1
|
Schmidt HF, Darwin CB, Sundaram MV. The Pax transcription factor EGL-38 links EGFR signaling to assembly of a cell type-specific apical extracellular matrix in the Caenorhabditis elegans vulva. Dev Biol 2025; 517:265-277. [PMID: 39489317 PMCID: PMC11631643 DOI: 10.1016/j.ydbio.2024.10.008] [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: 09/05/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The surface of epithelial tissues is covered by an apical extracellular matrix (aECM). The aECMs of different tissues have distinct compositions to serve distinct functions, yet how a particular cell type assembles the proper aECM is not well understood. We used the cell type-specific matrix of the C. elegans vulva to investigate the connection between cell identity and matrix assembly. The vulva is an epithelial tube composed of seven cell types descending from EGFR/Ras-dependent (1°) and Notch-dependent (2°) lineages. Vulva aECM contains multiple Zona Pellucida domain (ZP) proteins, which are a common component of aECMs across life. ZP proteins LET-653 and CUTL-18 assemble on 1° cell surfaces, while NOAH-1 assembles on a subset of 2° surfaces. All three ZP genes are broadly transcribed, indicating that cell type-specific ZP assembly must be determined by features of the destination cell surface. The paired box (Pax) transcription factor EGL-38 promotes assembly of 1° matrix and prevents inappropriate assembly of 2° matrix, suggesting that EGL-38 promotes expression of one or more ZP matrix organizers. Our results connect the known signaling pathways and various downstream effectors to EGL-38/Pax expression and the ZP matrix component of vulva cell fate execution. We propose that dedicated transcriptional networks may contribute to cell-appropriate assembly of aECM in many epithelial organs.
Collapse
Affiliation(s)
- Helen F Schmidt
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - Chelsea B Darwin
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Schmidt HF, Darwin CB, Sundaram MV. The Pax transcription factor EGL-38 links EGFR signaling to assembly of a cell-type specific apical extracellular matrix in the Caenorhabditis elegans vulva. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611291. [PMID: 39282387 PMCID: PMC11398461 DOI: 10.1101/2024.09.04.611291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The surface of epithelial tissues is covered by an apical extracellular matrix (aECM). The aECMs of different tissues have distinct compositions to serve distinct functions, yet how a particular cell type assembles the proper aECM is not well understood. We used the cell-type specific matrix of the C. elegans vulva to investigate the connection between cell identity and matrix assembly. The vulva is an epithelial tube composed of seven cell types descending from EGFR/Ras-dependent (1°) and Notch-dependent (2°) lineages. Vulva aECM contains multiple Zona Pellucida domain (ZP) proteins, which are a common component of aECMs across life. ZP proteins LET-653 and CUTL-18 assemble on 1° cell surfaces, while NOAH-1 assembles on a subset of 2° surfaces. All three ZP genes are broadly transcribed, indicating that cell-type specific ZP assembly must be determined by features of the destination cell surface. The paired box (Pax) transcription factor EGL-38 promotes assembly of 1° matrix and prevents inappropriate assembly of 2° matrix, suggesting that EGL-38 promotes expression of one or more ZP matrix organizers. Our results connect the known signaling pathways and various downstream effectors to EGL-38/Pax expression and the ZP matrix component of vulva cell fate execution. We propose that dedicated transcriptional networks may contribute to cell-appropriate assembly of aECM in many epithelial organs.
Collapse
Affiliation(s)
- Helen F Schmidt
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chelsea B Darwin
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Paczkó M, Vörös D, Szabó P, Jékely G, Szathmáry E, Szilágyi A. A neural network-based model framework for cell-fate decisions and development. Commun Biol 2024; 7:323. [PMID: 38486083 PMCID: PMC10940658 DOI: 10.1038/s42003-024-05985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 02/28/2024] [Indexed: 03/18/2024] Open
Abstract
Gene regulatory networks (GRNs) fulfill the essential function of maintaining the stability of cellular differentiation states by sustaining lineage-specific gene expression, while driving the progression of development. However, accounting for the relative stability of intermediate differentiation stages and their divergent trajectories remains a major challenge for models of developmental biology. Here, we develop an empirical data-based associative GRN model (AGRN) in which regulatory networks store multilineage stage-specific gene expression profiles as associative memory patterns. These networks are capable of responding to multiple instructive signals and, depending on signal timing and identity, can dynamically drive the differentiation of multipotent cells toward different cell state attractors. The AGRN dynamics can thus generate diverse lineage-committed cell populations in a robust yet flexible manner, providing an attractor-based explanation for signal-driven cell fate decisions during differentiation and offering a readily generalizable modelling tool that can be applied to a wide variety of cell specification systems.
Collapse
Affiliation(s)
- Mátyás Paczkó
- Institute of Evolution, HUN-REN Centre for Ecological Research, Konkoly-Thege M. út 29-33, 1121, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117, Budapest, Hungary
| | - Dániel Vörös
- Institute of Evolution, HUN-REN Centre for Ecological Research, Konkoly-Thege M. út 29-33, 1121, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117, Budapest, Hungary
| | - Péter Szabó
- Institute of Evolution, HUN-REN Centre for Ecological Research, Konkoly-Thege M. út 29-33, 1121, Budapest, Hungary
| | - Gáspár Jékely
- Living Systems Institute, University of Exeter, Stocker Road 4QD, EX4, Exeter, UK
| | - Eörs Szathmáry
- Institute of Evolution, HUN-REN Centre for Ecological Research, Konkoly-Thege M. út 29-33, 1121, Budapest, Hungary.
- Center for the Conceptual Foundations of Science, Parmenides Foundation, Hindenburgstr. 15, 82343, Pöcking, Germany.
- Department of Plant Systematics, Ecology and Theoretical Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117, Budapest, Hungary.
| | - András Szilágyi
- Institute of Evolution, HUN-REN Centre for Ecological Research, Konkoly-Thege M. út 29-33, 1121, Budapest, Hungary
| |
Collapse
|
4
|
Liu J, Murray JI. Mechanisms of lineage specification in Caenorhabditis elegans. Genetics 2023; 225:iyad174. [PMID: 37847877 PMCID: PMC11491538 DOI: 10.1093/genetics/iyad174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
The studies of cell fate and lineage specification are fundamental to our understanding of the development of multicellular organisms. Caenorhabditis elegans has been one of the premiere systems for studying cell fate specification mechanisms at single cell resolution, due to its transparent nature, the invariant cell lineage, and fixed number of somatic cells. We discuss the general themes and regulatory mechanisms that have emerged from these studies, with a focus on somatic lineages and cell fates. We next review the key factors and pathways that regulate the specification of discrete cells and lineages during embryogenesis and postembryonic development; we focus on transcription factors and include numerous lineage diagrams that depict the expression of key factors that specify embryonic founder cells and postembryonic blast cells, and the diverse somatic cell fates they generate. We end by discussing some future perspectives in cell and lineage specification.
Collapse
Affiliation(s)
- Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - John Isaac Murray
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Okubo K, Kaneko K. Heterosis of fitness and phenotypic variance in the evolution of a diploid gene regulatory network. PNAS NEXUS 2022; 1:pgac097. [PMID: 36741431 PMCID: PMC9896930 DOI: 10.1093/pnasnexus/pgac097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/24/2022] [Indexed: 02/07/2023]
Abstract
Heterosis describes the phenomenon, whereby a hybrid population has higher fitness than an inbred population, which has previously been explained by either Mendelian dominance or overdominance under the general assumption of a simple genotype-phenotype relationship. However, recent studies have demonstrated that genes interact through a complex gene regulatory network (GRN). Furthermore, phenotypic variance is reportedly lower for heterozygotes, and the origin of such variance-related heterosis remains elusive. Therefore, a theoretical analysis linking heterosis to GRN evolution and stochastic gene expression dynamics is required. Here, we investigated heterosis related to fitness and phenotypic variance in a system with interacting genes by numerically evolving diploid GRNs. According to the results, the heterozygote population exhibited higher fitness than the homozygote population, indicating fitness-related heterosis resulting from evolution. In addition, the heterozygote population exhibited lower noise-related phenotypic variance in expression levels than the homozygous population, implying that the heterozygote population is more robust to noise. Furthermore, the distribution of the ratio of heterozygote phenotypic variance to homozygote phenotypic variance exhibited quantitative similarity with previous experimental results. By applying dominance and differential gene expression rather than only a single gene expression model, we confirmed the correlation between heterosis and differential gene expression. We explain our results by proposing that the convex high-fitness region is evolutionarily shaped in the genetic space to gain noise robustness under genetic mixing through sexual reproduction. These results provide new insights into the effects of GRNs on variance-related heterosis and differential gene expression.
