gene expression profiles at cellular resolution. This information is provided by colorimetric RNA in situ hybridization that can elucidate expression of genes in their native context and does so at cellular resolution. We generated what is to our knowledge the first genome-wide transcriptome atlas by RNA in situ hybridization of an entire mammalian organism, the developing mouse at embryonic day 14.5. This digital transcriptome atlas, the Eurexpress atlas (http://www.eurexpress.org), consists of a searchable database of annotated images that can be interactively viewed. We generated anatomy-based expression profiles for over 18,000 coding genes and over 400 microRNAs. We identified 1,002 tissue-specific genes that are a source of novel tissue-specific markers for 37 different anatomical structures. The quality and the resolution of the data revealed novel molecular domains for several developing structures, such as the telencephalon, a novel organization for the hypothalamus, and insight on the Wnt network involved in renal epithelial differentiation during kidney development. The digital transcriptome atlas is a powerful resource to determine co-expression of genes, to identify cell populations and lineages, and to identify functional associations between genes relevant to development and disease. In situ hybridization (ISH) can be used to visualize gene expression in cells and tissues in their native context. High-throughput ISH using nonradioactive RNA probes allowed the Eurexpress consortium to generate a comprehensive, interactive, and freely accessible digital gene expression atlas, the Eurexpress transcriptome atlas (http://www.eurexpress.org), of the E14.5 mouse embryo. Expression data for over 15,000 genes were annotated for hundreds of anatomical structures, thus allowing us to systematically identify tissue-specific and tissue-overlapping gene networks. We illustrate the value of the Eurexpress atlas by finding novel regional subdivisions in the developing brain. We also use the transcriptome atlas to allocate specific components of the complex Wnt signaling pathway to kidney development, and we identify regionally expressed genes in liver that may be markers of hematopoietic stem cell differentiation.
Collapse
Affiliation(s)
| | - Sandro Banfi
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Marc Sultan
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lars Geffers
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Santosh Anand
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - David Rozado
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Alon Magen
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | | | - Ivana Peluso
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Nathalie Lin-Marq
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Muriel Koch
- Institut Clinique de la Souris, Illkirch, France
| | - Marchesa Bilio
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | | | - Roberta Verde
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | | | | | - Juliette Cicchini
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Elodie Perroud
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Shprese Mehmeti
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Emilie Dagand
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Asja Nürnberger
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Katja Schmidt
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Katja Metz
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Norbert Brieske
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Cindy Springer
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Ana Martinez Hernandez
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Sarah Herzog
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Frauke Grabbe
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Cornelia Sieverding
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Barbara Fischer
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Kathrin Schrader
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Maren Brockmeyer
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Sarah Dettmer
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Christin Helbig
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | | | | | - Carole Mura
- Institut Clinique de la Souris, Illkirch, France
| | | | - Raquel Garcia-Lopez
- Experimental Embryology Lab, Instituto de Neurociencias, Universidad Miguel Hernandez, San Juan de Alicante, Spain
| | - Diego Echevarria
- Experimental Embryology Lab, Instituto de Neurociencias, Universidad Miguel Hernandez, San Juan de Alicante, Spain
| | - Eduardo Puelles
- Experimental Embryology Lab, Instituto de Neurociencias, Universidad Miguel Hernandez, San Juan de Alicante, Spain
| | - Elena Garcia-Calero
- Experimental Embryology Lab, Instituto de Neurociencias, Universidad Miguel Hernandez, San Juan de Alicante, Spain
| | | | - Markus Uhr
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Christine Kauck
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
| | - Guangjie Feng
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Nestor Milyaev
