Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Development ; 140(1): 82-92, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23154412

ABSTRACT

During development, extracellular signaling molecules interact with intracellular gene networks to control the specification, pattern and size of organs. One such signaling molecule is Hedgehog (Hh). Hh is known to act as a morphogen, instructing different fates depending on the distance to its source. However, how Hh, when signaling across a cell field, impacts organ-specific transcriptional networks is still poorly understood. Here, we investigate this issue during the development of the Drosophila ocellar complex. The development of this sensory structure, which is composed of three simple eyes (or ocelli) located at the vertices of a triangular patch of cuticle on the dorsal head, depends on Hh signaling and on the definition of three domains: two areas of eya and so expression--the prospective anterior and posterior ocelli--and the intervening interocellar domain. Our results highlight the role of the homeodomain transcription factor engrailed (en) both as a target and as a transcriptional repressor of hh signaling in the prospective interocellar region. Furthermore, we identify a requirement for the Notch pathway in the establishment of en maintenance in a Hh-independent manner. Therefore, hh signals transiently during the specification of the interocellar domain, with en being required here for hh signaling attenuation. Computational analysis further suggests that this network design confers robustness to signaling noise and constrains phenotypic variation. In summary, using genetics and modeling we have expanded the ocellar gene network to explain how the interaction between the Hh gradient and this gene network results in the generation of stable mutually exclusive gene expression domains. In addition, we discuss some general implications our model may have in some Hh-driven gene networks.


Subject(s)
Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eye/embryology , Gene Regulatory Networks/physiology , Hedgehog Proteins/genetics , Animals , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Gene Targeting/methods , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Models, Genetic , Repressor Proteins/metabolism , Repressor Proteins/physiology , Transcription Factors/genetics , Transcription Factors/physiology
2.
Nucleic Acids Res ; 41(6): 3600-18, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23393190

ABSTRACT

Co-option of cis-regulatory modules has been suggested as a mechanism for the evolution of expression sites during development. However, the extent and mechanisms involved in mobilization of cis-regulatory modules remains elusive. To trace the history of non-coding elements, which may represent candidate ancestral cis-regulatory modules affirmed during chordate evolution, we have searched for conserved elements in tunicate and vertebrate (Olfactores) genomes. We identified, for the first time, 183 non-coding sequences that are highly conserved between the two groups. Our results show that all but one element are conserved in non-syntenic regions between vertebrate and tunicate genomes, while being syntenic among vertebrates. Nevertheless, in all the groups, they are significantly associated with transcription factors showing specific functions fundamental to animal development, such as multicellular organism development and sequence-specific DNA binding. The majority of these regions map onto ultraconserved elements and we demonstrate that they can act as functional enhancers within the organism of origin, as well as in cross-transgenesis experiments, and that they are transcribed in extant species of Olfactores. We refer to the elements as 'Olfactores conserved non-coding elements'.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Urochordata/genetics , Vertebrates/genetics , Animals , Base Sequence , Conserved Sequence , Dogs , Fishes/genetics , Gene Regulatory Networks , Genes, Homeobox , Genetic Loci , Genome , Humans , Mammals/genetics , Mice , Synteny , Transcription, Genetic
3.
Zygote ; 23(3): 426-46, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24717667

ABSTRACT

Sea urchin embryos initiate cell specifications at the 16-cell stage by forming the mesomeres, macromeres and micromeres according to the relative position of the cells in the animal-vegetal axis. The most vegetal cells, micromeres, autonomously differentiate into skeletons and induce the neighbouring macromere cells to become mesoendoderm in the ß-catenin-dependent Wnt8 signalling pathway. Although the underlying molecular mechanism for this progression is largely unknown, we have previously reported that the initial events might be triggered by the Ca2+ influxes through the egg-originated L-type Ca2+ channels distributed asymmetrically along the animal-vegetal axis and through the stretch-dependent Ca2+channels expressed specifically in the micromere at the 4th cleavage. In this communication, we have examined whether one of the earliest Ca2+ targets, protein kinase C (PKC), plays a role in cell specification upstream of ß-catenin. To this end, we surveyed the expression pattern of ß-catenin in early embryos in the presence or absence of the specific peptide inhibitor of Hemicentrotus pulcherrimus PKC (HpPKC-I). Unlike previous knowledge, we have found that the initial nuclear entrance of ß-catenin does not take place in the micromeres, but in the macromeres at the 16-cell stage. Using the HpPKC-I, we have demonstrated further that PKC not only determines cell-specific nucleation of ß-catenin, but also regulates a variety of cell specification events in the early sea urchin embryos by modulating the cell adhesion structures, actin dynamics, intracellular Ca2+ signalling, and the expression of key transcription factors.