Collapse
Affiliation(s)
- Kenji Okubo
- Research Center for Integrative Evolutionary Science, the Graduate University for Advanced Studies, SOKENDAI, Hayama, Kanagawa, 240-0193, Japan
| | | |
Collapse
|
6
|
Zhang Q, Hrach H, Mangone M, Reiner DJ. Identifying the Caenorhabditis elegans vulval transcriptome. G3 (BETHESDA, MD.) 2022; 12:jkac091. [PMID: 35551383 PMCID: PMC9157107 DOI: 10.1093/g3journal/jkac091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022]
Abstract
Development of the Caenorhabditis elegans vulva is a classic model of organogenesis. This system, which starts with 6 equipotent cells, encompasses diverse types of developmental event, including developmental competence, multiple signaling events to control precise and faithful patterning of three cell fates, execution and proliferation of specific cell lineages, and a series of sophisticated morphogenetic events. Early events have been subjected to extensive mutational and genetic investigations and later events to cell biological analyses. We infer the existence of dramatically changing profiles of gene expression that accompanies the observed changes in development. Yet, except from serendipitous discovery of several transcription factors expressed in dynamic patterns in vulval lineages, our knowledge of the transcriptomic landscape during vulval development is minimal. This study describes the composition of a vulva-specific transcriptome. We used tissue-specific harvesting of mRNAs via immunoprecipitation of epitope-tagged poly(A) binding protein, PAB-1, heterologously expressed by a promoter known to express GFP in vulval cells throughout their development. The identified transcriptome was small but tightly interconnected. From this data set, we identified several genes with identified functions in development of the vulva and validated more with promoter-GFP reporters of expression. For one target, lag-1, promoter-GFP expression was limited but a fluorescent tag of the endogenous protein revealed extensive expression. Thus, we have identified a transcriptome of C. elegans vulval lineages as a launching pad for exploration of functions of these genes in organogenesis.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| | - Heather Hrach
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85281, USA
- Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, Tempe, AZ 85281, USA
| | - Marco Mangone
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85281, USA
- Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, Tempe, AZ 85281, USA
| | - David J Reiner
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| |
Collapse
|
7
|
Moradifard S, Hoseinbeyki M, Emam MM, Parchiniparchin F, Ebrahimi-Rad M. Association of the Sp1 binding site and -1997 promoter variations in COL1A1 with osteoporosis risk: The application of meta-analysis and bioinformatics approaches offers a new perspective for future research. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2020; 786:108339. [PMID: 33339581 DOI: 10.1016/j.mrrev.2020.108339] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 08/11/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022]
Abstract
As a complex disease, osteoporosis is influenced by several genetic markers. Many studies have examined the link between the Sp1 binding site +1245 G > T (rs1800012) and -1997 G > T (rs1107946) variations in the COL1A1 gene with osteoporosis risk. However, the findings of these studies have been contradictory; therefore, we performed a meta-analysis to aggregate additional information and obtain increased statistical power to more efficiently estimate this correlation. A meta-analysis was conducted with studies published between 1991-2020 that were identified by a systematic electronic search of the Scopus and Clarivate Analytics databases. Studies with bone mineral density (BMD) data and complete genotypes of the single-nucleotide variations (SNVs) for the overall and postmenopausal female population were included in this meta-analysis and analyzed using the R metaphor package. A relationship between rs1800012 and significantly decreased BMD values at the lumbar spine and femoral neck was found in individuals carrying the "ss" versus the "SS" genotype in the overall population according to a random effects model (p < 0.0001). Similar results were also found in the postmenopausal female population (p = 0.003 and 0.0002, respectively). Such findings might be an indication of increased osteoporosis risk in both studied groups in individuals with the "ss" genotype. Although no association was identified between the -1997 G > T and low BMD in the overall population, those individuals with the "GT" genotype showed a higher level of BMD than those with "GG" in the subgroup analysis (p = 0.007). To determine which transcription factor (TF) might bind to the -1997 G > T in COL1A1, 45 TFs were identified based on bioinformatics predictions. According to the GSE35958 microarray dataset, 16 of 45 TFs showed differential expression profiles in osteoporotic human mesenchymal stem cells relative to normal samples from elderly donors. By identifying candidate TFs for the -1997 G > T site, our study offers a new perspective for future research.
Collapse
Affiliation(s)
| | | | - Mohammad Mehdi Emam
- Rheumatology Ward, Loghman Hospital, Shahid Beheshti Medical University (SBMU), Tehran, Iran
| | | | | |
Collapse
|
8
|
Cohen JD, Sparacio AP, Belfi AC, Forman-Rubinsky R, Hall DH, Maul-Newby H, Frand AR, Sundaram MV. A multi-layered and dynamic apical extracellular matrix shapes the vulva lumen in Caenorhabditis elegans. eLife 2020; 9:e57874. [PMID: 32975517 PMCID: PMC7544507 DOI: 10.7554/elife.57874] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Biological tubes must develop and maintain their proper diameter to transport materials efficiently. These tubes are molded and protected in part by apical extracellular matrices (aECMs) that line their lumens. Despite their importance, aECMs are difficult to image in vivo and therefore poorly understood. The Caenorhabditis elegans vulva has been a paradigm for understanding many aspects of organogenesis. Here we describe the vulva luminal matrix, which contains chondroitin proteoglycans, Zona Pellucida (ZP) domain proteins, and other glycoproteins and lipid transporters related to those in mammals. Confocal and transmission electron microscopy revealed, with unprecedented detail, a complex and dynamic aECM. Different matrix factors assemble on the apical surfaces of each vulva cell type, with clear distinctions seen between Ras-dependent (1°) and Notch-dependent (2°) cell types. Genetic perturbations suggest that chondroitin and other aECM factors together generate a structured scaffold that both expands and constricts lumen shape.
Collapse
Affiliation(s)
- Jennifer D Cohen
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Alessandro P Sparacio
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Alexandra C Belfi
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Rachel Forman-Rubinsky
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Hannah Maul-Newby
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Alison R Frand
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| |
Collapse
|
9
|
Abete-Luzi P, Fukushige T, Yun S, Krause MW, Eisenmann DM. New Roles for the Heterochronic Transcription Factor LIN-29 in Cuticle Maintenance and Lipid Metabolism at the Larval-to-Adult Transition in Caenorhabditis elegans. Genetics 2020; 214:669-690. [PMID: 31974205 PMCID: PMC7054012 DOI: 10.1534/genetics.119.302860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/21/2020] [Indexed: 11/18/2022] Open
Abstract
Temporal regulation of gene expression is a crucial aspect of metazoan development. In the roundworm Caenorhabditis elegans, the heterochronic pathway controls multiple developmental events in a time-specific manner. The most downstream effector of this pathway, the zinc-finger transcription factor LIN-29, acts in the last larval stage (L4) to regulate elements of the larval-to-adult switch. Here, we explore new LIN-29 targets and their implications for this developmental transition. We used RNA-sequencing to identify genes differentially expressed between animals misexpressing LIN-29 at an early time point and control animals. Among 230 LIN-29-activated genes, we found that genes encoding cuticle collagens were overrepresented. Interestingly, expression of lin-29 and some of these collagens was increased in adults with cuticle damage, suggesting a previously unknown function for LIN-29 in adult cuticle maintenance. On the other hand, genes involved in fat metabolism were enriched among 350 LIN-29-downregulated targets. We used mass spectrometry to assay lipid content in animals overexpressing LIN-29 and observed reduced fatty acid levels. Many LIN-29-repressed genes are normally expressed in the intestine, suggesting cell-nonautonomous regulation. We identified several LIN-29 upregulated genes encoding signaling molecules that may act as mediators in the regulation of intestinally expressed genes encoding fat metabolic enzymes and vitellogenins. Overall, our results support the model of LIN-29 as a major regulator of adult cuticle synthesis and integrity, and as the trigger for metabolic changes that take place at the important transition from rapid growth during larval life to slower growth and offspring production during adulthood.
Collapse
Affiliation(s)
- Patricia Abete-Luzi
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | - Tetsunari Fukushige
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Sijung Yun
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Michael W Krause
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - David M Eisenmann
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| |
Collapse
|
10
|
Fernandez-Valverde SL, Aguilera F, Ramos-Díaz RA. Inference of Developmental Gene Regulatory Networks Beyond Classical Model Systems: New Approaches in the Post-genomic Era. Integr Comp Biol 2019; 58:640-653. [PMID: 29917089 DOI: 10.1093/icb/icy061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The advent of high-throughput sequencing (HTS) technologies has revolutionized the way we understand the transformation of genetic information into morphological traits. Elucidating the network of interactions between genes that govern cell differentiation through development is one of the core challenges in genome research. These networks are known as developmental gene regulatory networks (dGRNs) and consist largely of the functional linkage between developmental control genes, cis-regulatory modules, and differentiation genes, which generate spatially and temporally refined patterns of gene expression. Over the last 20 years, great advances have been made in determining these gene interactions mainly in classical model systems, including human, mouse, sea urchin, fruit fly, and worm. This has brought about a radical transformation in the fields of developmental biology and evolutionary biology, allowing the generation of high-resolution gene regulatory maps to analyze cell differentiation during animal development. Such maps have enabled the identification of gene regulatory circuits and have led to the development of network inference methods that can recapitulate the differentiation of specific cell-types or developmental stages. In contrast, dGRN research in non-classical model systems has been limited to the identification of developmental control genes via the candidate gene approach and the characterization of their spatiotemporal expression patterns, as well as to the discovery of cis-regulatory modules via patterns of sequence conservation and/or predicted transcription-factor binding sites. However, thanks to the continuous advances in HTS technologies, this scenario is rapidly changing. Here, we give a historical overview on the architecture and elucidation of the dGRNs. Subsequently, we summarize the approaches available to unravel these regulatory networks, highlighting the vast range of possibilities of integrating multiple technical advances and theoretical approaches to expand our understanding on the global gene regulation during animal development in non-classical model systems. Such new knowledge will not only lead to greater insights into the evolution of molecular mechanisms underlying cell identity and animal body plans, but also into the evolution of morphological key innovations in animals.
Collapse
Affiliation(s)
- Selene L Fernandez-Valverde
- CONACYT, Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Centro de Investigación y de Estudios Avanzados del IPN, Irapuato, Guanajuato, Mexico
| | - Felipe Aguilera
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile
| | - René Alexander Ramos-Díaz
- CONACYT, Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Centro de Investigación y de Estudios Avanzados del IPN, Irapuato, Guanajuato, Mexico
| |
Collapse
|
11
|
Shin H, Reiner DJ. The Signaling Network Controlling C. elegans Vulval Cell Fate Patterning. J Dev Biol 2018; 6:E30. [PMID: 30544993 PMCID: PMC6316802 DOI: 10.3390/jdb6040030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/17/2022] Open
Abstract
EGF, emitted by the Anchor Cell, patterns six equipotent C. elegans vulval precursor cells to assume a precise array of three cell fates with high fidelity. A group of core and modulatory signaling cascades forms a signaling network that demonstrates plasticity during the transition from naïve to terminally differentiated cells. In this review, we summarize the history of classical developmental manipulations and molecular genetics experiments that led to our understanding of the signals governing this process, and discuss principles of signal transduction and developmental biology that have emerged from these studies.
