1
|
Amiri EE, Tenger-Trolander A, Li M, Thomas Julian A, Kasan K, Sanders SA, Blythe S, Schmidt-Ott U. Conservation of symmetry breaking at the level of chromatin accessibility between fly species with unrelated anterior determinants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632851. [PMID: 39868093 PMCID: PMC11760685 DOI: 10.1101/2025.01.13.632851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Establishing the anterior-posterior body axis is a fundamental process during embryogenesis, and the fruit fly, Drosophila melanogaster, provides one of the best-known case studies of this process. In Drosophila, localized mRNA of bicoid serves as anterior determinant (AD). Bicoid engages in a concentration-dependent competition with nucleosomes and initiates symmetry-breaking along the AP axis by promoting chromatin accessibility at the loci of transcription factor (TF) genes that are expressed in the anterior of the embryo. However, ADs of other fly species are unrelated and structurally distinct, and little is known about how they function. We addressed this question in the moth fly, Clogmia albipunctata, in which a maternally expressed transcript isoform of the pair-rule segmentation gene odd-paired is localized in the anterior egg and has been co-opted as AD. We provide a de novo assembly and annotation of the Clogmia genome and describe how knockdown of zelda and maternal odd-paired transcript affect chromatin accessibility and expression of TF-encoding loci. The results of these experiments suggest direct roles of Cal-Zld in opening and closing chromatin during nuclear cleavage cycles and show that Clogmia's maternal odd-paired activity promotes chromatin accessibility and anterior expression during the early phase of zygotic genome activation at Clogmia's homeobrain and sloppy-paired loci. We conclude that unrelated dipteran ADs initiate anterior-posterior axis-specification at the level of enhancer accessibility and that homeobrain and sloppy-paired homologs may serve a more widely conserved role in the initiation of anterior pattern formation given their early anterior expression and function in head development in other insects.
Collapse
Affiliation(s)
- Ezra E. Amiri
- The University of Chicago, Department of Organismal Biology and Anatomy, 1027 East 57 Street, Chicago, Illinois 60637, USA
| | - Ayse Tenger-Trolander
- The University of Chicago, Department of Organismal Biology and Anatomy, 1027 East 57 Street, Chicago, Illinois 60637, USA
| | - Muzi Li
- The University of Chicago, Department of Organismal Biology and Anatomy, 1027 East 57 Street, Chicago, Illinois 60637, USA
| | - Alexander Thomas Julian
- Illinois Institute of Technology, Department of Biology, 3105 South Dearborn Street, Chicago, Illinois 60616, USA
| | - Koray Kasan
- The University of Chicago, Department of Organismal Biology and Anatomy, 1027 East 57 Street, Chicago, Illinois 60637, USA
| | - Sheri A. Sanders
- Notre Dame University, 252 Galvin Life Science Center/Freimann Life Science Center, Notre Dame, Indiana 46556, USA
| | - Shelby Blythe
- Northwestern University, Department of Molecular Biosciences, 2205 Tech Drive, Evanston, Illinois 60208, USA
- Northwestern University and The University of Chicago, National Institute for Theory and Mathematics in Biology, 875 North Michigan Avenue, Suite 3500, Chicago, Illinois 60611, USA
| | - Urs Schmidt-Ott
- The University of Chicago, Department of Organismal Biology and Anatomy, 1027 East 57 Street, Chicago, Illinois 60637, USA
| |
Collapse
|
2
|
Sokolowski TR, Gregor T, Bialek W, Tkačik G. Deriving a genetic regulatory network from an optimization principle. Proc Natl Acad Sci U S A 2025; 122:e2402925121. [PMID: 39752518 PMCID: PMC11725783 DOI: 10.1073/pnas.2402925121] [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: 02/13/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
Many biological systems operate near the physical limits to their performance, suggesting that aspects of their behavior and underlying mechanisms could be derived from optimization principles. However, such principles have often been applied only in simplified models. Here, we explore a detailed mechanistic model of the gap gene network in the Drosophila embryo, optimizing its 50+ parameters to maximize the information that gene expression levels provide about nuclear positions. This optimization is conducted under realistic constraints, such as limits on the number of available molecules. Remarkably, the optimal networks we derive closely match the architecture and spatial gene expression profiles observed in the real organism. Our framework quantifies the tradeoffs involved in maximizing functional performance and allows for the exploration of alternative network configurations, addressing the question of which features are necessary and which are contingent. Our results suggest that multiple solutions to the optimization problem might exist across closely related organisms, offering insights into the evolution of gene regulatory networks.
Collapse
Affiliation(s)
- Thomas R. Sokolowski
- Institute of Science and Technology Austria, KlosterneuburgAT-3400, Austria
- Frankfurt Institute for Advanced Studies, Frankfurt am MainDE-60438, Germany
| | - Thomas Gregor
- Joseph Henry Laboratory of Physics and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Department of Stem Cell and Developmental Biology, UMR3738, Institut Pasteur, ParisFR-75015, France
| | - William Bialek
- Joseph Henry Laboratory of Physics and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ08544
- Center for Studies in Physics and Biology, Rockefeller University, New York, NY10065
| | - Gašper Tkačik
- Institute of Science and Technology Austria, KlosterneuburgAT-3400, Austria
| |
Collapse
|
3
|
Itoe MA, Shaw WR, Stryapunina I, Vidoudez C, Peng D, Du EW, Rinvee TA, Singh N, Yan Y, Hulai O, Thornburg KE, Catteruccia F. Maternal lipid mobilization is essential for embryonic development in the malaria vector Anopheles gambiae. PLoS Biol 2024; 22:e3002960. [PMID: 39689130 PMCID: PMC11703037 DOI: 10.1371/journal.pbio.3002960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/06/2025] [Accepted: 11/29/2024] [Indexed: 12/19/2024] Open
Abstract
Lipid metabolism is an essential component in reproductive physiology. While lipid mobilization has been implicated in the growth of Plasmodium falciparum malaria parasites in their Anopheles vectors, the role of this process in the reproductive biology of these mosquitoes remains elusive. Here, we show that impairing lipolysis in Anopheles gambiae, the major malaria vector, leads to embryonic lethality. Embryos derived from females in which we silenced the triglyceride lipase AgTL2 or the lipid storage droplet AgLSD1 develop normally during early embryogenesis but fail to hatch due to severely impaired metabolism. Embryonic lethality is efficiently recapitulated by exposing adult females to broad-spectrum lipase inhibitors prior to blood feeding, unveiling lipolysis as a potential target for inducing mosquito sterility. Our findings provide mechanistic insights into the importance of maternal lipid mobilization in embryonic health that may inform studies on human reproduction.
Collapse
Affiliation(s)
- Maurice A. Itoe
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - W. Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Iryna Stryapunina
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Charles Vidoudez
- Harvard Center for Mass Spectrometry, Cambridge, Massachusetts, United States of America
| | - Duo Peng
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Esrah W. Du
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Tasneem A. Rinvee
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Naresh Singh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Yan Yan
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Oleksandr Hulai
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Kate E. Thornburg
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| |
Collapse
|
4
|
Stryapunina I, Itoe MA, Trinh Q, Vidoudez C, Du E, Mendoza L, Hulai O, Kauffman J, Carew J, Shaw WR, Catteruccia F. Precise coordination between nutrient transporters ensures fertility in the malaria mosquito Anopheles gambiae. PLoS Genet 2024; 20:e1011145. [PMID: 38285728 PMCID: PMC10852252 DOI: 10.1371/journal.pgen.1011145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/08/2024] [Accepted: 01/20/2024] [Indexed: 01/31/2024] Open
Abstract
Females from many mosquito species feed on blood to acquire nutrients for egg development. The oogenetic cycle has been characterized in the arboviral vector Aedes aegypti, where after a bloodmeal, the lipid transporter lipophorin (Lp) shuttles lipids from the midgut and fat body to the ovaries, and a yolk precursor protein, vitellogenin (Vg), is deposited into the oocyte by receptor-mediated endocytosis. Our understanding of how the roles of these two nutrient transporters are mutually coordinated is however limited in this and other mosquito species. Here, we demonstrate that in the malaria mosquito Anopheles gambiae, Lp and Vg are reciprocally regulated in a timely manner to optimize egg development and ensure fertility. Defective lipid transport via Lp knockdown triggers abortive ovarian follicle development, leading to misregulation of Vg and aberrant yolk granules. Conversely, depletion of Vg causes an upregulation of Lp in the fat body in a manner that appears to be at least partially dependent on target of rapamycin (TOR) signaling, resulting in excess lipid accumulation in the developing follicles. Embryos deposited by Vg-depleted mothers are completely inviable, and are arrested early during development, likely due to severely reduced amino acid levels and protein synthesis. Our findings demonstrate that the mutual regulation of these two nutrient transporters is essential to safeguard fertility by ensuring correct nutrient balance in the developing oocyte, and validate Vg and Lp as two potential candidates for mosquito control.
Collapse
Affiliation(s)
- Iryna Stryapunina
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Maurice A. Itoe
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Queenie Trinh
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Charles Vidoudez
- Harvard Center for Mass Spectrometry, Cambridge, Massachusetts, United States of America
| | - Esrah Du
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Lydia Mendoza
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Oleksandr Hulai
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jamie Kauffman
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - John Carew
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - W. Robert Shaw
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Flaminia Catteruccia
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| |
Collapse
|
5
|
Santos-Moreno J, Tasiudi E, Kusumawardhani H, Stelling J, Schaerli Y. Robustness and innovation in synthetic genotype networks. Nat Commun 2023; 14:2454. [PMID: 37117168 PMCID: PMC10147661 DOI: 10.1038/s41467-023-38033-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/13/2023] [Indexed: 04/30/2023] Open
Abstract
Genotype networks are sets of genotypes connected by small mutational changes that share the same phenotype. They facilitate evolutionary innovation by enabling the exploration of different neighborhoods in genotype space. Genotype networks, first suggested by theoretical models, have been empirically confirmed for proteins and RNAs. Comparative studies also support their existence for gene regulatory networks (GRNs), but direct experimental evidence is lacking. Here, we report the construction of three interconnected genotype networks of synthetic GRNs producing three distinct phenotypes in Escherichia coli. Our synthetic GRNs contain three nodes regulating each other by CRISPR interference and governing the expression of fluorescent reporters. The genotype networks, composed of over twenty different synthetic GRNs, provide robustness in face of mutations while enabling transitions to innovative phenotypes. Through realistic mathematical modeling, we quantify robustness and evolvability for the complete genotype-phenotype map and link these features mechanistically to GRN motifs. Our work thereby exemplifies how GRN evolution along genotype networks might be driving evolutionary innovation.