Subject(s)
Calcium/metabolism , Protein Kinase C/metabolism , Sea Urchins/embryology , beta Catenin/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Ectoderm/drug effects , Ectoderm/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Female , Gastrula/drug effects , Gastrula/metabolism , Gene Expression Regulation, Developmental/drug effects , Male , Molecular Sequence Data , Mouth/cytology , Mouth/embryology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Sea Urchins/metabolism , Signal Transduction , beta Catenin/genetics , beta Catenin/immunology
4.
PLoS Pathog ; 6(6): e1000938, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20548942

ABSTRACT

Transcriptional regulatory networks (TRNs) transduce environmental signals into coordinated output expression of the genome. Accordingly, they are central for the adaptation of bacteria to their living environments and in host-pathogen interactions. Few attempts have been made to describe a TRN for a human pathogen, because even in model organisms, such as Escherichia coli, the analysis is hindered by the large number of transcription factors involved. In light of the paucity of regulators, the gastric human pathogen Helicobacter pylori represents a very appealing system for understanding how bacterial TRNs are wired up to support infection in the host. Herein, we review and analyze the available molecular and "-omic" data in a coherent ensemble, including protein-DNA and protein-protein interactions relevant for transcriptional control of pathogenic responses. The analysis covers approximately 80% of the annotated H. pylori regulators, and provides to our knowledge the first in-depth description of a TRN for an important pathogen. The emerging picture indicates a shallow TRN, made of four main modules (origons) that process the physiological responses needed to colonize the gastric niche. Specific network motifs confer distinct transcriptional response dynamics to the TRN, while long regulatory cascades are absent. Rather than having a plethora of specialized regulators, the TRN of H. pylori appears to transduce separate environmental inputs by using different combinations of a small set of regulators.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Helicobacter Infections/genetics , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Helicobacter pylori/isolation & purification , Homeostasis/genetics , Humans , Transcription, Genetic
5.
Dev Dyn ; 240(1): 75-85, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21104743

ABSTRACT

The retinal determination gene network (RDGN) constitutes a paradigm of a gene network controlling organ specification and growth. In this study, we probed the RDGN in the Drosophila ocelli, a set of simple eyes located on the fly's dorsal head, by studying the expression, regulation, and function of toy, hth, eya, and so, members of the Pax6, Meis, Eya, and Six gene families. Our results highlight the role of the pax6 gene toy, together with the hh signaling pathway, in the initiation of eya and so expression; the engagement of eya and so in a feedback loop necessary for their full expression; and the interplay between hh signaling and hth as a mechanism of organ size control, as general regulatory steps in the specification of visual organs.


Subject(s)
Body Patterning/genetics , Drosophila Proteins/physiology , Drosophila/embryology , Drosophila/genetics , Eye/embryology , Homeodomain Proteins/physiology , Trans-Activators/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Eye/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Eye Proteins/physiology , Gene Expression Regulation, Developmental , Gene Regulatory Networks/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Models, Biological , Morphogenesis/genetics , Organ Size/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
6.
Dev Biol ; 344(2): 569-77, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20599903

ABSTRACT

Organ development is a complex process in which the activity of scores of interacting transcription factors and signaling pathways need to be integrated with proliferative growth. Developmental gene regulatory networks (GRNs) allow capturing essential regulatory pathways at a systems-level and provide an effective way of approaching such complexity. However typical GRNs studies focus on very early embryonic stages (usually pre-gastrulation) or late stages, when there is little or no cell proliferation, and therefore do not consider how organ growth is integrated in the developmental process. This can be conveniently investigated in the Drosophila melanogaster eye primordium. Here we present a working model meant to illustrate how during a critical period, the second larval stage, changes in cells' proliferative pattern are coordinated with the initiation of the Retinal Determination (RD) gene program. Such changes are regulated in response to two different sources of signal (Wnt1/wg and BMP2/4/dpp) produced by the anterior and posterior ends of the primordium, respectively. The dpp signaling is necessary to trigger the RD program. However in order for it to be effective, cells receiving Dpp have to be out of the wg signaling range. This is obtained thanks to the proliferative growth that precedes the onset of RD expression. With this network model many of the gene regulatory steps previously known to participate in growth and patterning are linked. Analysis of the model highlights a few essential regulatory principles, as well as poses new questions. In addition, these principles might operate during the growth and patterning of other organs.