Collapse
Affiliation(s)
- Hanna Shin
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
| | - David J Reiner
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
- College of Medicine, Texas A & M University, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Mok DZL, Sternberg PW, Inoue T. Morphologically defined sub-stages of C. elegans vulval development in the fourth larval stage. BMC DEVELOPMENTAL BIOLOGY 2015; 15:26. [PMID: 26066484 PMCID: PMC4464634 DOI: 10.1186/s12861-015-0076-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/03/2015] [Indexed: 01/06/2023]
Abstract
Background During the fourth larval (L4) stage, vulval cells of C. elegans undergo extensive morphogenesis accompanied by changes in gene expression. This phase of vulval development, occurring after the well-studied induction of vulval cells, is not well understood but is potentially a useful context in which to study how a complex temporal sequence of events is regulated during development. However, a system for precisely describing different phases of vulval development in the L4 stage has been lacking. Results We defined ten sub-stages of L4 based on morphological criteria as observed using Nomarski microscopy (L4.0 ~ L4.9). Precise timing of each sub-stage at 20 °C was determined. We also re-examined the timing of expression for several gene expression markers, and correlated the sub-stages with the timing of other developmental events in the vulva and the uterus. Conclusions This scheme allows the developmental timing of an L4 individual to be determined at approximately one-hour resolution without the need to resort to time course experiments. These well-defined developmental stages will enable more precise description of gene expression and other developmental events.
Collapse
Affiliation(s)
- Darren Z L Mok
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore
| | - Paul W Sternberg
- HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Takao Inoue
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore. .,HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
13
|
Ecsedi M, Rausch M, Großhans H. The let-7 microRNA directs vulval development through a single target. Dev Cell 2015; 32:335-44. [PMID: 25669883 DOI: 10.1016/j.devcel.2014.12.018] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/31/2014] [Accepted: 12/18/2014] [Indexed: 11/16/2022]
Abstract
The let-7 microRNA (miRNA) regulates stemness in animals ranging from worms to humans. However, the cause of the dramatic vulval rupturing phenotype of let-7 mutant C. elegans has remained unknown. Consistent with the notion that miRNAs function by coordinately tuning the expression of many targets, bursting may result from joint dysregulation of several targets, possibly in the epidermis. Alternatively, overexpression of LET-60/RAS, a key vulva development gene and a phylogenetically conserved target of let-7, may be responsible. Here, we show that let-7 functions in the vulval-uterine system to ensure vulval integrity but that regulation of most targets of let-7, including LET-60/RAS, is dispensable. Using CRISPR-Cas9 to edit endogenous let-7 target sites, we found that regulation of LIN-41/TRIM71 alone is necessary and sufficient to prevent vulval rupturing. Hence, let-7 does not function to reduce gene expression noise broadly, but to direct vulval development through extensive regulation of a single, defined target.
Collapse
Affiliation(s)
- Matyas Ecsedi
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; University of Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Magdalene Rausch
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; University of Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Helge Großhans
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland.
| |
Collapse
|
14
|
Schindler AJ, Sherwood DR. Morphogenesis of the caenorhabditis elegans vulva. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 2:75-95. [PMID: 23418408 DOI: 10.1002/wdev.87] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Understanding how cells move, change shape, and alter cellular behaviors to form organs, a process termed morphogenesis, is one of the great challenges of developmental biology. Formation of the Caenorhabditis elegans vulva is a powerful, simple, and experimentally accessible model for elucidating how morphogenetic processes produce an organ. In the first step of vulval development, three epithelial precursor cells divide and differentiate to generate 22 cells of 7 different vulval subtypes. The 22 vulval cells then rearrange from a linear array into a tube, with each of the seven cell types undergoing characteristic morphogenetic behaviors that construct the vulva. Vulval morphogenesis entails many of the same cellular activities that underlie organogenesis and tissue formation across species, including invagination, lumen formation, oriented cell divisions, cell–cell adhesion, cell migration, cell fusion, extracellular matrix remodeling, and cell invasion. Studies of vulval development have led to pioneering discoveries in a number of these processes and are beginning to bridge the gap between the pathways that specify cells and their connections to morphogenetic behaviors. The simplicity of the vulva and the experimental tools available in C. elegans will continue to make vulval morphogenesis a powerful paradigm to further our understanding of the largely mysterious mechanisms that build tissues and organs.
Collapse
|
15
|
Sethi AJ, Angerer RC, Angerer LM. Multicolor labeling in developmental gene regulatory network analysis. Methods Mol Biol 2014; 1128:249-62. [PMID: 24567220 DOI: 10.1007/978-1-62703-974-1_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
The sea urchin embryo is an important model system for developmental gene regulatory network (GRN) analysis. This chapter describes the use of multicolor fluorescent in situ hybridization (FISH) as well as a combination of FISH and immunohistochemistry in sea urchin embryonic GRN studies. The methods presented here can be applied to a variety of experimental settings where accurate spatial resolution of multiple gene products is required for constructing a developmental GRN.
Collapse
Affiliation(s)
- Aditya J Sethi
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
16
|
Vandenbon A, Kumagai Y, Akira S, Standley DM. A novel unbiased measure for motif co-occurrence predicts combinatorial regulation of transcription. BMC Genomics 2012; 13 Suppl 7:S11. [PMID: 23282148 PMCID: PMC3521209 DOI: 10.1186/1471-2164-13-s7-s11] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Multiple transcription factors (TFs) are involved in the generation of gene expression patterns, such as tissue-specific gene expression and pleiotropic immune responses. However, how combinations of TFs orchestrate diverse gene expression patterns is poorly understood. Here we propose a new measure for regulatory motif co-occurrence and a new methodology to systematically identify TF pairs significantly co-occurring in a set of promoter sequences. Results Initial analyses suggest that non-CpG promoters have a higher potential for combinatorial regulation than CpG island-associated promoters, and that co-occurrences are strongly influenced by motif similarity. We applied our method to large-scale gene expression data from various tissues, and showed how our measure for motif co-occurrence is not biased by motif over-representation. Our method identified, amongst others, the binding motifs of HNF1 and FOXP1 to be significantly co-occurring in promoters of liver/kidney specific genes. Binding sites tend to be positioned proximally to each other, suggesting interactions exist between this pair of transcription factors. Moreover, the binding sites of several TFs were found to co-occur with NF-κB and IRF sites in sets of genes with similar expression patterns in dendritic cells after Toll-like receptor stimulation. Of these, we experimentally verified that CCAAT enhancer binding protein alpha positively regulates its target promoters synergistically with NF-κB. Conclusions Both computational and experimental results indicate that the proposed method can clarify TF interactions that could not be observed by currently available prediction methods.
Collapse
Affiliation(s)
- Alexis Vandenbon
- Laboratory of Systems Immunology, Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | | | | | | |
Collapse
|
17
|
Meelkop E, Temmerman L, Janssen T, Suetens N, Beets I, Van Rompay L, Shanmugam N, Husson SJ, Schoofs L. PDF receptor signaling in Caenorhabditis elegans modulates locomotion and egg-laying. Mol Cell Endocrinol 2012; 361:232-40. [PMID: 22579613 DOI: 10.1016/j.mce.2012.05.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 03/26/2012] [Accepted: 05/02/2012] [Indexed: 01/22/2023]
Abstract
In Caenorhabditis elegans, pdfr-1 encodes three receptors of the secretin receptor family. These G protein-coupled receptors are activated by three neuropeptides, pigment dispersing factors 1a, 1b and 2, which are encoded by pdf-1 and pdf-2. We isolated a PDF receptor loss-of-function allele (lst34) by means of a mutagenesis screen and show that the PDF signaling system is involved in locomotion and egg-laying. We demonstrate that the pdfr-1 mutant phenocopies the defective locomotor behavior of the pdf-1 mutant and that pdf-1 and pdf-2 behave antagonistically. All three PDF receptor splice variants are involved in the regulation of locomotor behavior. Cell specific rescue experiments show that this pdf mediated behavior is regulated by neurons rather than body wall muscles. We also show that egg-laying patterns of pdf-1 and pdf-2 mutants are affected, but not those of pdfr-1 mutants, pointing to a novel role for the PDF-system in the regulation of egg-laying.