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Chuang Kee Ong
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Lalit Kumar
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - MeiSze Lam
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Colin A. Semple
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Attila Gyenesei
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Uwe Radelof
- RZPD—Deutsches Ressourcenzentrum für Genomforschung, Berlin, Germany
| | - Hans Lehrach
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Duncan R. Davidson
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| | - Pascal Dollé
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Inserm U 964, CNRS UMR 7104, Faculté de Médecine, Université de Strasbourg; Illkirch, France
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| | - Stylianos E. Antonarakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- University Hospitals of Geneva, Geneva, Switzerland
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| | - Marie-Laure Yaspo
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| | - Salvador Martinez
- Experimental Embryology Lab, Instituto de Neurociencias, Universidad Miguel Hernandez, San Juan de Alicante, Spain
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| | - Richard A. Baldock
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| | - Gregor Eichele
- Genes and Behavior Department, Max Planck Institute of Biophysical Chemistry, Goettingen, Germany
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Department of Pediatrics, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, United States of America
- * E-mail: (DRD); (PD); (SEA); (M-LY); (SM); (RAB); (GE); (AB)
| |
Collapse
|
39
|
Hertel N, Redies C. Absence of layer-specific cadherin expression profiles in the neocortex of the reeler mutant mouse. ACTA ACUST UNITED AC 2010; 21:1105-17. [PMID: 20847152 DOI: 10.1093/cercor/bhq183] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cadherins are a superfamily of Ca(2+)-dependent cell surface glycoproteins that play a morphogenetic role in a wide variety of developmental processes. They provide a code of potentially adhesive cues for layer formation in mammalian cerebral cortex. One of the animal models used for studying corticogenesis is the reeler mouse. Previous investigations showed that radial neuronal migration is impaired in this mutant, possibly resulting in an inversion of cortical layers. However, the extent of this "outside-in" cortical layering remains unclear. In the present study, we investigated the mRNA expression of cadherins (Cdh4, Cdh6, Cdh7, Cdh8, Pcdh8, Pcdh9, Pcdh11, Pcdh17, and Pcdh19) in the cerebral cortex of wild-type (wt) mice and reeler mutants. All cadherins show a layer-specific expression profile in wt mice, but, in reeler cortex, cadherin-expressing cells are distributed widely across the radial dimension. The altered layering in reeler mutants completely disrupts the radial expression of cadherins, which is more patchy, rather than laminar. Regionalized gradient-like expression of cadherins is preserved. Our findings are compatible with a model, in which the ubiquitous dispersion of cadherin-expressing cells results from a dysgenesis of radial glial cells and a misrouting of migrating neuroblasts.
Collapse
Affiliation(s)
- Nicole Hertel
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743 Jena, Germany
| | | |
Collapse
|
40
|
Alibés A, Nadra AD, De Masi F, Bulyk ML, Serrano L, Stricher F. Using protein design algorithms to understand the molecular basis of disease caused by protein-DNA interactions: the Pax6 example. Nucleic Acids Res 2010; 38:7422-31. [PMID: 20685816 PMCID: PMC2995082 DOI: 10.1093/nar/gkq683] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Quite often a single or a combination of protein mutations is linked to specific diseases. However, distinguishing from sequence information which mutations have real effects in the protein’s function is not trivial. Protein design tools are commonly used to explain mutations that affect protein stability, or protein–protein interaction, but not for mutations that could affect protein–DNA binding. Here, we used the protein design algorithm FoldX to model all known missense mutations in the paired box domain of Pax6, a highly conserved transcription factor involved in eye development and in several diseases such as aniridia. The validity of FoldX to deal with protein–DNA interactions was demonstrated by showing that high levels of accuracy can be achieved for mutations affecting these interactions. Also we showed that protein-design algorithms can accurately reproduce experimental DNA-binding logos. We conclude that 88% of the Pax6 mutations can be linked to changes in intrinsic stability (77%) and/or to its capabilities to bind DNA (30%). Our study emphasizes the importance of structure-based analysis to understand the molecular basis of diseases and shows that protein–DNA interactions can be analyzed to the same level of accuracy as protein stability, or protein–protein interactions.