Collapse
Affiliation(s)
- Javier Santos-Moreno
- Department of Fundamental Microbiology, University of Lausanne, Biophore Building, 1015, Lausanne, Switzerland
- Department of Medicine and Life Sciences, Pompeu Fabra University, 00803, Barcelona, Spain
| | - Eve Tasiudi
- Department of Biosystems Science and Engineering, ETH Zurich and SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Hadiastri Kusumawardhani
- Department of Fundamental Microbiology, University of Lausanne, Biophore Building, 1015, Lausanne, Switzerland
| | - Joerg Stelling
- Department of Biosystems Science and Engineering, ETH Zurich and SIB Swiss Institute of Bioinformatics, Basel, Switzerland.
| | - Yolanda Schaerli
- Department of Fundamental Microbiology, University of Lausanne, Biophore Building, 1015, Lausanne, Switzerland.
| |
Collapse
|
6
|
Cheatle Jarvela AM, Trelstad CS, Pick L. Anterior-posterior patterning of segments in Anopheles stephensi offers insights into the transition from sequential to simultaneous segmentation in holometabolous insects. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2023; 340:116-130. [PMID: 34734470 PMCID: PMC9061899 DOI: 10.1002/jez.b.23102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 11/10/2022]
Abstract
The gene regulatory network for segmentation in arthropods offers valuable insights into how networks evolve owing to the breadth of species examined and the extremely detailed knowledge gained in the model organism Drosophila melanogaster. These studies have shown that Drosophila's network represents a derived state that acquired changes to accelerate segment patterning, whereas most insects specify segments gradually as the embryo elongates. Such heterochronic shifts in segmentation have potentially emerged multiple times within holometabolous insects, resulting in many mechanistic variants and difficulties in isolating underlying commonalities that permit such shifts. Recent studies identified regulatory genes that work as timing factors, coordinating gene expression transitions during segmentation. These studies predict that changes in timing factor deployment explain shifts in segment patterning relative to other developmental events. Here, we test this hypothesis by characterizing the temporal and spatial expression of the pair-rule patterning genes in the malaria vector mosquito, Anopheles stephensi. This insect is a Dipteran (fly), like Drosophila, but represents an ancient divergence within this clade, offering a useful counterpart for evo-devo studies. In mosquito embryos, we observe anterior to posterior sequential addition of stripes for many pair-rule genes and a wave of broad timer gene expression across this axis. Segment polarity gene stripes are added sequentially in the wake of the timer gene wave and the full pattern is not complete until the embryo is fully elongated. This "progressive segmentation" mode in Anopheles displays commonalities with both Drosophila's rapid segmentation mechanism and sequential modes used by more distantly related insects.
Collapse
Affiliation(s)
- Alys M. Cheatle Jarvela
- Department of Entomology, University of Maryland, College Park, 4291 Fieldhouse Drive, College Park, MD 20742, U.S.A
| | - Catherine S. Trelstad
- Department of Entomology, University of Maryland, College Park, 4291 Fieldhouse Drive, College Park, MD 20742, U.S.A
| | - Leslie Pick
- Department of Entomology, University of Maryland, College Park, 4291 Fieldhouse Drive, College Park, MD 20742, U.S.A
| |
Collapse
|
7
|
Zheng L, Byadgi O, Rakhshaninejad M, Nauwynck H. Upregulation of torso-like protein (perforin) and granzymes B and G in non-adherent, lymphocyte-like haemocytes during a WSSV infection in shrimp. FISH & SHELLFISH IMMUNOLOGY 2022; 128:676-683. [PMID: 35985630 DOI: 10.1016/j.fsi.2022.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
Invertebrates only have an innate immunity in which haemocytes play an important role. In our lab, 5 subpopulations of haemocytes were identified in the past by an iodixanol density gradient: hyalinocytes, granulocytes, semi-granulocytes and two subpopulations of non-phagocytic cells. For the two latter subpopulations, the haemocytes have small cytoplasm rims, do not adhere to the bottom of plastic cell-culture grade wells and present folds in the nucleus. These characteristics are similar to those of mammalian lymphocytes. Therefore, they were designated lymphocyte-like haemocytes. Although little is known about their function, we hypothesize, based on their morphology, that they may have a cytotoxic activity. First, a fast isolation technique was developed to separate the non-adherent haemocytes from the adherent haemocytes. After 60 min incubation on cell culture plates, the non-adherent haemocytes were collected. The purity reached 93% as demonstrated by flow cytometry and light microscopy upon a Hematoxylin and Eosin (H&E) staining. Cytotoxicity by lymphocytes is mediated by molecules such as perforin and granzymes and therefore, we searched for their genes in the shrimp genome. Genes coding for a torso-like protein, granzyme B and granzyme G were identified. Primers were designed and RT-PCR/RT-qPCR assays were developed. The results demonstrated that torso-like protein, granzyme B and granzyme G were mainly expressed in non-adherent haemocytes. The shrimp torso-like protein gene was most related to that of the crab torso-like protein; granzyme B gene was most related to that of mouse granzyme B and granzyme G gene was most related to that of zebrafish granzyme G. In a 72-hour in vivo WSSV infection challenge, the mRNA expression of shrimp torso-like protein, granzyme B and granzyme G in haemocytes was increasing over time, which indicated that torso-like protein, granzyme B and granzyme G of shrimp haemocytes are involved in the immune response during a viral infection. In the future, antibodies will be raised against these proteins for more in-depth functional analyses.
Collapse
Affiliation(s)
- Liping Zheng
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium.
| | - Omkar Byadgi
- International Program in Ornamental Fish Technology and Aquatic Animal Health, National Pingtung University of Science and Technology, 91201, Pingtung, Taiwan
| | - Mostafa Rakhshaninejad
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| |
Collapse
|
8
|
Blondel L, Besse S, Rivard EL, Ylla G, Extavour CG. Evolution of a cytoplasmic determinant: evidence for the biochemical basis of functional evolution of the novel germ line regulator oskar. Mol Biol Evol 2021; 38:5491-5513. [PMID: 34550378 PMCID: PMC8662646 DOI: 10.1093/molbev/msab284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Germ line specification is essential in sexually reproducing organisms. Despite their critical role, the evolutionary history of the genes that specify animal germ cells is heterogeneous and dynamic. In many insects, the gene oskar is required for the specification of the germ line. However, the germ line role of oskar is thought to be a derived role resulting from co-option from an ancestral somatic role. To address how evolutionary changes in protein sequence could have led to changes in the function of Oskar protein that enabled it to regulate germ line specification, we searched for oskar orthologs in 1,565 publicly available insect genomic and transcriptomic data sets. The earliest-diverging lineage in which we identified an oskar ortholog was the order Zygentoma (silverfish and firebrats), suggesting that oskar originated before the origin of winged insects. We noted some order-specific trends in oskar sequence evolution, including whole gene duplications, clade-specific losses, and rapid divergence. An alignment of all known 379 Oskar sequences revealed new highly conserved residues as candidates that promote dimerization of the LOTUS domain. Moreover, we identified regions of the OSK domain with conserved predicted RNA binding potential. Furthermore, we show that despite a low overall amino acid conservation, the LOTUS domain shows higher conservation of predicted secondary structure than the OSK domain. Finally, we suggest new key amino acids in the LOTUS domain that may be involved in the previously reported Oskar−Vasa physical interaction that is required for its germ line role.
Collapse
Affiliation(s)
- Leo Blondel
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Savandara Besse
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Emily L Rivard
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Guillem Ylla
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Cassandra G Extavour
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
9
|
Chipman AD. The evolution of the gene regulatory networks patterning the Drosophila Blastoderm. Curr Top Dev Biol 2021; 139:297-324. [PMID: 32450964 DOI: 10.1016/bs.ctdb.2020.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Drosophila blastoderm gene regulatory network is one of the best studied networks in biology. It is composed of a series of tiered sub-networks that act sequentially to generate a primary segmental pattern. Many of these sub-networks have been studied in other arthropods, allowing us to reconstruct how each of them evolved over the transition from the arthropod ancestor to the situation seen in Drosophila today. I trace the evolution of each of these networks, showing how some of them have been modified significantly in Drosophila relative to the ancestral state while others are largely conserved across evolutionary timescales. I compare the putative ancestral arthropod segmentation network with that found in Drosophila and discuss how and why it has been modified throughout evolution, and to what extent this modification is unusual.
Collapse
Affiliation(s)
- Ariel D Chipman
- The Department of Ecology, Evolution & Behavior, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem, Israel.
| |
Collapse
|
10
|
Nascimento PVP, Almeida-Oliveira F, Macedo-Silva A, Ausina P, Motinha C, Sola-Penna M, Majerowicz D. Gene annotation of nuclear receptor superfamily genes in the kissing bug Rhodnius prolixus and the effects of 20-hydroxyecdysone on lipid metabolism. INSECT MOLECULAR BIOLOGY 2021; 30:297-314. [PMID: 33455040 DOI: 10.1111/imb.12696] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/29/2020] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
The hormone 20-hydroxyecdysone is fundamental for regulating moulting and metamorphosis in immature insects, and it plays a role in physiological regulation in adult insects. This hormone acts by binding and activating a receptor, the ecdysone receptor, which is part of the nuclear receptor gene superfamily. Here, we analyse the genome of the kissing bug Rhodnius prolixus to annotate the nuclear receptor superfamily genes. The R. prolixus genome displays a possible duplication of the HNF4 gene. All the analysed insect organs express most nuclear receptor genes as shown by RT-PCR. The quantitative PCR analysis showed that the RpEcR and RpUSP genes are highly expressed in the testis, while the RpHNF4-1 and RpHNF4-2 genes are more active in the fat body and ovaries and in the anterior midgut, respectively. Feeding does not induce detectable changes in the expression of these genes in the fat body. However, the expression of the RpHNF4-2 gene is always higher than that of RpHNF4-1. Treating adult females with 20-hydroxyecdysone increased the amount of triacylglycerol stored in the fat bodies by increasing their lipogenic capacity. These results indicate that 20-hydroxyecdysone acts on the lipid metabolism of adult insects, although the underlying mechanism is not clear.