Collapse
Affiliation(s)
- Ellen Meelkop
- Research Group of Functional Genomics and Proteomics, Department of Biology, KU Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Bae YK, Sung JY, Kim YN, Kim S, Hong KM, Kim HT, Choi MS, Kwon JY, Shim J. An in vivo C. elegans model system for screening EGFR-inhibiting anti-cancer drugs. PLoS One 2012; 7:e42441. [PMID: 22957020 PMCID: PMC3434183 DOI: 10.1371/journal.pone.0042441] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 07/09/2012] [Indexed: 11/20/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a well-established target for cancer treatment. EGFR tyrosine kinase (TK) inhibitors, such as gefinitib and erlotinib, have been developed as anti-cancer drugs. Although non-small cell lung carcinoma with an activating EGFR mutation, L858R, responds well to gefinitib and erlotinib, tumors with a doubly mutated EGFR, T790M-L858R, acquire resistance to these drugs. The C. elegans EGFR homolog LET-23 and its downstream signaling pathway have been studied extensively to provide insight into regulatory mechanisms conserved from C. elegans to humans. To develop an in vivo screening system for potential cancer drugs targeting specific EGFR mutants, we expressed three LET-23 chimeras in which the TK domain was replaced with either the human wild-type TK domain (LET-23::hEGFR-TK), a TK domain with the L858R mutation (LET-23::hEGFR-TK[L858R]), or a TK domain with the T790M-L858R mutations (LET-23::hEGFR-TK[T790M-L858R]) in C. elegans vulval cells using the let-23 promoter. The wild-type hEGFR-TK chimeric protein rescued the let-23 mutant phenotype, and the activating mutant hEGFR-TK chimeras induced a multivulva (Muv) phenotype in a wild-type C. elegans background. The anti-cancer drugs gefitinib and erlotinib suppressed the Muv phenotype in LET-23::hEGFR-TK[L858R]-expressing transgenic animals, but not in LET-23::hEGFR-TK[T790M-L858R] transgenic animals. As a pilot screen, 8,960 small chemicals were tested for Muv suppression, and AG1478 (an EGFR-TK inhibitor) and U0126 (a MEK inhibitor) were identified as potential inhibitors of EGFR-mediated biological function. In conclusion, transgenic C. elegans expressing chimeric LET-23::hEGFR-TK proteins are a model system that can be used in mutation-specific screens for new anti-cancer drugs.
Collapse
Affiliation(s)
- Young-Ki Bae
- Comparative Biomedicine Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Jee Young Sung
- Pediatric Oncology Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Yong-Nyun Kim
- Comparative Biomedicine Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Sunshin Kim
- New Experimental Therapeutics Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Kyeong Man Hong
- Cancer Cell and Molecular Biology Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Heung Tae Kim
- Center for Lung Cancer, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Min Sung Choi
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Jae Young Kwon
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Jaegal Shim
- Comparative Biomedicine Research Branch, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
- * E-mail:
| |
Collapse
|
19
|
Kim J, Choi M, Kim JR, Jin H, Kim VN, Cho KH. The co-regulation mechanism of transcription factors in the human gene regulatory network. Nucleic Acids Res 2012; 40:8849-61. [PMID: 22798495 PMCID: PMC3467061 DOI: 10.1093/nar/gks664] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The co-regulation of transcription factors (TFs) has been widely observed in various species. Why is such a co-regulation mechanism needed for transcriptional regulation? To answer this question, the following experiments and analyses were performed. First, examination of the human gene regulatory network (GRN) indicated that co-regulation was significantly enriched in the human GRN. Second, mathematical simulation of an artificial regulatory network showed that the co-regulation mechanism was related to the biphasic dose-response patterns of TFs. Third, the relationship between the co-regulation mechanism and the biphasic dose-response pattern was confirmed using microarray experiments examining different time points and different doses of the toxicant tetrachlorodibenzodioxin. Finally, two mathematical models were constructed to mimic highly co-regulated networks (HCNs) and little co-regulated networks (LCNs), and we found that HCNs were more robust to parameter perturbation than LCNs, whereas LCNs were faster in adaptation to environmental changes than HCNs.
Collapse
Affiliation(s)
- Junil Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
20
|
Linhart C, Halperin Y, Darom A, Kidron S, Broday L, Shamir R. A novel candidate cis-regulatory motif pair in the promoters of germline and oogenesis genes in C. elegans. Genome Res 2011; 22:76-83. [PMID: 21930893 DOI: 10.1101/gr.115626.110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In this study we report on a novel pair of cis-regulatory motifs in promoter sequences of the nematode Caenorhabditis elegans. The motif pair exhibits extraordinary genomic traits: The order and the orientation of the two motifs are highly specific, and the distance between them is almost always one of two frequent distances. In contrast, the sequence between the motifs is variable across occurrences. Thus, the motif pair constitutes a nearly combinatorial sequence configuration. We further show that this module is conserved among, and unique to, the entire Caenorhabditis genus. By analyzing several gene expression data sets, our data suggest that this motif pair may function in germline development, oogenesis, and early embryogenesis. Finally, we verify that the motifs are indeed functional cis-regulatory elements using reporter constructs in transgenic C. elegans.
Collapse
Affiliation(s)
- Chaim Linhart
- School of Computer Science, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | | | |
Collapse
|
21
|
Ulm EA, Sleiman SF, Chamberlin HM. Developmental functions for the Caenorhabditis elegans Sp protein SPTF-3. Mech Dev 2011; 128:428-41. [PMID: 21884786 DOI: 10.1016/j.mod.2011.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/12/2011] [Accepted: 08/14/2011] [Indexed: 10/17/2022]
Abstract
Sp factors are important for animal development and the transcriptional regulation of a wide variety of genes. How they influence the developmental decisions of individual cells within the organism, however, is poorly understood. To better understand the developmental functions for Sp transcription factors, we have characterized the functions of Caenorhabditis elegans SPTF-3 using RNAi knockdown and a non-null, hypomorphic mutant allele. We find that disruption of sptf-3 confers a variety of developmental defects, including defects in development of the egg-laying system, oocyte production, and embryonic morphogenesis. sptf-3 mutants exhibit defects in vulval lineage polarity, a phenotype previously only observed in mutants defective in Wnt signaling. We show that the embryonic function of sptf-3 is dependent on germline activity, arguing that the gene has an important maternal contribution to embryonic development. An evaluation of reporter gene expression suggests that SPTF-3 exhibits specificity, in that it can influence the expression of a given gene in some cells but not others, and that SPTF-3 participates in the maintenance of gene expression states in differentiated cells. We propose SPTF-3 provides a good model to study the in vivo functions for Sp transcription factors during animal development.
Collapse
Affiliation(s)
- Elizabeth A Ulm
- Department of Molecular Genetics, Ohio State University, 484 W.12th Ave., Columbus, OH 43210, USA.
| | | | | |
Collapse
|
22
|
Schindler AJ, Sherwood DR. The transcription factor HLH-2/E/Daughterless regulates anchor cell invasion across basement membrane in C. elegans. Dev Biol 2011; 357:380-91. [PMID: 21784067 DOI: 10.1016/j.ydbio.2011.07.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 06/17/2011] [Accepted: 07/07/2011] [Indexed: 10/18/2022]
Abstract
Cell invasion through basement membrane is a specialized cellular behavior critical for many developmental processes and leukocyte trafficking. Invasive cellular behavior is also inappropriately co-opted during cancer progression. Acquisition of an invasive phenotype is accompanied by changes in gene expression that are thought to coordinate the steps of invasion. The transcription factors responsible for these changes in gene expression, however, are largely unknown. C. elegans anchor cell (AC) invasion is a genetically tractable in vivo model of invasion through basement membrane. AC invasion requires the conserved transcription factor FOS-1A, but other transcription factors are thought to act in parallel to FOS-1A to control invasion. Here we identify the transcription factor HLH-2, the C. elegans ortholog of Drosophila Daughterless and vertebrate E proteins, as a regulator of AC invasion. Reduction of HLH-2 function by RNAi or with a hypomorphic allele causes defects in AC invasion. Genetic analysis indicates that HLH-2 has functions outside of the FOS-1A pathway. Using expression analysis, we identify three genes that are transcriptionally regulated by HLH-2: the protocadherin cdh-3, and two genes encoding secreted extracellular matrix proteins, mig-6/papilin and him-4/hemicentin. Further, we show that reduction of HLH-2 function causes defects in polarization of F-actin to the invasive cell membrane, a process required for the AC to generate protrusions that breach the basement membrane. This work identifies HLH-2 as a regulator of the invasive phenotype in the AC, adding to our understanding of the transcriptional networks that control cell invasion.
Collapse
|
23
|
Zhang S, Jin W, Huang Y, Su W, Yang J, Feng Z. Profiling a Caenorhabditis elegans behavioral parametric dataset with a supervised K-means clustering algorithm identifies genetic networks regulating locomotion. J Neurosci Methods 2011; 197:315-23. [PMID: 21376755 PMCID: PMC3084513 DOI: 10.1016/j.jneumeth.2011.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 02/11/2011] [Accepted: 02/17/2011] [Indexed: 12/11/2022]
Abstract
Defining genetic networks underlying animal behavior in a high throughput manner is an important but challenging task that has not yet been achieved for any organism. Using Caenorhabditis elegans, we collected quantitative parametric data related to various aspects of locomotion from wild type and 31 mutant worm strains with single mutations in genes functioning in sensory reception, neurotransmission, G-protein signaling, neuromuscular control or other facets of motor regulation. We applied unsupervised and constrained K-means clustering algorithms to the data and found that the genes that clustered together due to the behavioral similarity of their mutants encoded proteins in the same signaling networks. This approach provides a framework to identify genes and genetic networks underlying worm neuromotor function in a high-throughput manner. A publicly accessible database harboring the visual and quantitative behavioral data collected in this study adds valuable information to the rapidly growing C. elegans databanks that can be employed in a similar context.