Collapse
Affiliation(s)
- Andreu Alibés
- EMBL/CRG Systems Biology Research Unit, Center for Genomic Regulation, UPF, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
41
|
Davis SW, Castinetti F, Carvalho LR, Ellsworth BS, Potok MA, Lyons RH, Brinkmeier ML, Raetzman LT, Carninci P, Mortensen AH, Hayashizaki Y, Arnhold IJP, Mendonça BB, Brue T, Camper SA. Molecular mechanisms of pituitary organogenesis: In search of novel regulatory genes. Mol Cell Endocrinol 2010; 323:4-19. [PMID: 20025935 PMCID: PMC2909473 DOI: 10.1016/j.mce.2009.12.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Defects in pituitary gland organogenesis are sometimes associated with congenital anomalies that affect head development. Lesions in transcription factors and signaling pathways explain some of these developmental syndromes. Basic research studies, including the characterization of genetically engineered mice, provide a mechanistic framework for understanding how mutations create the clinical characteristics observed in patients. Defects in BMP, WNT, Notch, and FGF signaling pathways affect induction and growth of the pituitary primordium and other organ systems partly by altering the balance between signaling pathways. The PITX and LHX transcription factor families influence pituitary and head development and are clinically relevant. A few later-acting transcription factors have pituitary-specific effects, including PROP1, POU1F1 (PIT1), and TPIT (TBX19), while others, such as NeuroD1 and NR5A1 (SF1), are syndromic, influencing development of other endocrine organs. We conducted a survey of genes transcribed in developing mouse pituitary to find candidates for cases of pituitary hormone deficiency of unknown etiology. We identified numerous transcription factors that are members of gene families with roles in syndromic or non-syndromic pituitary hormone deficiency. This collection is a rich source for future basic and clinical studies.
Collapse
Affiliation(s)
- S W Davis
- University of Michigan Medical School, Ann Arbor, MI 41809-5618, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
de Boer BA, Ruijter JM, Voorbraak FPJM, Moorman AFM. More than a decade of developmental gene expression atlases: where are we now? Nucleic Acids Res 2010; 37:7349-59. [PMID: 19822576 PMCID: PMC2794177 DOI: 10.1093/nar/gkp819] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
To unravel regulatory networks of genes functioning during embryonic development, information on in situ gene expression is required. Enormous amounts of such data are available in literature, where each paper reports on a limited number of genes and developmental stages. The best way to make these data accessible is via spatio-temporal gene expression atlases. Eleven atlases, describing developing vertebrates and covering at least 100 genes, were reviewed. This review focuses on: (i) the used anatomical framework, (ii) the handling of input data and (iii) the retrieval of information. Our aim is to provide insights into both the possibilities of the atlases, as well as to describe what more than a decade of developmental gene expression atlases can teach us about the requirements of the design of the ‘ideal atlas’. This review shows that most ingredients needed to develop the ideal atlas are already applied to some extent in at least one of the discussed atlases. A review of these atlases shows that the ideal atlas should be based on a spatial framework, i.e. a series of 3D reference models, which is anatomically annotated using an ontology with sufficient resolution, both for relations as well as for anatomical terms.
Collapse
Affiliation(s)
- Bouke A de Boer
- Heart Failure Research Center, Department of Anatomy and Embryology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
43
|
Carson J, Ju T, Bello M, Thaller C, Warren J, Kakadiaris IA, Chiu W, Eichele G. Automated pipeline for atlas-based annotation of gene expression patterns: application to postnatal day 7 mouse brain. Methods 2010; 50:85-95. [PMID: 19698790 PMCID: PMC2818703 DOI: 10.1016/j.ymeth.2009.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 08/10/2009] [Accepted: 08/13/2009] [Indexed: 02/08/2023] Open
Abstract
Massive amounts of image data have been collected and continue to be generated for representing cellular gene expression throughout the mouse brain. Critical to exploiting this key effort of the post-genomic era is the ability to place these data into a common spatial reference that enables rapid interactive queries, analysis, data sharing, and visualization. In this paper, we present a set of automated protocols for generating and annotating gene expression patterns suitable for the establishment of a database. The steps include imaging tissue slices, detecting cellular gene expression levels, spatial registration with an atlas, and textual annotation. Using high-throughput in situ hybridization to generate serial sets of tissues displaying gene expression, this process was applied toward the establishment of a database representing over 200 genes in the postnatal day 7 mouse brain. These data using this protocol are now well-suited for interactive comparisons, analysis, queries, and visualization.