Collapse
Affiliation(s)
- P V P Nascimento
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - F Almeida-Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - A Macedo-Silva
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P Ausina
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - C Motinha
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - M Sola-Penna
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - D Majerowicz
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Chiu M, Trigg B, Taracena M, Wells M. Diverse cellular morphologies during lumen maturation in Anopheles gambiae larval salivary glands. INSECT MOLECULAR BIOLOGY 2021; 30:210-230. [PMID: 33305876 PMCID: PMC8142555 DOI: 10.1111/imb.12689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/29/2020] [Accepted: 12/07/2020] [Indexed: 06/12/2023]
Abstract
Mosquitoes are the greatest animal threat to human health, causing hundreds of millions of infections and around 1 million deaths each year. All mosquito-borne pathogens must traverse the salivary glands (SGs) to be transmitted to the next host, making this organ an ideal target for interventions. The adult SG develops from precursor cells located in the larval SG duct bud. Characterization of the larval SG has been limited. We sought to better understand larval SG architecture, secretion and gene expression. We developed an optimized method for larval SG staining and surveyed hundreds of larval stage 4 (L4) SGs using fluorescence confocal microscopy. Remarkable variation in SG cell and chromatin organization differed among individuals and across the L4 stage. Lumen formation occurred during L4 stage through secretion likely involving a coincident cellular apical lipid enrichment and extracellular vesicle-like structures. Meta-analysis of microarray data showed that larval SG gene expression is divergent from adult SGs, more similar to larval gastric cecae, but different from other larval gut compartments. This work highlights the variable cell architecture of larval Anopheles gambiae SGs and provides candidate targets for genetic strategies aiming to disrupt SGs and transmission of mosquito-borne pathogens.
Collapse
Affiliation(s)
- M Chiu
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Malaria Research Institute, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - B Trigg
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Malaria Research Institute, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - M Taracena
- Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - M Wells
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Malaria Research Institute, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biomedical Sciences, Idaho College of Osteopathic Medicine (ICOM), Meridian, Idaho, USA
| |
Collapse
|
12
|
Adams KL, Abernathy DG, Willett BC, Selland EK, Itoe MA, Catteruccia F. Wolbachia cifB induces cytoplasmic incompatibility in the malaria mosquito vector. Nat Microbiol 2021; 6:1575-1582. [PMID: 34819638 PMCID: PMC8612931 DOI: 10.1038/s41564-021-00998-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 10/18/2021] [Indexed: 12/04/2022]
Abstract
Wolbachia, a maternally inherited intracellular bacterial species, can manipulate host insect reproduction by cytoplasmic incompatibility (CI), which results in embryo lethality in crosses between infected males and uninfected females. CI is encoded by two prophage genes, cifA and cifB. Wolbachia, coupled with the sterile insect technique, has been used in field trials to control populations of the dengue vector Aedes albopictus, but CI-inducing strains are not known to infect the malaria vector Anopheles gambiae. Here we show that cifA and cifB can induce conditional sterility in the malaria vector An. gambiae. We used transgenic expression of these Wolbachia-derived genes in the An. gambiae germline to show that cifB is sufficient to cause embryonic lethality and that cifB-induced sterility is rescued by cifA expression in females. When we co-expressed cifA and cifB in male mosquitoes, the CI phenotype was attenuated. In female mosquitoes, cifB impaired fertility, which was overcome by co-expression of cifA. Our findings pave the way towards using CI to control malaria mosquito vectors.
Collapse
Affiliation(s)
- Kelsey L. Adams
- grid.38142.3c000000041936754XDepartment of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Daniel G. Abernathy
- grid.38142.3c000000041936754XDepartment of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Bailey C. Willett
- grid.38142.3c000000041936754XDepartment of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Emily K. Selland
- grid.38142.3c000000041936754XDepartment of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Maurice A. Itoe
- grid.38142.3c000000041936754XDepartment of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
13
|
Lemke S, Kale G, Urbansky S. Comparing gastrulation in flies: Links between cell biology and the evolution of embryonic morphogenesis. Mech Dev 2020. [DOI: 10.1016/j.mod.2020.103648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
14
|
Cheatle Jarvela AM, Trelstad CS, Pick L. Regulatory gene function handoff allows essential gene loss in mosquitoes. Commun Biol 2020; 3:540. [PMID: 32999445 PMCID: PMC7528073 DOI: 10.1038/s42003-020-01203-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/02/2020] [Indexed: 11/09/2022] Open
Abstract
Regulatory genes are often multifunctional and constrained, which results in evolutionary conservation. It is difficult to understand how a regulatory gene could be lost from one species’ genome when it is essential for viability in closely related species. The gene paired is a classic Drosophila pair-rule gene, required for formation of alternate body segments in diverse insect species. Surprisingly, paired was lost in mosquitoes without disrupting body patterning. Here, we demonstrate that a paired family member, gooseberry, has acquired paired-like expression in the malaria mosquito Anopheles stephensi. Anopheles-gooseberry CRISPR-Cas9 knock-out mutants display pair-rule phenotypes and alteration of target gene expression similar to what is seen in Drosophila and beetle paired mutants. Thus, paired was functionally replaced by the related gene, gooseberry, in mosquitoes. Our findings document a rare example of a functional replacement of an essential regulatory gene and provide a mechanistic explanation of how such loss can occur. Cheatle Jarvela et al. demonstrate in the mosquito Anopheles stephensi that the paired gene was functionally replaced by the gene gooseberry, even though paired is essential in other insects such as fruit flies and beetles. This study contributes to the understanding of how essential genes are lost despite their importance during development.
Collapse
Affiliation(s)
| | | | - Leslie Pick
- Department of Entomology, University of Maryland, Collage Park, MD, USA.
| |
Collapse
|
15
|
Abstract
ABSTRACT
There is now compelling evidence that many arthropods pattern their segments using a clock-and-wavefront mechanism, analogous to that operating during vertebrate somitogenesis. In this Review, we discuss how the arthropod segmentation clock generates a repeating sequence of pair-rule gene expression, and how this is converted into a segment-polarity pattern by ‘timing factor’ wavefronts associated with axial extension. We argue that the gene regulatory network that patterns segments may be relatively conserved, although the timing of segmentation varies widely, and double-segment periodicity appears to have evolved at least twice. Finally, we describe how the repeated evolution of a simultaneous (Drosophila-like) mode of segmentation within holometabolan insects can be explained by heterochronic shifts in timing factor expression plus extensive pre-patterning of the pair-rule genes.
Collapse
Affiliation(s)
- Erik Clark
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
| | - Andrew D. Peel
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Michael Akam
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
| |
Collapse
|
16
|
Kulkarni A, Extavour CG. The Cricket Gryllus bimaculatus: Techniques for Quantitative and Functional Genetic Analyses of Cricket Biology. Results Probl Cell Differ 2019; 68:183-216. [PMID: 31598857 DOI: 10.1007/978-3-030-23459-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
All extant species are an outcome of nature's "experiments" during evolution, and hence multiple species need to be studied and compared to gain a thorough understanding of evolutionary processes. The field of evolutionary developmental biology (evo-devo) aspires to expand the number of species studied, because most functional genetic studies in animals have been limited to a small number of "traditional" model organisms, many of which belong to the same phylum (Chordata). The phylum Arthropoda, and particularly its component class Insecta, possesses many important characteristics that are considered favorable and attractive for evo-devo research, including an astonishing diversity of extant species and a wide disparity in body plans. The development of the most thoroughly investigated insect genetic model system to date, the fruit fly Drosophila melanogaster (a holometabolous insect), appears highly derived with respect to other insects and indeed with respect to most arthropods. In comparison, crickets (a basally branching hemimetabolous insect lineage compared to the Holometabola) are thought to embody many developmental features that make them more representative of insects. Here we focus on crickets as emerging models to study problems in a wide range of biological areas and summarize the currently available molecular, genomic, forward and reverse genetic, imaging and computational tool kit that has been established or adapted for cricket research. With an emphasis on the cricket species Gryllus bimaculatus, we highlight recent efforts made by the scientific community in establishing this species as a laboratory model for cellular biology and developmental genetics. This broad toolkit has the potential to accelerate many traditional areas of cricket research, including studies of adaptation, evolution, neuroethology, physiology, endocrinology, regeneration, and reproductive behavior. It may also help to establish newer areas, for example, the use of crickets as animal infection model systems and human food sources.
Collapse
Affiliation(s)
- Arpita Kulkarni
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Cassandra G Extavour
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
17
|
Chung CY, Hsiao YM, Huang TY, Chang TH, Chang CC. Germline expression of the hunchback orthologues in the asexual viviparous aphids: a conserved feature within the Aphididae. INSECT MOLECULAR BIOLOGY 2018; 27:752-765. [PMID: 29892979 DOI: 10.1111/imb.12514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In animals, differentiation of germline from soma usually takes place during embryogenesis. Genes and their products that are preferentially expressed in the embryonic germ cells are regarded as candidates for maintaining germline fate or promoting germline identity. In Drosophila, for example, the protein encoded by the germline gene vasa is specifically restricted to the germ cells, while products of the gap gene hunchback (hb), a somatic gene, are preferentially expressed in the neuroblasts. In this study, we report the expression of both messenger RNA and protein encoded by Aphb, an hb orthologue in the asexual viviparous pea aphid Acyrthosiphon pisum, in germ cells as well as in neuroblasts. We infer that expression of Aphb messenger RNA in the germ cells during the formation of germaria is required for the anterior localization of Aphb in the protruding oocytes. Germarial expression and anterior localization of ApKrüppel was also identified but, unlike Aphb, its expression was not detected in the migrating germ cells. Very similar patterns of hb expression were also identified in the green peach aphid Myzus persicae, suggesting that germline expression of hb is conserved within the Aphididae. To date, this pattern of hb germline expression has not been reported in other insects.