Collapse
Affiliation(s)
- Shijie Zhang
- Department of Pharmacology, School of Medicine Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
- Department of Electrical Engineering and Computer Science, School of Engineering Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
| | - Wei Jin
- Department of Electrical Engineering and Computer Science, School of Engineering Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
| | - Ying Huang
- Department of Pharmacology, School of Medicine Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
| | - Wei Su
- Department of Electrical Engineering and Computer Science, School of Engineering Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
| | - Jiong Yang
- Department of Electrical Engineering and Computer Science, School of Engineering Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
| | - Zhaoyang Feng
- Department of Pharmacology, School of Medicine Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
- Department of Electrical Engineering and Computer Science, School of Engineering Case Western Reserve University 10900 Euclid Avenue Cleveland, OH 44106
- Department of Physiology, School of Medicine, Xi’an Jiaotong University, 76 Yanta West Road, Xi’an, Shaanxi, China 710061
| |
Collapse
|
24
|
Smith CJ, Watson JD, Spencer WC, O’Brien T, Cha B, Albeg A, Treinin M, Miller DM. Time-lapse imaging and cell-specific expression profiling reveal dynamic branching and molecular determinants of a multi-dendritic nociceptor in C. elegans. Dev Biol 2010; 345:18-33. [PMID: 20537990 PMCID: PMC2919608 DOI: 10.1016/j.ydbio.2010.05.502] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 05/19/2010] [Accepted: 05/19/2010] [Indexed: 10/19/2022]
Abstract
Nociceptive neurons innervate the skin with complex dendritic arbors that respond to pain-evoking stimuli such as harsh mechanical force or extreme temperatures. Here we describe the structure and development of a model nociceptor, the PVD neuron of C. elegans, and identify transcription factors that control morphogenesis of the PVD dendritic arbor. The two PVD neuron cell bodies occupy positions on either the right (PVDR) or left (PVDL) sides of the animal in posterior-lateral locations. Imaging with a GFP reporter revealed a single axon projecting from the PVD soma to the ventral cord and an elaborate, highly branched arbor of dendritic processes that envelop the animal with a web-like array directly beneath the skin. Dendritic branches emerge in a step-wise fashion during larval development and may use an existing network of peripheral nerve cords as guideposts for key branching decisions. Time-lapse imaging revealed that branching is highly dynamic with active extension and withdrawal and that PVD branch overlap is prevented by a contact-dependent self-avoidance, a mechanism that is also employed by sensory neurons in other organisms. With the goal of identifying genes that regulate dendritic morphogenesis, we used the mRNA-tagging method to produce a gene expression profile of PVD during late larval development. This microarray experiment identified>2,000 genes that are 1.5X elevated relative to all larval cells. The enriched transcripts encode a wide range of proteins with potential roles in PVD function (e.g., DEG/ENaC and Trp channels) or development (e.g., UNC-5 and LIN-17/frizzled receptors). We used RNAi and genetic tests to screen 86 transcription factors from this list and identified eleven genes that specify PVD dendritic structure. These transcription factors appear to control discrete steps in PVD morphogenesis and may either promote or limit PVD branching at specific developmental stages. For example, time-lapse imaging revealed that MEC-3 (LIM homeodomain) is required for branch initiation in early larval development whereas EGL-44 (TEAD domain) prevents ectopic PVD branching in the adult. A comparison of PVD-enriched transcripts to a microarray profile of mammalian nociceptors revealed homologous genes with potentially shared nociceptive functions. We conclude that PVD neurons display striking structural, functional and molecular similarities to nociceptive neurons from more complex organisms and can thus provide a useful model system in which to identify evolutionarily conserved determinants of nociceptor fate.
Collapse
Affiliation(s)
- Cody J. Smith
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232-8240 Phone: (615) 343-3447
| | - Joseph D. Watson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232-8240 Phone: (615) 343-3447
| | - W. Clay Spencer
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232-8240 Phone: (615) 343-3447
| | - Tim O’Brien
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232-8240 Phone: (615) 343-3447
| | - Byeong Cha
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232-8240 Phone: (615) 343-3447
| | - Adi Albeg
- Department of Medical Neurobiology, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Millet Treinin
- Department of Medical Neurobiology, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - David M. Miller
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232-8240 Phone: (615) 343-3447
- Vanderbilt Kennedy Center
- Program in Neuroscience, Vanderbilt University
| |
Collapse
|
25
|
Kubo A, Suzuki N, Yuan X, Nakai K, Satoh N, Imai KS, Satou Y. Genomic cis-regulatory networks in the early Ciona intestinalis embryo. Development 2010; 137:1613-23. [PMID: 20392745 DOI: 10.1242/dev.046789] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Precise spatiotemporal gene expression during animal development is achieved through gene regulatory networks, in which sequence-specific transcription factors (TFs) bind to cis-regulatory elements of target genes. Although numerous cis-regulatory elements have been identified in a variety of systems, their global architecture in the gene networks that regulate animal development is not well understood. Here, we determined the structure of the core networks at the cis-regulatory level in early embryos of the chordate Ciona intestinalis by chromatin immunoprecipitation (ChIP) of 11 TFs. The regulatory systems of the 11 TF genes examined were tightly interconnected with one another. By combining analysis of the ChIP data with the results of previous comprehensive analyses of expression profiles and knockdown of regulatory genes, we found that most of the previously determined interactions are direct. We focused on cis-regulatory networks responsible for the Ciona mesodermal tissues by examining how the networks specify these tissues at the level of their cis-regulatory architecture. We also found many interactions that had not been predicted by simple gene knockdown experiments, and we showed that a significant fraction of TF-DNA interactions make major contributions to the regulatory control of target gene expression.
Collapse
Affiliation(s)
- Atsushi Kubo
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Challenges for modeling global gene regulatory networks during development: Insights from Drosophila. Dev Biol 2010; 340:161-9. [DOI: 10.1016/j.ydbio.2009.10.032] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 10/14/2009] [Accepted: 10/21/2009] [Indexed: 12/26/2022]
|
27
|
C. elegans BED domain transcription factor BED-3 controls lineage-specific cell proliferation during organogenesis. Dev Biol 2009; 338:226-36. [PMID: 20005870 PMCID: PMC2862168 DOI: 10.1016/j.ydbio.2009.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 11/09/2009] [Accepted: 12/04/2009] [Indexed: 11/23/2022]
Abstract
The control of cell division is critical to organogenesis, but how this control is achieved is not fully understood. We found that mutations in bed-3, encoding a BED Zn-finger domain transcription factor, confer a phenotype where a specific set of cell divisions during vulval organogenesis is lost. Unlike general cell cycle regulators in Caenorhabditis elegans, the function of bed-3 is restricted to specific lineages. Transcriptional reporters suggest that bed-3 is expressed in a limited number of cell types including vulval cells whose divisions are affected in bed-3 mutants. A bed-3 mutation also affects the expression pattern of the cdh-3 cadherin gene in the vulva. The phenotype of bed-3 mutants is similar to the phenotype caused by mutations in cog-1 (Nkx6), a component of a gene regulatory network controlling cell type specific gene expression in the vulval lineage. These results suggest that bed-3 is a key component linking the gene regulatory network controlling cell-type specification to control of cell division during vulval organogenesis.
Collapse
|
28
|
Vermeirssen V, Joshi A, Michoel T, Bonnet E, Casneuf T, Van de Peer Y. Transcription regulatory networks in Caenorhabditis elegans inferred through reverse-engineering of gene expression profiles constitute biological hypotheses for metazoan development. MOLECULAR BIOSYSTEMS 2009; 5:1817-30. [PMID: 19763340 DOI: 10.1039/b908108a] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Differential gene expression governs the development, function and pathology of multicellular organisms. Transcription regulatory networks study differential gene expression at a systems level by mapping the interactions between regulatory proteins and target genes. While microarray transcription profiles are the most abundant data for gene expression, it remains challenging to correctly infer the underlying transcription regulatory networks. The reverse-engineering algorithm LeMoNe (learning module networks) uses gene expression profiles to extract ensemble transcription regulatory networks of coexpression modules and their prioritized regulators. Here we apply LeMoNe to a compendium of microarray studies of the worm Caenorhabditis elegans. We obtain 248 modules with a regulation program for 5020 genes and 426 regulators and a total of 24 012 predicted transcription regulatory interactions. Through GO enrichment analysis, comparison with the gene-gene association network WormNet and integration of other biological data, we show that LeMoNe identifies functionally coherent coexpression modules and prioritizes regulators that relate to similar biological processes as the module genes. Furthermore, we can predict new functional relationships for uncharacterized genes and regulators. Based on modules involved in molting, meiosis and oogenesis, ciliated sensory neurons and mitochondrial metabolism, we illustrate the value of LeMoNe as a biological hypothesis generator for differential gene expression in greater detail. In conclusion, through reverse-engineering of C. elegans expression data, we obtained transcription regulatory networks that can provide further insight into metazoan development.
Collapse
|
29
|
Jovelin R. Rapid sequence evolution of transcription factors controlling neuron differentiation in Caenorhabditis. Mol Biol Evol 2009; 26:2373-86. [PMID: 19589887 DOI: 10.1093/molbev/msp142] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Whether phenotypic evolution proceeds predominantly through changes in regulatory sequences is a controversial issue in evolutionary genetics. Ample evidence indicates that the evolution of gene regulatory networks via changes in cis-regulatory sequences is an important determinant of phenotypic diversity. However, recent experimental work suggests that the role of transcription factor (TF) divergence in developmental evolution may be underestimated. In order to help understand what levels of constraints are acting on the coding sequence of developmental regulatory genes, evolutionary rates were investigated among 48 TFs required for neuronal development in Caenorhabditis elegans. Allelic variation was then sampled for 28 of these genes within a population of the related species Caenorhabditis remanei. Neuronal TFs are more divergent, both within and between species, than structural genes. TFs affecting different neuronal classes are under different levels of selective constraints. The regulatory genes controlling the differentiation of chemosensory neurons evolve particularly fast and exhibit higher levels of within- and between-species nucleotide variation than TFs required for the development of several neuronal classes and TFs required for motorneuron differentiation. The TFs affecting chemosensory neuron development are also more divergent than chemosensory genes expressed in the neurons they differentiate. These results illustrate that TFs are not as highly constrained as commonly thought and suggest that the role of divergence in developmental regulatory genes during the evolution of gene regulatory networks requires further attention.