Collapse
Affiliation(s)
- James Carson
- Biological Monitoring and Modeling Group, Pacific Northwest National Laboratory, Richland, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Surmann-Schmitt C, Widmann N, Dietz U, Saeger B, Eitzinger N, Nakamura Y, Rattel M, Latham R, Hartmann C, von der Mark H, Schett G, von der Mark K, Stock M. Wif-1 is expressed at cartilage-mesenchyme interfaces and impedes Wnt3a-mediated inhibition of chondrogenesis. J Cell Sci 2009; 122:3627-37. [PMID: 19755491 DOI: 10.1242/jcs.048926] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Wnt factors are involved in the regulation of all steps of cartilage development. The activity of Wnt factors is generally regulated at the extracellular level by factors like the Dkk family, sFRPs, Cerberus and Wnt inhibitory factor 1 (Wif-1). Here we report that Wif-1 is highly expressed at cartilage-mesenchyme interfaces of the early developing skeleton. In fetal and postnatal skeletal development, Wif-1 is expressed in a sharply restricted zone in the upper hyaline layer of epiphyseal and articular cartilage and in trabecular bone. Coimmunoprecipitation and pull-down assays using recombinant Wif-1 and Wnt factors show specific binding of Wif-1 to Wnt3a, Wnt4, Wnt5a, Wnt7a, Wnt9a and Wnt11. Moreover, Wif-1 was able to block Wnt3a-mediated activation of the canonical Wnt signalling pathway. Consequently, Wif-1 impaired growth of mesenchymal precursor cells and neutralised Wnt3a-mediated inhibition of chondrogenesis in micromass cultures of embryonic chick limb-bud cells. These results identify Wif-1 as a novel extracellular Wnt modulator in cartilage biology.
Collapse
Affiliation(s)
- Cordula Surmann-Schmitt
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Buchanan AV, Sholtis S, Richtsmeier J, Weiss KM. What are genes "for" or where are traits "from"? What is the question? Bioessays 2009; 31:198-208. [PMID: 19204992 PMCID: PMC2807122 DOI: 10.1002/bies.200800133] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
For at least a century it has been known that multiple factors play a role in the development of complex traits, and yet the notion that there are genes "for" such traits, which traces back to Mendel, is still widespread. In this paper, we illustrate how the Mendelian model has tacitly encouraged the idea that we can explain complexity by reducing it to enumerable genes. By this approach many genes associated with simple as well as complex traits have been identified. But the genetic architecture of biological traits, or how they are made, remains largely unknown. In essence, this reflects the tension between reductionism as the current "modus operandi" of science, and the emerging knowledge of the nature of complex traits. Recent interest in systems biology as a unifying approach indicates a reawakened acceptance of the complexity of complex traits, though the temptation is to replace "gene for" thinking by comparably reductionistic "network for" concepts. Both approaches implicitly mix concepts of variants and invariants in genetics. Even the basic question is unclear: what does one need to know to "understand" the genetic basis of complex traits? New operational ideas about how to deal with biological complexity are needed.
Collapse
Affiliation(s)
- Anne V Buchanan
- Department of Anthropology, The Pennsylvania State University, University Park, PA 16802, USA.