Subject(s)
Drosophila melanogaster/growth & development , Drosophila/embryology , Eye/embryology , Animals , Cell Proliferation , Drosophila/genetics , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye/metabolism , Gene Regulatory Networks , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Wnt1 Protein
7.
Dev Biol ; 322(2): 425-34, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18718463

ABSTRACT

During sea urchin embryogenesis the spdri gene participates in two separate Gene Regulatory Networks (GRNs): the Primary Mesenchyme Cells' (PMCs) and the Oral Ectoderm's one. In both cases, activation of the gene follows initial specification events [Amore, G., Yavrouian, R., Peterson, K., Ransick, A., McClay, D., Davidson, E., 2003. Spdeadringer, a sea urchin embryo gene required separately in skeletogenic and oral ectoderm gene regulatory networks. Dev. Biol. 261, 55-81.]. We identified a portion of genomic DNA ("4.7IL" -3456;+389) which is sufficient to replicate sdpri's expression pattern in experiments of transgenesis, using a GFP reporter. In our experiments, the activation kinetic of 4.7IL-GFP was similar to that of the endogenous gene and the reporter responded to known spdri's transcriptional regulators (Ets1, Alx1, Gsc and Dri). Here we present a dissection of this regulatory region and a description of the modules involved in spdri's transcriptional regulation. Both in the PMCs' and Oral Ectoderm's expression phases, activation of spdri is obtained through the integration of three kinds of inputs: positive and globally distributed ones; negative ones (that prevent ectopic expression); positive and tissue-specific ones. Our results allow to expand the map of the regulatory connections at the spdri node, both in the PMCs and in the Oral Ectoderm Gene Regulatory Networks (GRNs).


Subject(s)
Ectoderm/embryology , Gene Regulatory Networks , Genetic Linkage , Sea Urchins/embryology , Animals , Ectoderm/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Mesoderm/embryology , Mesoderm/metabolism , Sea Urchins/genetics , Sea Urchins/metabolism , Transcriptional Activation
8.
PLoS One ; 7(2): e30428, 2012.
Article in English | MEDLINE | ID: mdl-22393353

ABSTRACT

In recent years, the preservation of biodiversity has become an important issue. Despite much public discussion, however, current practices in the food industry seldom take account of its potential economic importance: on the contrary, the introduction of industrialized agriculture practices over large areas has often resulted in a dramatic reduction in biodiversity.In this paper, we report on the remarkable degree of biodiversity in the wine yeast populations naturally present in a small area of Sicily (Italy) where traditional (non-industrial) winery practices are still in place. Out of more than 900 Saccharomyces yeast isolates recovered from late spontaneous fermentations, we detected at least 209 strains. Most interestingly, when evaluated at the fermentation and technological level, a number of isolates were found to be superior to industrial yeast strains. Out of a selected group, isolates from two strains were used for experimental fermentations in a winery environment and the quality of the wines produced was assessed at the technological, quality and sensory levels. Given that the characteristics of the wines produced were found to be industrially appealing, the study demonstrated the economic potential of preserving the patrimony of Sicilian yeast biodiversity and highlighted the importance of maintaining traditional wine making practices.