Collapse
Affiliation(s)
- Richard Jovelin
- Center for Ecology and Evolutionary Biology, University of Oregon, Oregon, USA.
| |
Collapse
|
30
|
Sethi AJ, Angerer RC, Angerer LM. Gene regulatory network interactions in sea urchin endomesoderm induction. PLoS Biol 2009; 7:e1000029. [PMID: 19192949 PMCID: PMC2634790 DOI: 10.1371/journal.pbio.1000029] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Accepted: 12/17/2008] [Indexed: 12/18/2022] Open
Abstract
A major goal of contemporary studies of embryonic development is to understand large sets of regulatory changes that accompany the phenomenon of embryonic induction. The highly resolved sea urchin pregastrular endomesoderm-gene regulatory network (EM-GRN) provides a unique framework to study the global regulatory interactions underlying endomesoderm induction. Vegetal micromeres of the sea urchin embryo constitute a classic endomesoderm signaling center, whose potential to induce archenteron formation from presumptive ectoderm was demonstrated almost a century ago. In this work, we ectopically activate the primary mesenchyme cell-GRN (PMC-GRN) that operates in micromere progeny by misexpressing the micromere determinant Pmar1 and identify the responding EM-GRN that is induced in animal blastomeres. Using localized loss-of -function analyses in conjunction with expression of endo16, the molecular definition of micromere-dependent endomesoderm specification, we show that the TGFbeta cytokine, ActivinB, is an essential component of this induction in blastomeres that emit this signal, as well as in cells that respond to it. We report that normal pregastrular endomesoderm specification requires activation of the Pmar1-inducible subset of the EM-GRN by the same cytokine, strongly suggesting that early micromere-mediated endomesoderm specification, which regulates timely gastrulation in the sea urchin embryo, is also ActivinB dependent. This study unexpectedly uncovers the existence of an additional uncharacterized micromere signal to endomesoderm progenitors, significantly revising existing models. In one of the first network-level characterizations of an intercellular inductive phenomenon, we describe an important in vivo model of the requirement of ActivinB signaling in the earliest steps of embryonic endomesoderm progenitor specification.
Collapse
|
31
|
Cis-regulatory mutations in the Caenorhabditis elegans homeobox gene locus cog-1 affect neuronal development. Genetics 2009; 181:1679-86. [PMID: 19189954 DOI: 10.1534/genetics.108.097832] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We apply here comparative genome hybridization as a novel tool to identify the molecular lesion in two Caenorhabditis elegans mutant strains that affect a neuronal cell fate decision. The phenotype of the mutant strains resembles those of the loss-of-function alleles of the cog-1 homeobox gene, an inducer of the fate of the gustatory neuron ASER. We find that both lesions map to the cis-regulatory control region of cog-1 and affect a phylogenetically conserved binding site for the C2H2 zinc-finger transcription factor CHE-1, a previously known regulator of cog-1 expression in ASER. Identification of this CHE-1-binding site as a critical regulator of cog-1 expression in the ASER in vivo represents one of the rare demonstrations of the in vivo relevance of an experimentally determined or predicted transcription-factor-binding site. Aside from the mutationally defined CHE-1-binding site, cog-1 contains a second, functional CHE-1-binding site, which in isolation is sufficient to drive reporter gene expression in the ASER but in an in vivo context is apparently insufficient for promoting appropriate ASER expression. The cis-regulatory control regions of other ASE-expressed genes also contain ASE motifs that can promote ASE neuron expression when isolated from their genomic context, but appear to depend on multiple ASE motifs in their normal genomic context. The multiplicity of cis-regulatory elements may ensure the robustness of gene expression.
Collapse
|
32
|
The Caenorhabditis elegans vulva: a post-embryonic gene regulatory network controlling organogenesis. Proc Natl Acad Sci U S A 2008; 105:20095-9. [PMID: 19104047 DOI: 10.1073/pnas.0806377105] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Caenorhabditis elegans vulva is an elegant model for dissecting a gene regulatory network (GRN) that directs postembryonic organogenesis. The mature vulva comprises seven cell types (vulA, vulB1, vulB2, vulC, vulD, vulE, and vulF), each with its own unique pattern of spatial and temporal gene expression. The mechanisms that specify these cell types in a precise spatial pattern are not well understood. Using reverse genetic screens, we identified novel components of the vulval GRN, including nhr-113 in vulA. Several transcription factors (lin-11, lin-29, cog-1, egl-38, and nhr-67) interact with each other and act in concert to regulate target gene expression in the diverse vulval cell types. For example, egl-38 (Pax2/5/8) stabilizes the vulF fate by positively regulating vulF characteristics and by inhibiting characteristics associated with the neighboring vulE cells. nhr-67 and egl-38 regulate cog-1, helping restrict its expression to vulE. Computational approaches have been successfully used to identify functional cis-regulatory motifs in the zmp-1 (zinc metalloproteinase) promoter. These results provide an overview of the regulatory network architecture for each vulval cell type.
Collapse
|
33
|
Marri S, Gupta BP. Dissection of lin-11 enhancer regions in Caenorhabditis elegans and other nematodes. Dev Biol 2008; 325:402-11. [PMID: 18950616 DOI: 10.1016/j.ydbio.2008.09.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Revised: 09/16/2008] [Accepted: 09/16/2008] [Indexed: 10/21/2022]
Abstract
The Caenorhabditis elegans LIM homeobox gene lin-11 plays crucial roles in the morphogenesis of the reproductive system and differentiation of several neurons. The expression of lin-11 in different tissues is regulated by enhancer regions located upstream as well as within lin-11 introns. These regions are functionally separable suggesting that multiple regulatory inputs operate to control the spatiotemporal pattern of lin-11 expression. To further dissect apart the nature of lin-11 regulation we focused on three Caenorhabditis species C. briggsae, C. remanei, and C. brenneri that are substantially diverged from C. elegans but share almost identical vulval morphology. We show that, in these species, the 5' region of lin-11 possesses conserved sequences to activate lin-11 expression in the reproductive system. Analysis of the in vivo role of these sequences in C. elegans has led to the identification of three functionally distinct enhancers for the vulva, VC neurons, and uterine pi lineage cells. We found that the pi enhancer is regulated by FOS homolog FOS-1 and LIN-12/Notch pathway effectors, LAG-1 (Su(H)/CBF1 family) and EGL-43 (EVI1 family). These results indicate that multiple factors cooperate to regulate pi-specific expression of lin-11 and together with other findings suggest that the mechanism of lin-11 regulation by LIN-12/Notch signaling is evolutionarily conserved in Caenorhabditis species. Our work demonstrates that 4-way comparison is a powerful tool to study conserved mechanisms of gene regulation in C. elegans and other nematodes.
Collapse
Affiliation(s)
- Sujatha Marri
- Department of Biology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | | |
Collapse
|
34
|
Nikolaev LG, Akopov SB, Chernov IP, Sverdlov ED. Maps of cis-Regulatory Nodes in Megabase Long Genome Segments are an Inevitable Intermediate Step Toward Whole Genome Functional Mapping. Curr Genomics 2008; 8:137-49. [PMID: 18660850 DOI: 10.2174/138920207780368178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 02/22/2007] [Accepted: 02/27/2007] [Indexed: 11/22/2022] Open
Abstract
The availability of complete human and other metazoan genome sequences has greatly facilitated positioning and analysis of various genomic functional elements, with initial emphasis on coding sequences. However, complete functional maps of sequenced eukaryotic genomes should include also positions of all non-coding regulatory elements. Unfortunately, experimental data on genomic positions of a multitude of regulatory sequences, such as enhancers, silencers, insulators, transcription terminators, and replication origins are very limited, especially at the whole genome level. Since most genomic regulatory elements (e.g. enhancers) are generally gene-, tissue-, or cell-specific, the prediction of these elements by computational methods is difficult and often ambiguous. Therefore, the development of high-throughput experimental approaches for identifying and mapping genomic functional elements is highly desirable. At the same time, the creation of whole-genome map of hundreds of thousands of regulatory elements in several hundreds of tissue/cell types is presently far beyond our capabilities. A possible alternative for the whole genome approach is to concentrate efforts on individual genomic segments and then to integrate the data obtained into a whole genome functional map. Moreover, the maps of polygenic fragments with functional cis-regulatory elements would provide valuable data on complex regulatory systems, including their variability and evolution. Here, we reviewed experimental approaches to the realization of these ideas, including our own developments of experimental techniques for selection of cis-acting functionally active DNA fragments from large (megabase-sized) segments of mammalian genomes.
Collapse
Affiliation(s)
- Lev G Nikolaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya,117997, Moscow, Russia
| | | | | | | |
Collapse
|
35
|
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: 33] [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
|
36
|
Shoguchi E, Hamaguchi M, Satoh N. Genome-wide network of regulatory genes for construction of a chordate embryo. Dev Biol 2008; 316:498-509. [DOI: 10.1016/j.ydbio.2008.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 12/29/2007] [Accepted: 01/09/2008] [Indexed: 11/26/2022]
|
37
|
Changing of the cell division axes drives vulva evolution in nematodes. Dev Biol 2008; 313:142-54. [DOI: 10.1016/j.ydbio.2007.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 09/20/2007] [Accepted: 10/07/2007] [Indexed: 02/06/2023]
|
38
|
Jakobsen JS, Braun M, Astorga J, Gustafson EH, Sandmann T, Karzynski M, Carlsson P, Furlong EE. Temporal ChIP-on-chip reveals Biniou as a universal regulator of the visceral muscle transcriptional network. Genes Dev 2007; 21:2448-60. [PMID: 17908931 PMCID: PMC1993875 DOI: 10.1101/gad.437607] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Smooth muscle plays a prominent role in many fundamental processes and diseases, yet our understanding of the transcriptional network regulating its development is very limited. The FoxF transcription factors are essential for visceral smooth muscle development in diverse species, although their direct regulatory role remains elusive. We present a transcriptional map of Biniou (a FoxF transcription factor) and Bagpipe (an Nkx factor) activity, as a first step to deciphering the developmental program regulating Drosophila visceral muscle development. A time course of chromatin immunoprecipitatation followed by microarray analysis (ChIP-on-chip) experiments and expression profiling of mutant embryos reveal a dynamic map of in vivo bound enhancers and direct target genes. While Biniou is broadly expressed, it regulates enhancers driving temporally and spatially restricted expression. In vivo reporter assays indicate that the timing of Biniou binding is a key trigger for the time span of enhancer activity. Although bagpipe and biniou mutants phenocopy each other, their regulatory potential is quite different. This network architecture was not apparent from genetic studies, and highlights Biniou as a universal regulator in all visceral muscle, regardless of its developmental origin or subsequent function. The regulatory connection of a number of Biniou target genes is conserved in mice, suggesting an ancient wiring of this developmental program.