| | | | | | | |
Collapse
|
46
|
Wolf LV, Yang Y, Wang J, Xie Q, Braunger B, Tamm ER, Zavadil J, Cvekl A. Identification of pax6-dependent gene regulatory networks in the mouse lens. PLoS One 2009; 4:e4159. [PMID: 19132093 PMCID: PMC2612750 DOI: 10.1371/journal.pone.0004159] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Accepted: 11/21/2008] [Indexed: 11/20/2022] Open
Abstract
Lineage-specific DNA-binding transcription factors regulate development by activating and repressing particular set of genes required for the acquisition of a specific cell type. Pax6 is a paired domain and homeodomain-containing transcription factor essential for development of central nervous, olfactory and visual systems, as well as endocrine pancreas. Haploinsufficiency of Pax6 results in perturbed lens development and homeostasis. Loss-of-function of Pax6 is incompatible with lens lineage formation and results in abnormal telencephalic development. Using DNA microarrays, we have identified 559 genes expressed differentially between 1-day old mouse Pax6 heterozygous and wild type lenses. Of these, 178 (31.8%) were similarly increased and decreased in Pax6 homozygous embryonic telencephalon [Holm PC, Mader MT, Haubst N, Wizenmann A, Sigvardsson M, Götz M (2007) Loss- and gain-of-function analyses reveals targets of Pax6 in the developing mouse telencephalon. Mol Cell Neurosci 34: 99–119]. In contrast, 381 (68.2%) genes were differently regulated between the lens and embryonic telencephalon. Differential expression of nine genes implicated in lens development and homeostasis: Cspg2, Igfbp5, Mab21l2, Nrf2f, Olfm3, Spag5, Spock1, Spon1 and Tgfb2, was confirmed by quantitative RT-PCR, with five of these genes: Cspg2, Mab21l2, Olfm3, Spag5 and Tgfb2, identified as candidate direct Pax6 target genes by quantitative chromatin immunoprecipitation (qChIP). In Mab21l2 and Tgfb2 promoter regions, twelve putative individual Pax6-binding sites were tested by electrophoretic mobility shift assays (EMSAs) with recombinant Pax6 proteins. This led to the identification of two and three sites in the respective Mab21l2 and Tgfb2 promoter regions identified by qChIPs. Collectively, the present studies represent an integrative genome-wide approach to identify downstream networks controlled by Pax6 that control mouse lens and forebrain development.
Collapse
Affiliation(s)
- Louise V. Wolf
- The Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Ying Yang
- The Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jinhua Wang
- NYU Cancer Institute, New York University Langone Medical Center, New York, New York, United States of America
| | - Qing Xie
- The Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Barbara Braunger
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Ernst R. Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Jiri Zavadil
- NYU Cancer Institute, New York University Langone Medical Center, New York, New York, United States of America
- Department of Pathology, New York University Langone Medical Center, New York, New York, United States of America
| | - Ales Cvekl
- The Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
47
|
Yakoby N, Bristow CA, Gong D, Schafer X, Lembong J, Zartman JJ, Halfon MS, Schüpbach T, Shvartsman SY. A combinatorial code for pattern formation in Drosophila oogenesis. Dev Cell 2008; 15:725-37. [PMID: 19000837 PMCID: PMC2822874 DOI: 10.1016/j.devcel.2008.09.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 08/27/2008] [Accepted: 09/17/2008] [Indexed: 10/21/2022]
Abstract
Two-dimensional patterning of the follicular epithelium in Drosophila oogenesis is required for the formation of three-dimensional eggshell structures. Our analysis of a large number of published gene expression patterns in the follicle cells suggests that they follow a simple combinatorial code based on six spatial building blocks and the operations of union, difference, intersection, and addition. The building blocks are related to the distribution of inductive signals, provided by the highly conserved epidermal growth factor receptor and bone morphogenetic protein signaling pathways. We demonstrate the validity of the code by testing it against a set of patterns obtained in a large-scale transcriptional profiling experiment. Using the proposed code, we distinguish 36 distinct patterns for 81 genes expressed in the follicular epithelium and characterize their joint dynamics over four stages of oogenesis. The proposed combinatorial framework allows systematic analysis of the diversity and dynamics of two-dimensional transcriptional patterns and guides future studies of gene regulation.
Collapse
Affiliation(s)
- Nir Yakoby
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemical Engineering, Princeton University, Princeton, NJ 08544, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lécuyer E, Tomancak P. Mapping the gene expression universe. Curr Opin Genet Dev 2008; 18:506-12. [PMID: 18809490 DOI: 10.1016/j.gde.2008.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Accepted: 08/21/2008] [Indexed: 11/17/2022]
Abstract
Methods to globally survey gene expression provide valuable insights into gene function during development. In particular, comprehensive in situ hybridization studies have demonstrated that gene expression patterns are extraordinarily diverse and new imaging techniques have been introduced to capture these patterns with higher resolution at the tissue, cellular, and subcellular levels. The analysis of massive image databases can be greatly facilitated by computer vision techniques once annotated image sets reach the crucial mass sufficient to train the computer in pattern recognition. Ultimately, genome-wide atlases of gene expression during development will record gene activity in living animals with at least cellular resolution and in the context of morphogenetic events. These emerging datasets will lead to great advances in the field of comparative genomics and revolutionize our ability to decipher and model developmental processes for a variety of organisms.