Subject(s)
Saccharomyces/genetics , Biodiversity , DNA Primers/genetics , DNA, Mitochondrial/metabolism , Ethanol/chemistry , Fermentation , Food Industry , Industrial Microbiology/methods , Phenotype , Polymorphism, Restriction Fragment Length , Reproducibility of Results , Saccharomyces cerevisiae/physiology , Sicily , Sulfites/chemistry , Wine
9.
PLoS One ; 3(12): e4025, 2008.
Article in English | MEDLINE | ID: mdl-19112499

ABSTRACT

Gene expression is widely perceived as exclusively controlled by the information contained in cis-regulatory regions. These are built in a modular way, each module being a cluster of binding sites for the transcription factors that control the level, the location and the time at which gene transcription takes place. On the other hand, results from our laboratory have shown that gene expression is affected by the compositional properties (GC levels) of the isochores in which genes are embedded, i.e. the genome context. To clarify how compositional genomic properties affect the way cis-regulatory information is utilized, we have changed the genome context of a GFP-reporter gene containing the complete cis-regulatory region of the gene spdeadringer (spdri), expressed during sea urchin embryogenesis. We have observed that GC levels higher or lower than those found in the natural genome context can alter the reporter expression pattern. We explain this as the result of an interference with the functionality of specific modules in the gene's cis-regulatory region. From these observations we derive the notion that the compositional properties of the genome context can affect cis-regulatory control of gene expression. Therefore although the way a gene works depends on the information contained in its cis-regulatory region, availability of such information depends on the compositional properties of the genomic context.


Subject(s)
Gene Expression Regulation, Developmental , Genome , Sea Urchins/embryology , Sea Urchins/genetics , Animals , DNA/metabolism , Embryonic Development , Gene Expression , Gene Expression Profiling , Genes, Reporter , Genetic Techniques , Genomics , Green Fluorescent Proteins/metabolism , Models, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
10.
Dev Biol ; 293(2): 555-64, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16574094

ABSTRACT

The Strongylocentrotus purpuratus cyclophilin1 gene (Sp-cyp1) is expressed exclusively in skeletogenic mesenchyme cells of the embryo, beginning in the micromere lineage of the early blastula stage and continuing after gastrulation during the syncytial deposition of the skeleton. This gene encodes a protein which is a member of the peptidyl-prolyl cis-trans isomerase (PPIase) family. Sp-cyp1 is among the differentiation genes activated in the skeletogenic territory as a terminal function of the endomesodermal gene regulatory network. Network perturbation analysis had predicted the skeletogenic regulators Ets1 and Deadringer (Dri) to be its driver inputs. Here, we show that a 218-bp cis-regulatory DNA fragment recapitulates skeletogenic Sp-cyp1 expression; that elimination of either Ets1 or Dri inputs severely depresses the activity of expression constructs containing this DNA fragment; and that Ets1 and Dri target sites within the 218 bp fragment are required for normal expression. This indicates that the predicted inputs are direct. Other studies indicate that the same inputs are evidently necessary for expression of several other skeletogenic differentiation genes, and these genes probably constitute a skeletogenic gene battery, defined by its Ets plus Dri regulatory inputs.


Subject(s)
Cyclophilins/genetics , Strongylocentrotus purpuratus/embryology , Strongylocentrotus purpuratus/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Endoderm/metabolism , Gene Expression Regulation, Developmental , Genetic Complementation Test , Mesoderm/metabolism , Molecular Sequence Data , Sequence Homology, Amino Acid
11.
Dev Biol ; 273(2): 226-43, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15328009

ABSTRACT

The Strongylocentrotus purpuratus hnf6 (Sphnf6) gene encodes a new member of the ONECUT family of transcription factors. The expression of hnf6 in the developing embryo is triphasic, and loss-of-function analysis shows that the Hnf6 protein is a transcription factor that has multiple distinct roles in sea urchin development. hnf6 is expressed maternally, and before gastrulation its transcripts are distributed globally. Early in development, its expression is required for the activation of PMC differentiation genes such as sm50, pm27, and msp130, but not for the activation of any known PMC regulatory genes, for example, alx, ets1, pmar1, or tbrain. Micromere transplantation experiments show that the gene is not involved in early micromere signaling. Early hnf6 expression is also required for expression of the mesodermal regulator gatac. The second known role of hnf6 is its participation after gastrulation in the oral ectoderm gene regulatory network (GRN), in which its expression is essential for the maintenance of the state of oral ectoderm specification. The third role is in the neurogenic ciliated band, which is foreshadowed exactly by a trapezoidal band of hnf6 expression at the border of the oral ectoderm and where it continues to be expressed through the end of embryogenesis. Neither oral ectoderm regulatory functions nor ciliated band formation occur normally in the absence of hnf6 expression.