Collapse
Affiliation(s)
| | - Martina Braun
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Jeanette Astorga
- Department of Cell and Molecular Biology, Goteborg University, SE-405 30 Goteborg, Sweden
| | | | - Thomas Sandmann
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Michal Karzynski
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Peter Carlsson
- Department of Cell and Molecular Biology, Goteborg University, SE-405 30 Goteborg, Sweden
| | - Eileen E.M. Furlong
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
- Corresponding author.E-MAIL ; FAX 49-6221-387166
| |
Collapse
|
39
|
Abstract
Controlling the differential expression of many thousands of genes is the most fundamental task of a developing organism. It requires an enormous computational device that has the capacity to process in parallel a vast number of regulatory inputs in the various cells of the embryo and come out with regulatory outputs that are tissue specific. The regulatory genome constitutes this computational device, comprising many thousands of processing units in the form of cis-regulatory modules. The interconnected cis-regulatory modules that control regulatory gene expression create a network that is the underlying mechanism of specification. In this review we use the gene regulatory network that governs endomesoderm specification in the sea urchin embryo to demonstrate the salient features of developmental gene regulatory networks and illustrate the information processing that is done by the regulatory sequences.
Collapse
|
40
|
Dupuy D, Bertin N, Hidalgo CA, Venkatesan K, Tu D, Lee D, Rosenberg J, Svrzikapa N, Blanc A, Carnec A, Carvunis AR, Pulak R, Shingles J, Reece-Hoyes J, Hunt-Newbury R, Viveiros R, Mohler WA, Tasan M, Roth FP, Le Peuch C, Hope IA, Johnsen R, Moerman DG, Barabási AL, Baillie D, Vidal M. Genome-scale analysis of in vivo spatiotemporal promoter activity in Caenorhabditis elegans. Nat Biotechnol 2007; 25:663-8. [PMID: 17486083 DOI: 10.1038/nbt1305] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 04/13/2007] [Indexed: 12/13/2022]
Abstract
Differential regulation of gene expression is essential for cell fate specification in metazoans. Characterizing the transcriptional activity of gene promoters, in time and in space, is therefore a critical step toward understanding complex biological systems. Here we present an in vivo spatiotemporal analysis for approximately 900 predicted C. elegans promoters (approximately 5% of the predicted protein-coding genes), each driving the expression of green fluorescent protein (GFP). Using a flow-cytometer adapted for nematode profiling, we generated 'chronograms', two-dimensional representations of fluorescence intensity along the body axis and throughout development from early larvae to adults. Automated comparison and clustering of the obtained in vivo expression patterns show that genes coexpressed in space and time tend to belong to common functional categories. Moreover, integration of this data set with C. elegans protein-protein interactome data sets enables prediction of anatomical and temporal interaction territories between protein partners.
Collapse
Affiliation(s)
- Denis Dupuy
- Center for Cancer Systems Biology, Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sandmann T, Girardot C, Brehme M, Tongprasit W, Stolc V, Furlong EE. A core transcriptional network for early mesoderm development in Drosophila melanogaster. Genes Dev 2007; 21:436-49. [PMID: 17322403 PMCID: PMC1804332 DOI: 10.1101/gad.1509007] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Embryogenesis is controlled by large gene-regulatory networks, which generate spatially and temporally refined patterns of gene expression. Here, we report the characteristics of the regulatory network orchestrating early mesodermal development in the fruitfly Drosophila, where the transcription factor Twist is both necessary and sufficient to drive development. Through the integration of chromatin immunoprecipitation followed by microarray analysis (ChIP-on-chip) experiments during discrete time periods with computational approaches, we identified >2000 Twist-bound cis-regulatory modules (CRMs) and almost 500 direct target genes. Unexpectedly, Twist regulates an almost complete cassette of genes required for cell proliferation in addition to genes essential for morophogenesis and cell migration. Twist targets almost 25% of all annotated Drosophila transcription factors, which may represent the entire set of regulators necessary for the early development of this system. By combining in vivo binding data from Twist, Mef2, Tinman, and Dorsal we have constructed an initial transcriptional network of early mesoderm development. The network topology reveals extensive combinatorial binding, feed-forward regulation, and complex logical outputs as prevalent features. In addition to binary activation and repression, we suggest that Twist binds to almost all mesodermal CRMs to provide the competence to integrate inputs from more specialized transcription factors.
Collapse
Affiliation(s)
- Thomas Sandmann
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Charles Girardot
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Marc Brehme
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Waraporn Tongprasit
- Genome Research Facility, NASA Ames Research Center, Moffet Field, California 94035, USA
| | - Viktor Stolc
- Genome Research Facility, NASA Ames Research Center, Moffet Field, California 94035, USA
| | - Eileen E.M. Furlong
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
- Corresponding author.E-MAIL ; FAX 49-6221-387166
| |
Collapse
|
42
|
Fernandes JS, Sternberg PW. The tailless ortholog nhr-67 regulates patterning of gene expression and morphogenesis in the C. elegans vulva. PLoS Genet 2007; 3:e69. [PMID: 17465684 PMCID: PMC1857733 DOI: 10.1371/journal.pgen.0030069] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Accepted: 03/16/2007] [Indexed: 12/31/2022] Open
Abstract
Regulation of spatio-temporal gene expression in diverse cell and tissue types is a critical aspect of development. Progression through Caenorhabditis elegans vulval development leads to the generation of seven distinct vulval cell types (vulA, vulB1, vulB2, vulC, vulD, vulE, and vulF), each with its own unique gene expression profile. The mechanisms that establish the precise spatial patterning of these mature cell types are largely unknown. Dissection of the gene regulatory networks involved in vulval patterning and differentiation would help us understand how cells generate a spatially defined pattern of cell fates during organogenesis. We disrupted the activity of 508 transcription factors via RNAi and assayed the expression of ceh-2, a marker for vulB fate during the L4 stage. From this screen, we identified the tailless ortholog nhr-67 as a novel regulator of gene expression in multiple vulval cell types. We find that one way in which nhr-67 maintains cell identity is by restricting inappropriate cell fusion events in specific vulval cells, namely vulE and vulF. nhr-67 exhibits a dynamic expression pattern in the vulval cells and interacts with three other transcriptional regulators cog-1 (Nkx6.1/6.2), lin-11 (LIM), and egl-38 (Pax2/5/8) to generate the composite expression patterns of their downstream targets. We provide evidence that egl-38 regulates gene expression in vulB1, vulC, vulD, vulE, as well as vulF cells. We demonstrate that the pairwise interactions between these regulatory genes are complex and vary among the seven cell types. We also discovered a striking regulatory circuit that affects a subset of the vulval lineages: cog-1 and nhr-67 inhibit both one another and themselves. We postulate that the differential levels and combinatorial patterns of lin-11, cog-1, and nhr-67 expression are a part of a regulatory code for the mature vulval cell types. During development, in which the single-celled egg generates a whole organism, cells become different from each other and form patterns of types of cells. It is these spatially defined fate patterns that underlie the formation of complex organs. Regulatory molecules called transcription factors influence the fate patterns that cells adopt. Understanding the role of these transcription factors and their interactions with other genes could tell us how cells establish a certain pattern of cell fates. This study focuses on studying how the seven cell types of the Caenorhabditis elegans vulva arise. This organ is one of the most intensively studied, and while the signaling network that initiates vulval development and sets the gross pattern of cell differentiation is well understood, the network of transcription factors that specifies the final cell fates is not understood. Here, we identify nhr-67, a new transcription factor that regulates patterning of cell fates in this organ. Transcription factors do not necessarily act alone, and we explore how NHR-67 works with three other regulatory factors (each with human homologs) to specify the different properties of the vulval cells. We also demonstrate that the interconnections of these transcription factors differ between these seven diverse cell types, which may partially account for how these cells acquire a certain pattern of cell fates.