Collapse
Affiliation(s)
- C-Y Chung
- Laboratory for Genetics and Development, Department of Entomology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Y-M Hsiao
- Laboratory for Genetics and Development, Department of Entomology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - T-Y Huang
- Laboratory for Genetics and Development, Department of Entomology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - T-H Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| | - C-C Chang
- Laboratory for Genetics and Development, Department of Entomology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
18
|
Jiménez-Guri E, Wotton KR, Jaeger J. tarsal-less is expressed as a gap gene but has no gap gene phenotype in the moth midge Clogmia albipunctata. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180458. [PMID: 30225035 PMCID: PMC6124123 DOI: 10.1098/rsos.180458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/18/2018] [Indexed: 06/08/2023]
Abstract
Gap genes are involved in segment determination during early development of the vinegar fly Drosophila melanogaster and other dipteran insects (flies, midges and mosquitoes). They are expressed in overlapping domains along the antero-posterior (A-P) axis of the blastoderm embryo. While gap domains cover the entire length of the A-P axis in Drosophila, there is a region in the blastoderm of the moth midge Clogmia albipunctata, which lacks canonical gap gene expression. Is a non-canonical gap gene functioning in this area? Here, we characterize tarsal-less (tal) in C. albipunctata. The homologue of tal in the flour beetle Tribolium castaneum (called milles-pattes, mlpt) is a bona fide gap gene. We find that Ca-tal is expressed in the region previously reported as lacking gap gene expression. Using RNA interference, we study the interaction of Ca-tal with gap genes. We show that Ca-tal is regulated by gap genes, but only has a very subtle effect on tailless (Ca-tll), while not affecting other gap genes at all. Moreover, cuticle phenotypes of Ca-tal depleted embryos do not show any gap phenotype. We conclude that Ca-tal is expressed and regulated like a gap gene, but does not function as a gap gene in C. albipunctata.
Collapse
Affiliation(s)
- Eva Jiménez-Guri
- EMBL/CRG Research Unit in Systems Biology, Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Centre for Ecology and Conservation, College of Life and Environmental Sciences, University of Exeter, Penryn, Cornwall TR10 9EZ, UK
| | - Karl R. Wotton
- EMBL/CRG Research Unit in Systems Biology, Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Johannes Jaeger
- EMBL/CRG Research Unit in Systems Biology, Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
19
|
Jacobs J, Atkins M, Davie K, Imrichova H, Romanelli L, Christiaens V, Hulselmans G, Potier D, Wouters J, Taskiran II, Paciello G, González-Blas CB, Koldere D, Aibar S, Halder G, Aerts S. The transcription factor Grainy head primes epithelial enhancers for spatiotemporal activation by displacing nucleosomes. Nat Genet 2018; 50:1011-1020. [PMID: 29867222 PMCID: PMC6031307 DOI: 10.1038/s41588-018-0140-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 04/06/2018] [Indexed: 12/21/2022]
Abstract
Transcriptional enhancers function as docking platforms for combinations of transcription factors (TFs) to control gene expression. How enhancer sequences determine nucleosome occupancy, TF recruitment and transcriptional activation in vivo remains unclear. Using ATAC-seq across a panel of Drosophila inbred strains, we found that SNPs affecting binding sites of the TF Grainy head (Grh) causally determine the accessibility of epithelial enhancers. We show that deletion and ectopic expression of Grh cause loss and gain of DNA accessibility, respectively. However, although Grh binding is necessary for enhancer accessibility, it is insufficient to activate enhancers. Finally, we show that human Grh homologs-GRHL1, GRHL2 and GRHL3-function similarly. We conclude that Grh binding is necessary and sufficient for the opening of epithelial enhancers but not for their activation. Our data support a model positing that complex spatiotemporal expression patterns are controlled by regulatory hierarchies in which pioneer factors, such as Grh, establish tissue-specific accessible chromatin landscapes upon which other factors can act.
Collapse
Affiliation(s)
- Jelle Jacobs
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Mardelle Atkins
- VIB Center for Cancer Biology, Leuven, Belgium
- KU Leuven, Department of Oncology, Leuven, Belgium
| | - Kristofer Davie
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Hana Imrichova
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Lucia Romanelli
- VIB Center for Cancer Biology, Leuven, Belgium
- KU Leuven, Department of Oncology, Leuven, Belgium
| | - Valerie Christiaens
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Gert Hulselmans
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Delphine Potier
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Jasper Wouters
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | | | - Giulia Paciello
- Politecnico di Torino, Automatics and Informatics, Turin, Italy
| | - Carmen B González-Blas
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Duygu Koldere
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Sara Aibar
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium
- KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Georg Halder
- VIB Center for Cancer Biology, Leuven, Belgium
- KU Leuven, Department of Oncology, Leuven, Belgium
| | - Stein Aerts
- VIB Center for Brain and Disease Research, Laboratory of Computational Biology, Leuven, Belgium.
- KU Leuven, Department of Human Genetics, Leuven, Belgium.
| |
Collapse
|
20
|
Clark E. Dynamic patterning by the Drosophila pair-rule network reconciles long-germ and short-germ segmentation. PLoS Biol 2017; 15:e2002439. [PMID: 28953896 PMCID: PMC5633203 DOI: 10.1371/journal.pbio.2002439] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 10/09/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023] Open
Abstract
Drosophila segmentation is a well-established paradigm for developmental pattern formation. However, the later stages of segment patterning, regulated by the "pair-rule" genes, are still not well understood at the system level. Building on established genetic interactions, I construct a logical model of the Drosophila pair-rule system that takes into account the demonstrated stage-specific architecture of the pair-rule gene network. Simulation of this model can accurately recapitulate the observed spatiotemporal expression of the pair-rule genes, but only when the system is provided with dynamic "gap" inputs. This result suggests that dynamic shifts of pair-rule stripes are essential for segment patterning in the trunk and provides a functional role for observed posterior-to-anterior gap domain shifts that occur during cellularisation. The model also suggests revised patterning mechanisms for the parasegment boundaries and explains the aetiology of the even-skipped null mutant phenotype. Strikingly, a slightly modified version of the model is able to pattern segments in either simultaneous or sequential modes, depending only on initial conditions. This suggests that fundamentally similar mechanisms may underlie segmentation in short-germ and long-germ arthropods.
Collapse
Affiliation(s)
- Erik Clark
- Laboratory for Development and Evolution, Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Hunding A, Baumgartner S. Ancient role of ten-m/ odz in segmentation and the transition from sequential to syncytial segmentation. Hereditas 2017; 154:8. [PMID: 28461810 PMCID: PMC5408475 DOI: 10.1186/s41065-017-0029-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023] Open
Abstract
Background Until recently, mechanisms of segmentation established for Drosophila served as a paradigm for arthropod segmentation. However, with the discovery of gene expression waves in vertebrate segmentation, another paradigm based on oscillations linked to axial growth was established. The Notch pathway and hairy delay oscillator are basic components of this mechanism, as is the wnt pathway. With the establishment of oscillations during segmentation of the beetle Tribolium, a common segmentation mechanism may have been present in the last common ancestor of vertebrates and arthropods. However, the Notch pathway is not involved in segmentation of the initial Drosophila embryo. In arthropods, the engrailed, wingless pair has a much more conserved function in segmentation than most of the hierarchy established for Drosophila. Results Here, we work backwards from this conserved pair by discussing possible mechanisms which could have taken over the role of the Notch pathway. We propose a pivotal role for the large transmembrane protein Ten-m/Odz. Ten-m/Odz may have had an ancient role in cell-cell communication, parallel to the Notch and wnt pathways. The Ten-m protein binds to the membrane with properties which resemble other membrane-based biochemical oscillators. Conclusion We propose that such a simple transition could have formed the initial scaffold, on top of which the hierarchy, observed in the syncytium of dipterans, could have evolved.
Collapse
Affiliation(s)
- Axel Hunding
- Biophysical Chemistry, Department of Chemistry S01, H. C. 0rsted Institute, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| | - Stefan Baumgartner
- Department of Experimental Medical Sciences, Lund University, BMC D10, 22184 Lund, Sweden
| |
Collapse
|
22
|
The Function and Evolution of Nuclear Receptors in Insect Embryonic Development. Curr Top Dev Biol 2017; 125:39-70. [DOI: 10.1016/bs.ctdb.2017.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
23
|
Schmidt-Ott U, Lynch JA. Emerging developmental genetic model systems in holometabolous insects. Curr Opin Genet Dev 2016; 39:116-128. [PMID: 27399647 DOI: 10.1016/j.gde.2016.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/25/2016] [Accepted: 06/08/2016] [Indexed: 01/08/2023]
Abstract
The number of insect species that are amenable to functional genetic studies is growing rapidly and provides many new research opportunities in developmental and evolutionary biology. The holometabolous insects represent a disproportionate percentage of animal diversity and are thus well positioned to provide model species for a wide variety of developmental processes. Here we discuss emerging holometabolous models, and review some recent breakthroughs. For example, flies and midges were found to use structurally unrelated long-range pattern organizers, butterflies and moths revealed extensive pattern formation during oogenesis, new imaging possibilities in the flour beetle Tribolium castaneum showed how embryos break free of their extraembryonic membranes, and the complex genetics governing interspecies difference in head shape were revealed in Nasonia wasps.
Collapse
Affiliation(s)
- Urs Schmidt-Ott
- Department of Organismal Biology and Anatomy, University of Chicago, United States.
| | - Jeremy A Lynch
- Department of Biological Sciences, University of Illinois at Chicago, United States.
| |
Collapse
|
24
|
Rothschild JB, Tsimiklis P, Siggia ED, François P. Predicting Ancestral Segmentation Phenotypes from Drosophila to Anopheles Using In Silico Evolution. PLoS Genet 2016; 12:e1006052. [PMID: 27227405 PMCID: PMC4882032 DOI: 10.1371/journal.pgen.1006052] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/23/2016] [Indexed: 12/23/2022] Open
Abstract
Molecular evolution is an established technique for inferring gene homology but regulatory DNA turns over so rapidly that inference of ancestral networks is often impossible. In silico evolution is used to compute the most parsimonious path in regulatory space for anterior-posterior patterning linking two Dipterian species. The expression pattern of gap genes has evolved between Drosophila (fly) and Anopheles (mosquito), yet one of their targets, eve, has remained invariant. Our model predicts that stripe 5 in fly disappears and a new posterior stripe is created in mosquito, thus eve stripe modules 3+7 and 4+6 in fly are homologous to 3+6 and 4+5 in mosquito. We can place Clogmia on this evolutionary pathway and it shares the mosquito homologies. To account for the evolution of the other pair-rule genes in the posterior we have to assume that the ancestral Dipterian utilized a dynamic method to phase those genes in relation to eve. The last common ancestor of the fruit fly (Drosophila) and mosquito (Anopheles) lived more than 200 Million years ago. Can we use available data on insects alive today to infer what their ancestor looked like? In this manuscript, we focus on early embryonic development, when stripes of genetic expression appear and define the location of insect segments (“segmentation”). We use an evolutionary algorithm to reconstruct and predict dynamics of genes controlling stripes in the last common ancestor of fly and mosquito. We predict a new and different combinatorial logic of stripe formation in mosquito compared to fly, which is fully consistent with development of intermediate species such as moth-fly (Clogmia). Our simulations further suggest that the dynamics of gene expression in this last common ancestor were similar to other insects, such as wasps (Nasonia). Our method illustrates how computational methods inspired by machine learning and non-linear physics can be used to infer gene dynamics in species that disappeared millions of years ago.