Subject(s)
Sea Urchins/embryology , Sea Urchins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cloning, Molecular , DNA, Complementary/genetics , Gene Expression Regulation, Developmental , Gene Targeting , Mesoderm/cytology , Models, Biological , Molecular Sequence Data , Oligonucleotides, Antisense/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sea Urchins/genetics , Transcription Factors/genetics
12.
Dev Biol ; 269(1): 152-64, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15081364

ABSTRACT

We have identified an NK2 family homeodomain transcription factor, SpNK2.1, in the sea urchin Strongylocentrotus purpuratus whose transcripts are initially detected within the apical plate ectoderm of the hatching blastula and are confined to the apical organ at least through 2 weeks of development. Protein localization studies demonstrate that SpNK2.1 is restricted to the apical plate epithelium, but is excluded from the nucleus of serotonergic neurons. The expression profile of SpNK2.1 is dictated via two separate regulatory systems. Initially, SpNK2.1 is restricted to the apical pole domain by beta-catenin-dependent processes operating along the animal-vegetal axis, as evidenced by an expansion of SpNK2.1 expression upon cadherin overexpression. Starting at gastrulation, expression in the apical plate is maintained by SpDri, the sea urchin orthologue of dead ringer. Abrogation of SpDri results in the downregulation of SpNK2.1 after gastrulation, but SpDri is not necessary for the initial activation of SpNK2.1. Loss of function experiments using SpNK2.1-specific morpholino antisense oligonucleotides and SpNK2.1 overexpression experiments do not disrupt embryonic development and have no effect upon the development of neuronal components of the apical organ. Nonetheless, SpNK2.1 defines a new early territory of the sea urchin embryo.


Subject(s)
Body Patterning/genetics , Ectoderm/metabolism , Homeodomain Proteins/genetics , Sea Urchins/embryology , Animals , Body Patterning/physiology , Homeodomain Proteins/metabolism , Sea Urchins/metabolism
13.
Dev Biol ; 261(1): 55-81, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12941621

ABSTRACT

The Spdeadringer (Spdri) gene encodes an ARID-class transcription factor not previously known in sea urchin embryos. We show that Spdri is a key player in two separate developmental gene regulatory networks (GRNs). Spdri is expressed in a biphasic manner, first, after 12 h and until ingression in the skeletogenic descendants of the large micromeres; second, after about 20 h in the oral ectoderm, where its transcripts remain present at 30-50 mRNA molecules/cell far into development. In both territories, the periods of Spdri expression follow prior territorial specification events. The functional significance of each phase of expression was assessed by determining the effect of an alphaSpdri morpholino antisense oligonucleotide (MASO) on expression of 17 different mesodermal genes, 8 different oral ectoderm genes, and 18 other genes expressed specifically during endomesoderm specification. These effects were measured by quantitative PCR, supplemented by whole-mount in situ hybridization and morphological observations. Spdri is shown to act in the micromere descendants in the pathways that result in the expression of batteries of terminal skeletogenic genes. But, in the oral ectoderm, the same gene participates in the central GRN controlling oral ectoderm identity. Spdri is linked in the oral ectoderm GRN with several other genes encoding transcriptional regulators that are expressed specifically in various regions of the oral ectoderm. If its expression is blocked by treatment with alphaSpdri MASO, oral-specific features disappear and expression of the aboral ectoderm marker spec1 encompasses the whole of the ectoderm. In addition to disappearance of the oral ectoderm, morphological consequences of alphaSpdri MASO treatment include failure of spiculogenesis and of correct primary mesenchyme cell (pmc) patterning in the postgastrular embryo, and also failure of gastrulation. To further analyze these phenotypes, chimeric embryos were constructed consisting of two labeled micromeres combined with micromereless 4th cleavage host embryos; either the micromeres or the hosts contained alphaSpdri MASO. These experiments showed that, while Spdri expression is required autonomously for expression of skeletogenic genes prior to ingression, complete skeletogenesis also requires the expression of oral ectoderm patterning information. Presentation of this information on the oral side of the blastocoel in turn depends on Spdri expression in the oral ectoderm. Failure of gastrulation is not due to indirect interference with endomesodermal specification per se, since all endomesodermal genes tested function normally in alphaSpdri MASO embryos. Part of its cause is interference by alphaSpdri MASO with a late signaling function on the part of the micromere descendants that is needed to complete clearance of the Soxb1 repressor of gastrulation from the prospective endoderm, but in addition there is a nonautonomous oral ectoderm effect.