Collapse
Affiliation(s)
- Jolene S Fernandes
- Division of Biology, California Institute of Technology, Pasadena, California, United States of America
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California, United States of America
| | - Paul W Sternberg
- Division of Biology, California Institute of Technology, Pasadena, California, United States of America
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
43
|
Rodrigues AJ, Coppola G, Santos C, Costa MDC, Ailion M, Sequeiros J, Geschwind DH, Maciel P. Functional genomics and biochemical characterization of the C. elegans orthologue of the Machado-Joseph disease protein ataxin-3. FASEB J 2007; 21:1126-36. [PMID: 17234717 DOI: 10.1096/fj.06-7002com] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Machado-Joseph disease (MJD) is the most common dominant spinocerebellar ataxia. MJD is caused by a CAG trinucleotide expansion in the ATXN3 gene, which encodes a protein named ataxin-3. Ataxin-3 has been proposed to act as a deubiquitinating enzyme in the ubiquitin-proteasome pathway and to be involved in transcriptional repression; nevertheless, its precise biological function(s) remains unknown. To gain further insight into the function of ataxin-3, we have identified the Caenorhabditis elegans orthologue of the ATXN3 gene and characterized its pattern of expression, developmental regulation, and subcellular localization. We demonstrate that, analogous to its human orthologue, C. elegans ataxin-3 has deubiquitinating activity in vitro against polyubiquitin chains with four or more ubiquitins, the minimum ubiquitin length for proteasomal targeting. To further evaluate C. elegans ataxin-3, we characterized the first known knockout animal models both phenotypically and biochemically, and found that the two C. elegans strains were viable and displayed no gross phenotype. To identify a molecular phenotype, we performed a large-scale microarray analysis of gene expression in both knockout strains. The data revealed a significant deregulation of core sets of genes involved in the ubiquitin-proteasome pathway, structure/motility, and signal transduction. This gene identification provides important clues that can help elucidate the specific biological role of ataxin-3 and unveil some of the physiological effects caused by its absence or diminished function.
Collapse
Affiliation(s)
- Ana-João Rodrigues
- Life and Health Sciences Research Institute, Health Sciences School, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Rajakumar V, Chamberlin HM. The Pax2/5/8 gene egl-38 coordinates organogenesis of the C. elegansegg-laying system. Dev Biol 2007; 301:240-53. [PMID: 17020758 DOI: 10.1016/j.ydbio.2006.08.068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2006] [Revised: 08/23/2006] [Accepted: 08/30/2006] [Indexed: 01/03/2023]
Abstract
Organogenesis requires coordinated development between different tissues and cells. The Pax family of transcription factors coordinates multiple developmental events in organs including the kidney, thyroid and the eye. Studying Pax factors in different organisms should identify unifying characteristics of organ development with implications to both development and disease. Here we investigate the function of the Pax2/5/8 transcription factor EGL-38 in coordinating development of the C. elegans egg-laying system. A functional egg-laying system requires cell fate specification events in the epithelial cells of the vulva as well as the mesodermal cells in the uterus of the somatic gonad. Using gene expression studies, genetic mutant analysis and genetic mosaics, we show that egl-38 has functions in both tissues of the organ to promote its development. We incorporate these results together with previous results to propose that EGL-38 plays multiple roles in the development of the egg-laying system, acting to both promote cell fate and to coordinate the development between different cell types. As the Pax2 gene performs similar roles in the development of the mammalian kidney, we show that coordinating organogenesis is a conserved function for Pax2/5/8 transcription factors.
Collapse
Affiliation(s)
- Vandana Rajakumar
- Department of Molecular Genetics, Ohio State University, 938 Biological Sciences Building, Columbus, OH 43210, USA
| | | |
Collapse
|
45
|
Abstract
In this essay I have attempted to provide clues relating to novel research avenues that are likely to have a broad impact on the field of stem cell biology. The specific examples, drawn from other areas, are meant to be instructive and are representative of many more similar efforts. I have suggested that the new areas of systems and synthetic biology may provide a truly deep level of understanding for many aspects of how stem cells make fate choices. Successful application of new avenues will require an integrative approach that combines experimental and computational techniques.
Collapse
Affiliation(s)
- Ihor R Lemischka
- Department of Molecular Biology, Washington Road, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
46
|
Choi MY, Romer AI, Hu M, Lepourcelet M, Mechoor A, Yesilaltay A, Krieger M, Gray PA, Shivdasani RA. A dynamic expression survey identifies transcription factors relevant in mouse digestive tract development. Development 2006; 133:4119-29. [PMID: 16971476 DOI: 10.1242/dev.02537] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Tissue-restricted transcription factors (TFs), which confer specialized cellular properties, are usually identified through sequence homology or cis-element analysis of lineage-specific genes; conventional modes of mRNA profiling often fail to report non-abundant TF transcripts. We evaluated the dynamic expression during mouse gut organogenesis of 1381 transcripts, covering nearly every known and predicted TF, and documented the expression of approximately 1000 TF genes in gastrointestinal development. Despite distinctive structures and functions, the stomach and intestine exhibit limited differences in TF genes. Among differentially expressed transcripts, a few are virtually restricted to the digestive tract, including Nr2e3, previously regarded as a photoreceptor-specific product. TFs that are enriched in digestive organs commonly serve essential tissue-specific functions, hence justifying a search for other tissue-restricted TFs. Computational data mining and experimental investigation focused interest on a novel homeobox TF, Isx, which appears selectively in gut epithelium and mirrors expression of the intestinal TF Cdx2. Isx-deficient mice carry a specific defect in intestinal gene expression: dysregulation of the high density lipoprotein (HDL) receptor and cholesterol transporter scavenger receptor class B, type I (Scarb1). Thus, integration of developmental gene expression with biological assessment, as described here for TFs, represents a powerful tool to investigate control of tissue differentiation.
Collapse
Affiliation(s)
- Michael Y Choi
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Balaji S, Babu MM, Iyer LM, Luscombe NM, Aravind L. Comprehensive analysis of combinatorial regulation using the transcriptional regulatory network of yeast. J Mol Biol 2006; 360:213-27. [PMID: 16762362 DOI: 10.1016/j.jmb.2006.04.029] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 04/01/2006] [Accepted: 04/14/2006] [Indexed: 11/23/2022]
Abstract
Studies on various model systems have shown that a relatively small number of transcription factors can set up strikingly complex spatial and temporal patterns of gene expression. This is achieved mainly by means of combinatorial or differential gene regulation, i.e. regulation of a gene by two or more transcription factors simultaneously or under different conditions. While a number of specific molecular details of the mechanisms of combinatorial regulation have emerged, our understanding of the general principles of combinatorial regulation on a genomic scale is still limited. In this work, we approach this problem by using the largest assembled transcriptional regulatory network for yeast. A specific network transformation procedure was used to obtain the co-regulatory network describing the set of all significant associations among transcription factors in regulating common target genes. Analysis of the global properties of the co-regulatory network suggested the presence of two classes of regulatory hubs: (i) those that make many co-regulatory associations, thus serving as integrators of disparate cellular processes; and (ii) those that make few co-regulatory associations, and thereby specifically regulate one or a few major cellular processes. Investigation of the local structure of the co-regulatory network revealed a significantly higher than expected modular organization, which might have emerged as a result of selection by functional constraints. These constraints probably emerge from the need for extensive modular backup and the requirement to integrate transcriptional inputs of multiple distinct functional systems. We then explored the transcriptional control of three major regulatory systems (ubiquitin signaling, protein kinase and transcriptional regulation systems) to understand specific aspects of their upstream control. As a result, we observed that ubiquitin E3 ligases are regulated primarily by unique transcription factors, whereas E1 and E2 enzymes share common transcription factors to a much greater extent. This suggested that the deployment of E3s unique to specific functional contexts may be mediated significantly at the transcriptional level. Likewise, we were able to uncover evidence for much higher upstream transcription control of transcription factors themselves, in comparison to components of other regulatory systems. We believe that the results presented here might provide a framework for testing the role of co-regulatory associations in eukaryotic transcriptional control.
Collapse
Affiliation(s)
- S Balaji
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda 20894, USA.
| | | | | | | | | |
Collapse
|
48
|
Kong YM, Macdonald RJ, Wen X, Yang P, Barbera VM, Swift GH. A comprehensive survey of DNA-binding transcription factor gene expression in human fetal and adult organs. Gene Expr Patterns 2006; 6:678-86. [PMID: 16487753 DOI: 10.1016/j.modgep.2006.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2005] [Accepted: 01/05/2006] [Indexed: 01/18/2023]
Abstract
A global survey of RNA from 14 fetal and 12 adult human organs by RT-PCR determined the expression patterns of 790 genes encoding DNA-binding transcription factors. The data can be sorted to identify sets of transcription factors with expression relatively restricted to a given organ or to particular organ groups. These data are a resource to help define the spectrum of transcription factor control, contribute to the elucidation of transcription factor cascades responsible for the development and maintenance of each organ, and provide a baseline to study the effects of disease or developmental defects.
Collapse
Affiliation(s)
- Y Megan Kong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, 75390, USA
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Development of the animal body plan is controlled by large gene regulatory networks (GRNs), and hence evolution of body plans must depend upon change in the architecture of developmental GRNs. However, these networks are composed of diverse components that evolve at different rates and in different ways. Because of the hierarchical organization of developmental GRNs, some kinds of change affect terminal properties of the body plan such as occur in speciation, whereas others affect major aspects of body plan morphology. A notable feature of the paleontological record of animal evolution is the establishment by the Early "Cambrian of virtually all phylum-level body plans. We identify a class of GRN component, the kernels" of the network, which, because of their developmental role and their particular internal structure, are most impervious to change. Conservation of phyletic body plans may have been due to the retention since pre-Cambrian time of GRN kernels, which underlie development of major body parts.
Collapse
Affiliation(s)
- Eric H Davidson
- Division of Biology 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| | | |
Collapse
|
50
|
Abstract
The synthesis of gene expression data and cis-regulatory analysis permits the elucidation of genomic regulatory networks. These networks provide a direct visualization of the functional interconnections among the regulatory genes and signaling components leading to cell-specific patterns of gene activity. Complex developmental processes are thereby illuminated in ways not revealed by the conventional analysis of individual genes. In this review, we describe emerging networks in several different model systems, and compare them with the gene regulatory network that controls dorsoventral patterning of the Drosophila embryo.
Collapse
Affiliation(s)
- Angelike Stathopoulos
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA.
| | | |
Collapse
|