Collapse
Affiliation(s)
- Jeremy B. Rothschild
- Physics Department, McGill University, Ernest Rutherford Physics Building, Montreal, Quebec, Canada
| | - Panagiotis Tsimiklis
- Physics Department, McGill University, Ernest Rutherford Physics Building, Montreal, Quebec, Canada
| | - Eric D. Siggia
- Center for Studies in Physics and Biology, The Rockefeller University, New York, New York, United States of America
| | - Paul François
- Physics Department, McGill University, Ernest Rutherford Physics Building, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
25
|
Crombach A, Wotton KR, Jiménez-Guri E, Jaeger J. Gap Gene Regulatory Dynamics Evolve along a Genotype Network. Mol Biol Evol 2016; 33:1293-307. [PMID: 26796549 PMCID: PMC4839219 DOI: 10.1093/molbev/msw013] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Developmental gene networks implement the dynamic regulatory mechanisms that pattern and shape the organism. Over evolutionary time, the wiring of these networks changes, yet the patterning outcome is often preserved, a phenomenon known as “system drift.” System drift is illustrated by the gap gene network—involved in segmental patterning—in dipteran insects. In the classic model organism Drosophila melanogaster and the nonmodel scuttle fly Megaselia abdita, early activation and placement of gap gene expression domains show significant quantitative differences, yet the final patterning output of the system is essentially identical in both species. In this detailed modeling analysis of system drift, we use gene circuits which are fit to quantitative gap gene expression data in M. abdita and compare them with an equivalent set of models from D. melanogaster. The results of this comparative analysis show precisely how compensatory regulatory mechanisms achieve equivalent final patterns in both species. We discuss the larger implications of the work in terms of “genotype networks” and the ways in which the structure of regulatory networks can influence patterns of evolutionary change (evolvability).
Collapse
Affiliation(s)
- Anton Crombach
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Karl R Wotton
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Eva Jiménez-Guri
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Johannes Jaeger
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
26
|
Franke FA, Mayer G. Expression study of the hunchback ortholog in embryos of the onychophoran Euperipatoides rowelli. Dev Genes Evol 2015; 225:207-19. [PMID: 26093940 DOI: 10.1007/s00427-015-0505-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/02/2015] [Indexed: 10/23/2022]
Abstract
Zinc finger transcription factors encoded by hunchback homologs play different roles in arthropods, including maternally mediated control, segmentation, and mesoderm and neural development. Knockdown experiments in spider and insect embryos have also revealed homeotic effects and gap phenotypes, the latter indicating a function of hunchback as a "gap gene". Although the expression pattern of hunchback has been analysed in representatives of all four major arthropod groups (chelicerates, myriapods, crustaceans and insects), nothing is known about its expression in one of the closest arthropod relatives, the Onychophora (velvet worms). We therefore examined the expression pattern of hunchback in embryos of the onychophoran Euperipatoides rowelli. Our transcriptomic and phylogenetic analyses revealed only one hunchback ortholog in this species. The putative Hunchback protein contains all nine zinc finger domains known from other protostomes. We found no indication of maternally contributed transcripts of hunchback in early embryos of E. rowelli. Its initial expression occurs in the ectodermal tissue of the antennal segment, followed by the jaw, slime papilla and trunk segments in an anterior-to-posterior progression. Later, hunchback expression is seen in the mesoderm of the developing limbs. A second "wave" of expression commences later in development in the antennal segment and continues posteriorly along each developing nerve cord. This expression is restricted to the neural tissues and does not show any segmental pattern. These findings are in line with the ancestral roles of hunchback in mesoderm and neural development, whereas we find no evidence for a putative function of hunchback as a "gap gene" in Onychophora.
Collapse
Affiliation(s)
- Franziska Anni Franke
- Animal Evolution & Development, Institute of Biology, University of Leipzig, Talstraße 33, D-04103, Leipzig, Germany,
| | | |
Collapse
|
27
|
Gordon KL, Arthur RK, Ruvinsky I. Phylum-Level Conservation of Regulatory Information in Nematodes despite Extensive Non-coding Sequence Divergence. PLoS Genet 2015; 11:e1005268. [PMID: 26020930 PMCID: PMC4447282 DOI: 10.1371/journal.pgen.1005268] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 05/09/2015] [Indexed: 11/28/2022] Open
Abstract
Gene regulatory information guides development and shapes the course of evolution. To test conservation of gene regulation within the phylum Nematoda, we compared the functions of putative cis-regulatory sequences of four sets of orthologs (unc-47, unc-25, mec-3 and elt-2) from distantly-related nematode species. These species, Caenorhabditis elegans, its congeneric C. briggsae, and three parasitic species Meloidogyne hapla, Brugia malayi, and Trichinella spiralis, represent four of the five major clades in the phylum Nematoda. Despite the great phylogenetic distances sampled and the extensive sequence divergence of nematode genomes, all but one of the regulatory elements we tested are able to drive at least a subset of the expected gene expression patterns. We show that functionally conserved cis-regulatory elements have no more extended sequence similarity to their C. elegans orthologs than would be expected by chance, but they do harbor motifs that are important for proper expression of the C. elegans genes. These motifs are too short to be distinguished from the background level of sequence similarity, and while identical in sequence they are not conserved in orientation or position. Functional tests reveal that some of these motifs contribute to proper expression. Our results suggest that conserved regulatory circuitry can persist despite considerable turnover within cis elements. To explore the phylogenetic limits of conservation of cis-regulatory elements, we used transgenesis to test the functions of enhancers of four genes from several species spanning the phylum Nematoda. While we found a striking degree of functional conservation among the examined cis elements, their DNA sequences lacked apparent conservation with the C. elegans orthologs. In fact, sequence similarity between C. elegans and the distantly related nematodes was no greater than would be expected by chance. Short motifs, similar to known regulatory sequences in C. elegans, can be detected in most of the cis elements. When tested, some of these sites appear to mediate regulatory function. However, they seem to have originated through motif turnover, rather than to have been preserved from a common ancestor. Our results suggest that gene regulatory networks are broadly conserved in the phylum Nematoda, but this conservation persists despite substantial reorganization of regulatory elements and could not be detected using naïve comparisons of sequence similarity.
Collapse
Affiliation(s)
- Kacy L. Gordon
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (KLG); (IR)
| | - Robert K. Arthur
- Department of Ecology and Evolution, The University of Chicago, Chicago, Illinois, United States of America
| | - Ilya Ruvinsky
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, Illinois, United States of America
- Department of Ecology and Evolution, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (KLG); (IR)
| |
Collapse
|
28
|
Duque T, Sinha S. What does it take to evolve an enhancer? A simulation-based study of factors influencing the emergence of combinatorial regulation. Genome Biol Evol 2015; 7:1415-31. [PMID: 25956793 PMCID: PMC4494070 DOI: 10.1093/gbe/evv080] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There is widespread interest today in understanding enhancers, which are regulatory elements typically harboring several transcription factor binding sites and mediating the combinatorial effect of transcription factors on gene expression. The evolution of enhancers poses interesting unanswered questions, for example, the evolutionary time taken for a typical enhancer to emerge or the factors shaping its evolution. Existing approaches to cis-regulatory evolution have often ignored the combinatorial nature and varied biochemical mechanisms of gene regulation encoded in enhancers. We report on our investigation of enhancer evolution through the use of PEBCRES, a framework for evolutionary simulation of enhancers that employs a mechanistic and well-supported sequence-to-expression model to assign fitness to the evolving enhancer genotype. We estimated the time necessary to evolve, from genomic background, enhancers capable of driving complex gene expression patterns similar to those involved in early development in Drosophila. We found the time-to-evolve to range between 0.5 and 10 Myr, and to vary greatly with the target expression pattern, complexity of the real enhancer known to encode that pattern, and the strength of input from specific transcription factors. To our knowledge, this is the first estimate of waiting times for realistic enhancers to evolve. The in silico evolved enhancers had, with a few interesting exceptions, site compositions similar to those seen in real enhancers for the same patterns. Our simulations also revealed that certain features of an enhancer might evolve not due to their biological function but as aids to the evolutionary process itself.
Collapse
Affiliation(s)
- Thyago Duque
- Department of Computer Science, University of Illinois at Urbana-Champaign
| | - Saurabh Sinha
- Department of Computer Science, University of Illinois at Urbana-Champaign Institute for Genomic Biology, University of Illinois at Urbana-Champaign
| |
Collapse
|
29
|
Wotton KR, Jiménez-Guri E, Crombach A, Janssens H, Alcaine-Colet A, Lemke S, Schmidt-Ott U, Jaeger J. Quantitative system drift compensates for altered maternal inputs to the gap gene network of the scuttle fly Megaselia abdita. eLife 2015; 4:e04785. [PMID: 25560971 PMCID: PMC4337606 DOI: 10.7554/elife.04785] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/02/2015] [Indexed: 12/20/2022] Open
Abstract
The segmentation gene network in insects can produce equivalent phenotypic outputs despite differences in upstream regulatory inputs between species. We investigate the mechanistic basis of this phenomenon through a systems-level analysis of the gap gene network in the scuttle fly Megaselia abdita (Phoridae). It combines quantification of gene expression at high spatio-temporal resolution with systematic knock-downs by RNA interference (RNAi). Initiation and dynamics of gap gene expression differ markedly between M. abdita and Drosophila melanogaster, while the output of the system converges to equivalent patterns at the end of the blastoderm stage. Although the qualitative structure of the gap gene network is conserved, there are differences in the strength of regulatory interactions between species. We term such network rewiring 'quantitative system drift'. It provides a mechanistic explanation for the developmental hourglass model in the dipteran lineage. Quantitative system drift is likely to be a widespread mechanism for developmental evolution.