Collapse
Affiliation(s)
- Eric Lécuyer
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
49
|
Fulp CT, Cho G, Marsh ED, Nasrallah IM, Labosky PA, Golden JA. Identification of Arx transcriptional targets in the developing basal forebrain. Hum Mol Genet 2008; 17:3740-60. [PMID: 18799476 PMCID: PMC2581427 DOI: 10.1093/hmg/ddn271] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mutations in the aristaless-related homeobox (ARX) gene are associated with multiple neurologic disorders in humans. Studies in mice indicate Arx plays a role in neuronal progenitor proliferation and development of the cerebral cortex, thalamus, hippocampus, striatum, and olfactory bulbs. Specific defects associated with Arx loss of function include abnormal interneuron migration and subtype differentiation. How disruptions in ARX result in human disease and how loss of Arx in mice results in these phenotypes remains poorly understood. To gain insight into the biological functions of Arx, we performed a genome-wide expression screen to identify transcriptional changes within the subpallium in the absence of Arx. We have identified 84 genes whose expression was dysregulated in the absence of Arx. This population was enriched in genes involved in cell migration, axonal guidance, neurogenesis, and regulation of transcription and includes genes implicated in autism, epilepsy, and mental retardation; all features recognized in patients with ARX mutations. Additionally, we found Arx directly repressed three of the identified transcription factors: Lmo1, Ebf3 and Shox2. To further understand how the identified genes are involved in neural development, we used gene set enrichment algorithms to compare the Arx gene regulatory network (GRN) to the Dlx1/2 GRN and interneuron transcriptome. These analyses identified a subset of genes in the Arx GRN that are shared with that of the Dlx1/2 GRN and that are enriched in the interneuron transcriptome. These data indicate Arx plays multiple roles in forebrain development, both dependent and independent of Dlx1/2, and thus provides further insights into the understanding of the mechanisms underlying the pathology of mental retardation and epilepsy phenotypes resulting from ARX mutations.
Collapse
Affiliation(s)
- Carl T Fulp
- Neuroscience Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
50
|
Hoffman BG, Zavaglia B, Witzsche J, Ruiz de Algara T, Beach M, Hoodless PA, Jones SJM, Marra MA, Helgason CD. Identification of transcripts with enriched expression in the developing and adult pancreas. Genome Biol 2008; 9:R99. [PMID: 18554416 PMCID: PMC2481431 DOI: 10.1186/gb-2008-9-6-r99] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 05/13/2008] [Accepted: 06/14/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Despite recent advances, the transcriptional hierarchy driving pancreas organogenesis remains largely unknown, in part due to the paucity of comprehensive analyses. To address this deficit we generated ten SAGE libraries from the developing murine pancreas spanning Theiler stages 17-26, making use of available Pdx1 enhanced green fluorescent protein (EGFP) and Neurog3 EGFP reporter strains, as well as tissue from adult islets and ducts. RESULTS We used a specificity metric to identify 2,536 tags with pancreas-enriched expression compared to 195 other mouse SAGE libraries. We subsequently grouped co-expressed transcripts with differential expression during pancreas development using K-means clustering. We validated the clusters first using quantitative real time PCR and then by analyzing the Theiler stage 22 pancreas in situ hybridization staining patterns of over 600 of the identified genes using the GenePaint database. These were then categorized into one of the five expression domains within the developing pancreas. Based on these results we identified a cascade of transcriptional regulators expressed in the endocrine pancreas lineage and, from this, we developed a predictive regulatory network describing beta-cell development. CONCLUSION Taken together, this work provides evidence that the SAGE libraries generated here are a valuable resource for continuing to elucidate the molecular mechanisms regulating pancreas development. Furthermore, our studies provide a comprehensive analysis of pancreas development, and insights into the regulatory networks driving this process are revealed.
Collapse
Affiliation(s)
- Brad G Hoffman
- Department of Cancer Endocrinology, BC Cancer Research Center, West 10th Ave, Vancouver, BC V5Z 1L3, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|