Subject(s)
Drosophila Proteins , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Sea Urchins/embryology , Sea Urchins/genetics , Amino Acid Sequence , Animals , Base Sequence , Chimera/genetics , DNA, Complementary/genetics , Ectoderm/cytology , Gastrula/cytology , Gene Expression Regulation, Developmental , Models, Biological , Molecular Sequence Data , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
14.
Dev Biol ; 246(1): 162-90, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12027441

ABSTRACT

We present the current form of a provisional DNA sequence-based regulatory gene network that explains in outline how endomesodermal specification in the sea urchin embryo is controlled. The model of the network is in a continuous process of revision and growth as new genes are added and new experimental results become available; see http://www.its.caltech.edu/~mirsky/endomeso.htm (End-mes Gene Network Update) for the latest version. The network contains over 40 genes at present, many newly uncovered in the course of this work, and most encoding DNA-binding transcriptional regulatory factors. The architecture of the network was approached initially by construction of a logic model that integrated the extensive experimental evidence now available on endomesoderm specification. The internal linkages between genes in the network have been determined functionally, by measurement of the effects of regulatory perturbations on the expression of all relevant genes in the network. Five kinds of perturbation have been applied: (1) use of morpholino antisense oligonucleotides targeted to many of the key regulatory genes in the network; (2) transformation of other regulatory factors into dominant repressors by construction of Engrailed repressor domain fusions; (3) ectopic expression of given regulatory factors, from genetic expression constructs and from injected mRNAs; (4) blockade of the beta-catenin/Tcf pathway by introduction of mRNA encoding the intracellular domain of cadherin; and (5) blockade of the Notch signaling pathway by introduction of mRNA encoding the extracellular domain of the Notch receptor. The network model predicts the cis-regulatory inputs that link each gene into the network. Therefore, its architecture is testable by cis-regulatory analysis. Strongylocentrotus purpuratus and Lytechinus variegatus genomic BAC recombinants that include a large number of the genes in the network have been sequenced and annotated. Tests of the cis-regulatory predictions of the model are greatly facilitated by interspecific computational sequence comparison, which affords a rapid identification of likely cis-regulatory elements in advance of experimental analysis. The network specifies genomically encoded regulatory processes between early cleavage and gastrula stages. These control the specification of the micromere lineage and of the initial veg(2) endomesodermal domain; the blastula-stage separation of the central veg(2) mesodermal domain (i.e., the secondary mesenchyme progenitor field) from the peripheral veg(2) endodermal domain; the stabilization of specification state within these domains; and activation of some downstream differentiation genes. Each of the temporal-spatial phases of specification is represented in a subelement of the network model, that treats regulatory events within the relevant embryonic nuclei at particular stages.


Subject(s)
Endoderm , Genes, Regulator , Mesoderm , Sea Urchins/embryology , Animals , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Science ; 295(5560): 1669-78, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11872831

ABSTRACT

Development of the body plan is controlled by large networks of regulatory genes. A gene regulatory network that controls the specification of endoderm and mesoderm in the sea urchin embryo is summarized here. The network was derived from large-scale perturbation analyses, in combination with computational methodologies, genomic data, cis-regulatory analysis, and molecular embryology. The network contains over 40 genes at present, and each node can be directly verified at the DNA sequence level by cis-regulatory analysis. Its architecture reveals specific and general aspects of development, such as how given cells generate their ordained fates in the embryo and why the process moves inexorably forward in developmental time.


Subject(s)
Endoderm/physiology , Gene Expression Regulation, Developmental , Genome , Mesoderm/physiology , Sea Urchins/embryology , Sea Urchins/genetics , Animals , Cell Lineage , Computational Biology , Embryonic Development , Endoderm/cytology , Gene Expression Profiling , Genes, Regulator , Mesoderm/cytology , Models, Biological , Models, Genetic , Morphogenesis , Regulatory Sequences, Nucleic Acid , Stem Cells/physiology , Systems Theory
SELECTION OF CITATIONS
SEARCH DETAIL