Collapse
Affiliation(s)
- Karl R Wotton
- European Molecular Biology Laboratory, CRG Systems Biology Research Unit, Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Eva Jiménez-Guri
- European Molecular Biology Laboratory, CRG Systems Biology Research Unit, Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Anton Crombach
- European Molecular Biology Laboratory, CRG Systems Biology Research Unit, Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Hilde Janssens
- European Molecular Biology Laboratory, CRG Systems Biology Research Unit, Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Anna Alcaine-Colet
- European Molecular Biology Laboratory, CRG Systems Biology Research Unit, Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Steffen Lemke
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, United States
| | - Urs Schmidt-Ott
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, United States
| | - Johannes Jaeger
- European Molecular Biology Laboratory, CRG Systems Biology Research Unit, Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
30
|
Cicin-Sain D, Pulido AH, Crombach A, Wotton KR, Jiménez-Guri E, Taly JF, Roma G, Jaeger J. SuperFly: a comparative database for quantified spatio-temporal gene expression patterns in early dipteran embryos. Nucleic Acids Res 2014; 43:D751-5. [PMID: 25404137 PMCID: PMC4383950 DOI: 10.1093/nar/gku1142] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
We present SuperFly (http://superfly.crg.eu), a relational database for quantified spatio-temporal expression data of segmentation genes during early development in different species of dipteran insects (flies, midges and mosquitoes). SuperFly has a special focus on emerging non-drosophilid model systems. The database currently includes data of high spatio-temporal resolution for three species: the vinegar fly Drosophila melanogaster, the scuttle fly Megaselia abdita and the moth midge Clogmia albipunctata. At this point, SuperFly covers up to 9 genes and 16 time points per species, with a total of 1823 individual embryos. It provides an intuitive web interface, enabling the user to query and access original embryo images, quantified expression profiles, extracted positions of expression boundaries and integrated datasets, plus metadata and intermediate processing steps. SuperFly is a valuable new resource for the quantitative comparative study of gene expression patterns across dipteran species. Moreover, it provides an interesting test set for systems biologists interested in fitting mathematical gene network models to data. Both of these aspects are essential ingredients for progress toward a more quantitative and mechanistic understanding of developmental evolution.
Collapse
Affiliation(s)
- Damjan Cicin-Sain
- EMBL/CRG Research Unit in Systems Biology, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Antonio Hermoso Pulido
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Anton Crombach
- EMBL/CRG Research Unit in Systems Biology, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Karl R Wotton
- EMBL/CRG Research Unit in Systems Biology, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Eva Jiménez-Guri
- EMBL/CRG Research Unit in Systems Biology, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Jean-François Taly
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Guglielmo Roma
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Johannes Jaeger
- EMBL/CRG Research Unit in Systems Biology, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain Bioinformatics Core Facility, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| |
Collapse
|
31
|
Ahuja A, Extavour CG. Patterns of molecular evolution of the germ line specification gene oskar suggest that a novel domain may contribute to functional divergence in Drosophila. Dev Genes Evol 2014; 224:65-77. [PMID: 24407548 DOI: 10.1007/s00427-013-0463-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 12/07/2013] [Indexed: 01/04/2023]
Abstract
In several metazoans including flies of the genus Drosophila, germ line specification occurs through the inheritance of maternally deposited cytoplasmic determinants, collectively called germ plasm. The novel insect gene oskar is at the top of the Drosophila germ line specification pathway, and also plays an important role in posterior patterning. A novel N-terminal domain of oskar (the Long Oskar domain) evolved in Drosophilids, but the role of this domain in oskar functional evolution is unknown. Trans-species transgenesis experiments have shown that oskar orthologs from different Drosophila species have functionally diverged, but the underlying selective pressures and molecular changes have not been investigated. As a first step toward understanding how Oskar function could have evolved, we applied molecular evolution analysis to oskar sequences from the completely sequenced genomes of 16 Drosophila species from the Sophophora subgenus, Drosophila virilis and Drosophila immigrans. We show that overall, this gene is subject to purifying selection, but that individual predicted structural and functional domains are subject to heterogeneous selection pressures. Specifically, two domains, the Drosophila-specific Long Osk domain and the region that interacts with the germ plasm protein Lasp, are evolving at a faster rate than other regions of oskar. Further, we provide evidence that positive selection may have acted on specific sites within these two domains on the D. virilis branch. Our domain-based analysis suggests that changes in the Long Osk and Lasp-binding domains are strong candidates for the molecular basis of functional divergence between the Oskar proteins of D. melanogaster and D. virilis. This molecular evolutionary analysis thus represents an important step towards understanding the role of an evolutionarily and developmentally critical gene in germ plasm evolution and assembly.
Collapse
Affiliation(s)
- Abha Ahuja
- Department of Organismic and Evolutionary Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA, 02138, USA,
| | | |
Collapse
|
32
|
Criscione F, Qi Y, Saunders R, Hall B, Tu Z. A unique Y gene in the Asian malaria mosquito Anopheles stephensi encodes a small lysine-rich protein and is transcribed at the onset of embryonic development. INSECT MOLECULAR BIOLOGY 2013; 22:433-41. [PMID: 23683123 PMCID: PMC3700585 DOI: 10.1111/imb.12034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
In many organisms the Y chromosome initiates sex determination and regulates male fertility and mating behaviour. However, molecular characterization of Y genes is rare outside of a few model species because it is difficult to clone and analyse repeat-rich heterochromatic Y sequences. In insects, Y genes are only well characterized in a small number of Drosophila species. Here we report the discovery of GUY1 (gene unique to the Y), a gene unique to the Y chromosome in the Asian malaria mosquito, Anopheles stephensi, using an approach that compares Illumina sequences separately obtained from male and female genomic DNA. Experimental evidence confirmed that GUY1 is a single copy gene found only on the Y chromosome. GUY1 is transcribed at the very onset of zygotic transcription and encodes a small lysine-rich protein that forms two alpha helices and shows DNA-binding properties. Interestingly, three helix-loop-helix proteins are key factors that determine sex in the early embryo in Drosophila melanogaster. Single embryo analysis indicated that GUY1 is only transcribed in male embryos and that the GUY1 promoter is functional in the early embryos. GUY1 may be used as a paternally inherited molecular marker. Further investigation of GUY1 will contribute to the genetic approaches to control mosquito-borne diseases.
Collapse
Affiliation(s)
- F Criscione
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
| | | | | | | | | |
Collapse
|
33
|
Ten Tusscher KHWJ. Mechanisms and constraints shaping the evolution of body plan segmentation. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2013; 36:54. [PMID: 23708840 DOI: 10.1140/epje/i2013-13054-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 05/07/2013] [Indexed: 06/02/2023]
Abstract
Segmentation of the major body axis into repeating units is arguably one of the major inventions in the evolution of animal body plan pattering. It is found in current day vertebrates, annelids and arthropods. Most segmented animals seem to use a clock-and-wavefront type mechanism in which oscillations emanating from a posterior growth zone become transformed into an anterior posterior sequence of segments. In contrast, few animals such as Drosophila use a complex gene regulatory hierarchy to simultaneously subdivide their entire body axis into segments. Here I discuss how in silico models simulating the evolution of developmental patterning can be used to investigate the forces and constraints that helped shape these two developmental modes. I perform an analysis of a series of previous simulation studies, exploiting the similarities and differences in their outcomes in relation to model characteristics to elucidate the circumstances and constraints likely to have been important for the evolution of sequential and simultaneous segmentation modes. The analysis suggests that constraints arising from the involved growth process and spatial patterning signal--posterior elongation producing a propagating wavefront versus a tissue wide morphogen gradient--and the evolutionary history--ancestral versus derived segmentation mode--strongly shaped both segmentation mechanisms. Furthermore, this implies that these patterning types are to be expected rather than random evolutionary outcomes and supports the likelihood of multiple parallel evolutionary origins.
Collapse
Affiliation(s)
- K H W J Ten Tusscher
- Theoretical Biology and Bioinformactics Group, Utrecht University, Padualaan 8, 3584, CH Utrecht, The Netherlands.
| |
Collapse
|
34
|
Yamamoto DS, Hatakeyama M, Matsuoka H. Artificial activation of mature unfertilized eggs in the malaria vector mosquito, Anopheles stephensi (Diptera, Culicidae). ACTA ACUST UNITED AC 2013; 216:2960-6. [PMID: 23619405 DOI: 10.1242/jeb.084293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the past decade, many transgenic lines of mosquitoes have been generated and analyzed, whereas the maintenance of a large number of transgenic lines requires a great deal of effort and cost. In vitro fertilization by an injection of cryopreserved sperm into eggs has been proven to be effective for the maintenance of strains in mammals. The technique of artificial egg activation is a prerequisite for the establishment of in vitro fertilization by sperm injection. We demonstrated that artificial egg activation is feasible in the malaria vector mosquito, Anopheles stephensi (Diptera, Culicidae). Nearly 100% of eggs dissected from virgin females immersed in distilled water darkened, similar to normally oviposited fertilized eggs. It was revealed by the cytological examination of chromosomes that meiotic arrest was relieved in these eggs approximately 20 min after incubation in water. Biochemical examinations revealed that MAPK (mitogen-activated protein kinase)/ERK (extracellular signal-regulated protein kinase) and MEK (MAPK/ERK kinase) were dephosphorylated similar to that in fertilized eggs. These results indicate that dissected unfertilized eggs were activated in distilled water and started development. Injection of distilled water into body cavity of the virgin blood-fed females also induced activation of a portion of eggs in the ovaries. The technique of artificial egg activation is expected to contribute to the success of in vitro fertilization in A. stephensi.
Collapse
Affiliation(s)
- Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi 329-0498, Japan.
| | | | | |
Collapse
|
35
|
Bickel RD, Cleveland HC, Barkas J, Jeschke CC, Raz AA, Stern DL, Davis GK. The pea aphid uses a version of the terminal system during oviparous, but not viviparous, development. EvoDevo 2013; 4:10. [PMID: 23552511 PMCID: PMC3639227 DOI: 10.1186/2041-9139-4-10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 02/18/2013] [Indexed: 01/03/2023] Open
Abstract
Background In most species of aphid, female nymphs develop into either sexual or asexual adults depending on the length of the photoperiod to which their mothers were exposed. The progeny of these sexual and asexual females, in turn, develop in dramatically different ways. The fertilized oocytes of sexual females begin embryogenesis after being deposited on leaves (oviparous development) while the oocytes of asexual females complete embryogenesis within the mother (viviparous development). Compared with oviparous development, viviparous development involves a smaller transient oocyte surrounded by fewer somatic epithelial cells and a smaller early embryo that comprises fewer cells. To investigate whether patterning mechanisms differ between the earliest stages of the oviparous and viviparous modes of pea aphid development, we examined the expression of pea aphid orthologs of genes known to specify embryonic termini in other insects. Results Here we show that pea aphid oviparous ovaries express torso-like in somatic posterior follicle cells and activate ERK MAP kinase at the posterior of the oocyte. In addition to suggesting that some posterior features of the terminal system are evolutionarily conserved, our detection of activated ERK in the oocyte, rather than in the embryo, suggests that pea aphids may transduce the terminal signal using a mechanism distinct from the one used in Drosophila. In contrast with oviparous development, the pea aphid version of the terminal system does not appear to be used during viviparous development, since we did not detect expression of torso-like in the somatic epithelial cells that surround either the oocyte or the blastoderm embryo and we did not observe restricted activated ERK in the oocyte. Conclusions We suggest that while oviparous oocytes and embryos may specify posterior fate through an aphid terminal system, viviparous oocytes and embryos employ a different mechanism, perhaps one that does not rely on an interaction between the oocyte and surrounding somatic cells. Together, these observations provide a striking example of a difference in the fundamental events of early development that is both environmentally induced and encoded by the same genome.
Collapse
Affiliation(s)
- Ryan D Bickel
- Department of Biology, Bryn Mawr College, Bryn Mawr, PA 19010, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Naggan Perl T, Schmid BG, Schwirz J, Chipman AD. The Evolution of the knirps Family of Transcription Factors in Arthropods. Mol Biol Evol 2013; 30:1348-57. [DOI: 10.1093/molbev/mst046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
37
|
Nguyen C, Andrews E, Le C, Sun L, Annan Z, Clemons A, Severson DW, Duman-Scheel M. Functional genetic characterization of salivary gland development in Aedes aegypti. EvoDevo 2013; 4:9. [PMID: 23497573 PMCID: PMC3599648 DOI: 10.1186/2041-9139-4-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 12/07/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Despite the devastating global impact of mosquito-borne illnesses on human health, very little is known about mosquito developmental biology. In this investigation, functional genetic analysis of embryonic salivary gland development was performed in Aedes aegypti, the dengue and yellow fever vector and an emerging model for vector mosquito development. Although embryonic salivary gland development has been well studied in Drosophila melanogaster, little is known about this process in mosquitoes or other arthropods. RESULTS Mosquitoes possess orthologs of many genes that regulate Drosophila melanogaster embryonic salivary gland development. The expression patterns of a large subset of these genes were assessed during Ae. aegypti development. These studies identified a set of molecular genetic markers for the developing mosquito salivary gland. Analysis of marker expression allowed for tracking of the progression of Ae. aegypti salivary gland development in embryos. In Drosophila, the salivary glands develop from placodes located in the ventral neuroectoderm. However, in Ae. aegypti, salivary marker genes are not expressed in placode-like patterns in the ventral neuroectoderm. Instead, marker gene expression is detected in salivary gland rudiments adjacent to the proventriculus. These observations highlighted the need for functional genetic characterization of mosquito salivary gland development. An siRNA- mediated knockdown strategy was therefore employed to investigate the role of one of the marker genes, cyclic-AMP response element binding protein A (Aae crebA), during Ae. aegypti salivary gland development. These experiments revealed that Aae crebA encodes a key transcriptional regulator of the secretory pathway in the developing Ae. aegypti salivary gland. CONCLUSIONS The results of this investigation indicated that the initiation of salivary gland development in Ae. aegypti significantly differs from that of D. melanogaster. Despite these differences, some elements of salivary gland development, including the ability of CrebA to regulate secretory gene expression, are conserved between the two species. These studies underscore the need for further analysis of mosquito developmental genetics and may foster comparative studies of salivary gland development in additional insect species.
Collapse
Affiliation(s)
- Chilinh Nguyen
- University of Notre Dame, Notre Dame, Eck Institute for Global Health and Department of Biological Sciences, Notre Dame, IN 46556, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Jiménez-Guri E, Huerta-Cepas J, Cozzuto L, Wotton KR, Kang H, Himmelbauer H, Roma G, Gabaldón T, Jaeger J. Comparative transcriptomics of early dipteran development. BMC Genomics 2013; 14:123. [PMID: 23432914 PMCID: PMC3616871 DOI: 10.1186/1471-2164-14-123] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 02/19/2013] [Indexed: 12/24/2022] Open
Abstract
Background Modern sequencing technologies have massively increased the amount of data available for comparative genomics. Whole-transcriptome shotgun sequencing (RNA-seq) provides a powerful basis for comparative studies. In particular, this approach holds great promise for emerging model species in fields such as evolutionary developmental biology (evo-devo). Results We have sequenced early embryonic transcriptomes of two non-drosophilid dipteran species: the moth midge Clogmia albipunctata, and the scuttle fly Megaselia abdita. Our analysis includes a third, published, transcriptome for the hoverfly Episyrphus balteatus. These emerging models for comparative developmental studies close an important phylogenetic gap between Drosophila melanogaster and other insect model systems. In this paper, we provide a comparative analysis of early embryonic transcriptomes across species, and use our data for a phylogenomic re-evaluation of dipteran phylogenetic relationships. Conclusions We show how comparative transcriptomics can be used to create useful resources for evo-devo, and to investigate phylogenetic relationships. Our results demonstrate that de novo assembly of short (Illumina) reads yields high-quality, high-coverage transcriptomic data sets. We use these data to investigate deep dipteran phylogenetic relationships. Our results, based on a concatenation of 160 orthologous genes, provide support for the traditional view of Clogmia being the sister group of Brachycera (Megaselia, Episyrphus, Drosophila), rather than that of Culicomorpha (which includes mosquitoes and blackflies).
Collapse
Affiliation(s)
- Eva Jiménez-Guri
- EMBL/CRG Research Unit in Systems Biology, Centre de Regulació Genòmica (CRG), and Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Duncan EJ, Benton MA, Dearden PK. Canonical terminal patterning is an evolutionary novelty. Dev Biol 2013; 377:245-61. [PMID: 23438815 DOI: 10.1016/j.ydbio.2013.02.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/08/2013] [Accepted: 02/14/2013] [Indexed: 10/27/2022]
Abstract
Patterning of the terminal regions of the Drosophila embryo is achieved by an exquisitely regulated signal that passes between the follicle cells of the ovary, and the developing embryo. This pathway, however, is missing or modified in other insects. Here we trace the evolution of this pathway by examining the origins and expression of its components. The three core components of this pathway: trunk, torso and torso-like have different evolutionary histories and have been assembled step-wise to form the canonical terminal patterning pathway of Drosophila and Tribolium. Trunk, torso and a gene unrelated to terminal patterning, prothoraciotrophic hormone (PTTH), show an intimately linked evolutionary history, with every holometabolous insect, except the honeybee, possessing both PTTH and torso genes. Trunk is more restricted in its phylogenetic distribution, present only in the Diptera and Tribolium and, surprisingly, in the chelicerate Ixodes scapularis, raising the possibility that trunk and torso evolved earlier than previously thought. In Drosophila torso-like restricts the activation of the terminal patterning pathway to the poles of the embryo. Torso-like evolved in the pan-crustacean lineage, but based on expression of components of the canonical terminal patterning system in the hemimetabolous insect Acyrthosiphon pisum and the holometabolous insect Apis mellifera, we find that the canonical terminal-patterning system is not active in these insects. We therefore propose that the ancestral function of torso-like is unrelated to terminal patterning and that torso-like has become co-opted into terminal patterning in the lineage leading to Coleoptera and Diptera. We also show that this co-option has not resulted in changes to the molecular function of this protein. Torso-like from the pea aphid, honeybee and Drosophila, despite being expressed in different patterns, are functionally equivalent. We propose that co-option of torso-like into restricting the activity of trunk and torso facilitated the final step in the evolution of this pathway; the capture of transcriptional control of target genes such as tailless and huckebein by this complex and novel patterning pathway.
Collapse
Affiliation(s)
- Elizabeth J Duncan
- Laboratory for Evolution and Development, Genetics Otago, Gravida, National Centre for Growth and Development, Department of Biochemistry, University of Otago, P.O. Box 56, Dunedin, Aotearoa, New Zealand.
| | | | | |
Collapse
|
40
|
Juhn J, James AA. Hybridization in situ of salivary glands, ovaries, and embryos of vector mosquitoes. J Vis Exp 2012:3709. [PMID: 22781778 PMCID: PMC3476388 DOI: 10.3791/3709] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Mosquitoes are vectors for a diverse set of pathogens including arboviruses, protozoan parasites and nematodes. Investigation of transcripts and gene regulators that are expressed in tissues in which the mosquito host and pathogen interact, and in organs involved in reproduction are of great interest for strategies to reduce mosquito-borne disease transmission and disrupt egg development. A number of tools have been employed to study and validate the temporal and tissue-specific regulation of gene expression. Here, we describe protocols that have been developed to obtain spatial information, which enhances our understanding of where specific genes are expressed and their products accumulate. The protocol described has been used to validate expression and determine accumulation patterns of transcripts in tissues related to mosquito-borne pathogen transmission, such as female salivary glands, as well as subcellular compartments of ovaries and embryos, which relate to mosquito reproduction and development. The following procedures represent an optimized methodology that improves the efficiency of various steps in the protocol without loss of target-specific hybridization signals. Guidelines for RNA probe preparation, dissection of soft tissues and the general procedure for fixation and hybridization are described in Part A, while steps specific for the collection, fixation, pre-hybridization and hybridization of mosquito embryos are detailed in Part B.
Collapse
Affiliation(s)
- Jennifer Juhn
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
| | | |
Collapse
|
41
|
Expression of pair rule gene orthologs in the blastoderm of a myriapod: evidence for pair rule-like mechanisms? BMC DEVELOPMENTAL BIOLOGY 2012; 12:15. [PMID: 22595029 PMCID: PMC3477074 DOI: 10.1186/1471-213x-12-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 04/11/2012] [Indexed: 01/14/2023]
Abstract
Background A hallmark of Drosophila segmentation is the stepwise subdivision of the body into smaller and smaller units, and finally into the segments. This is achieved by the function of the well-understood segmentation gene cascade. The first molecular sign of a segmented body appears with the action of the pair rule genes, which are expressed as transversal stripes in alternating segments. Drosophila development, however, is derived, and in most other arthropods only the anterior body is patterned (almost) simultaneously from a pre-existing field of cells; posterior segments are added sequentially from a posterior segment addition zone. A long-standing question is to what extent segmentation mechanisms known from Drosophila may be conserved in short-germ arthropods. Despite the derived developmental modes, it appears more likely that conserved mechanisms can be found in anterior patterning. Results Expression analysis of pair rule gene orthologs in the blastoderm of the pill millipede Glomeris marginata (Myriapoda: Diplopoda) suggests that these genes are generally involved in segmenting the anterior embryo. We find that the Glomeris pairberry-1 ( pby-1) gene is expressed in a pair rule pattern that is also found in insects and a chelicerate, the mite Tetraynchus urticae. Other Glomeris pair rule gene orthologs are expressed in double segment wide domains in the blastoderm, which at subsequent stages split into two stripes in adjacent segments. Conclusions The expression patterns of the millipede pair rule gene orthologs resemble pair rule patterning in Drosophila and other insects, and thus represent evidence for the presence of an ancestral pair rule-like mechanism in myriapods. We discuss the possibilities that blastoderm patterning may be conserved in long-germ and short-germ arthropods, and that a posterior double segmental mechanism may be present in short-germ arthropods.
Collapse
|
42
|
Schmitt-Engel C, Cerny AC, Schoppmeier M. A dual role for nanos and pumilio in anterior and posterior blastodermal patterning of the short-germ beetle Tribolium castaneum. Dev Biol 2012; 364:224-35. [DOI: 10.1016/j.ydbio.2012.01.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 01/09/2012] [Accepted: 01/20/2012] [Indexed: 11/15/2022]
|
43
|
Andrioli LP. Toward new Drosophila paradigms. Genesis 2012; 50:585-98. [DOI: 10.1002/dvg.22019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 02/07/2012] [Accepted: 02/08/2012] [Indexed: 11/07/2022]
|
44
|
Control of RNP motility and localization by a splicing-dependent structure in oskar mRNA. Nat Struct Mol Biol 2012; 19:441-9. [PMID: 22426546 DOI: 10.1038/nsmb.2257] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 01/07/2012] [Indexed: 11/08/2022]
Abstract
oskar RNA localization to the posterior pole of the Drosophila melanogaster oocyte requires splicing of the first intron and the exon junction complex (EJC) core proteins. The functional link between splicing, EJC deposition and oskar localization has been unclear. Here we demonstrate that the EJC associates with oskar mRNA upon splicing in vitro and that Drosophila EJC deposition is constitutive and conserved. Our in vivo analysis reveals that splicing creates the spliced oskar localization element (SOLE), whose structural integrity is crucial for ribonucleoprotein motility and localization in the oocyte. Splicing thus has a dual role in oskar mRNA localization: assembling the SOLE and depositing the EJC required for mRNA transport. The SOLE complements the EJC in formation of a functional unit that, together with the oskar 3' UTR, maintains proper kinesin-based motility of oskar mRNPs and posterior mRNA targeting.
Collapse
|
45
|
Jaeger J, Crombach A. Life's attractors : understanding developmental systems through reverse engineering and in silico evolution. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 751:93-119. [PMID: 22821455 DOI: 10.1007/978-1-4614-3567-9_5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We propose an approach to evolutionary systems biology which is based on reverse engineering of gene regulatory networks and in silico evolutionary simulations. We infer regulatory parameters for gene networks by fitting computational models to quantitative expression data. This allows us to characterize the regulatory structure and dynamical repertoire of evolving gene regulatory networks with a reasonable amount of experimental and computational effort. We use the resulting network models to identify those regulatory interactions that are conserved, and those that have diverged between different species. Moreover, we use the models obtained by data fitting as starting points for simulations of evolutionary transitions between species. These simulations enable us to investigate whether such transitions are random, or whether they show stereotypical series of regulatory changes which depend on the structure and dynamical repertoire of an evolving network. Finally, we present a case study-the gap gene network in dipterans (flies, midges, and mosquitoes)-to illustrate the practical application of the proposed methodology, and to highlight the kind of biological insights that can be gained by this approach.
Collapse
|
46
|
A conserved developmental patterning network produces quantitatively different output in multiple species of Drosophila. PLoS Genet 2011; 7:e1002346. [PMID: 22046143 PMCID: PMC3203197 DOI: 10.1371/journal.pgen.1002346] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 08/27/2011] [Indexed: 11/18/2022] Open
Abstract
Differences in the level, timing, or location of gene expression can contribute to alternative phenotypes at the molecular and organismal level. Understanding the origins of expression differences is complicated by the fact that organismal morphology and gene regulatory networks could potentially vary even between closely related species. To assess the scope of such changes, we used high-resolution imaging methods to measure mRNA expression in blastoderm embryos of Drosophila yakuba and Drosophila pseudoobscura and assembled these data into cellular resolution atlases, where expression levels for 13 genes in the segmentation network are averaged into species-specific, cellular resolution morphological frameworks. We demonstrate that the blastoderm embryos of these species differ in their morphology in terms of size, shape, and number of nuclei. We present an approach to compare cellular gene expression patterns between species, while accounting for varying embryo morphology, and apply it to our data and an equivalent dataset for Drosophila melanogaster. Our analysis reveals that all individual genes differ quantitatively in their spatio-temporal expression patterns between these species, primarily in terms of their relative position and dynamics. Despite many small quantitative differences, cellular gene expression profiles for the whole set of genes examined are largely similar. This suggests that cell types at this stage of development are conserved, though they can differ in their relative position by up to 3–4 cell widths and in their relative proportion between species by as much as 5-fold. Quantitative differences in the dynamics and relative level of a subset of genes between corresponding cell types may reflect altered regulatory functions between species. Our results emphasize that transcriptional networks can diverge over short evolutionary timescales and that even small changes can lead to distinct output in terms of the placement and number of equivalent cells. For a gene to function properly, it must be active in the right place, at the right time, and in the right amount. Changes in any of these features can lead to observable differences between individuals and species and in some cases can lead to disease. We do not currently understand how the position, timing, and amount of gene expression is encoded in DNA sequence. One approach to this problem is to compare how gene expression differs between species and to try to relate changes in DNA sequence to changes in gene expression. Here, we take the first step by comparing gene expression patterns at high spatial and temporal resolution between embryos of three species of fruit flies. We develop methods for comparing gene expression in individual cells, which allow us to control for variation in the size, shape, and number of nuclei between embryos. We find measurable quantitative differences in the patterns for all individual genes that we have examined. However, by considering all genes in our dataset at once, we show that many genes are changing together, leading to largely equivalent types of cells in these three species.
Collapse
|
47
|
Abstract
The nuclear receptors (NRs) of metazoans are an ancient family of transcription factors defined by conserved DNA- and ligand-binding domains (DBDs and LBDs, respectively). The Drosophila melanogaster genome project revealed 18 canonical NRs (with DBDs and LBDs both present) and 3 receptors with the DBD only. Annotation of subsequently sequenced insect genomes revealed only minor deviations from this pattern. A renewed focus on functional analysis of the isoforms of insect NRs is therefore required to understand the diverse roles of these transcription factors in embryogenesis, metamorphosis, reproduction, and homeostasis. One insect NR, ecdysone receptor (EcR), functions as a receptor for the ecdysteroid molting hormones of insects. Researchers have developed nonsteroidal ecdysteroid agonists for EcR that disrupt molting and can be used as safe pesticides. An exciting new technology allows EcR to be used in chimeric, ligand-inducible gene-switch systems with applications in pest management and medicine.
Collapse
Affiliation(s)
- Susan E Fahrbach
- Department of Biology, Wake Forest University, Winston-Salem, North Carolina 27109, USA.
| | | | | |
Collapse
|
48
|
Janssen R, Budd GE, Damen WG. Gene expression suggests conserved mechanisms patterning the heads of insects and myriapods. Dev Biol 2011; 357:64-72. [DOI: 10.1016/j.ydbio.2011.05.670] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 05/20/2011] [Accepted: 05/25/2011] [Indexed: 01/31/2023]
|
49
|
Behura SK, Haugen M, Flannery E, Sarro J, Tessier CR, Severson DW, Duman-Scheel M. Comparative genomic analysis of Drosophila melanogaster and vector mosquito developmental genes. PLoS One 2011; 6:e21504. [PMID: 21754989 PMCID: PMC3130749 DOI: 10.1371/journal.pone.0021504] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/30/2011] [Indexed: 11/18/2022] Open
Abstract
Genome sequencing projects have presented the opportunity for analysis of developmental genes in three vector mosquito species: Aedes aegypti, Culex quinquefasciatus, and Anopheles gambiae. A comparative genomic analysis of developmental genes in Drosophila melanogaster and these three important vectors of human disease was performed in this investigation. While the study was comprehensive, special emphasis centered on genes that 1) are components of developmental signaling pathways, 2) regulate fundamental developmental processes, 3) are critical for the development of tissues of vector importance, 4) function in developmental processes known to have diverged within insects, and 5) encode microRNAs (miRNAs) that regulate developmental transcripts in Drosophila. While most fruit fly developmental genes are conserved in the three vector mosquito species, several genes known to be critical for Drosophila development were not identified in one or more mosquito genomes. In other cases, mosquito lineage-specific gene gains with respect to D. melanogaster were noted. Sequence analyses also revealed that numerous repetitive sequences are a common structural feature of Drosophila and mosquito developmental genes. Finally, analysis of predicted miRNA binding sites in fruit fly and mosquito developmental genes suggests that the repertoire of developmental genes targeted by miRNAs is species-specific. The results of this study provide insight into the evolution of developmental genes and processes in dipterans and other arthropods, serve as a resource for those pursuing analysis of mosquito development, and will promote the design and refinement of functional analysis experiments.
Collapse
Affiliation(s)
- Susanta K. Behura
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Morgan Haugen
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
| | - Ellen Flannery
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Joseph Sarro
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Charles R. Tessier
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
| | - David W. Severson
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
| | - Molly Duman-Scheel
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
- * E-mail:
| |
Collapse
|
50
|
Haugen M, Flannery E, Tomchaney M, Mori A, Behura SK, Severson DW, Duman-Scheel M. Semaphorin-1a is required for Aedes aegypti embryonic nerve cord development. PLoS One 2011; 6:e21694. [PMID: 21738767 PMCID: PMC3124551 DOI: 10.1371/journal.pone.0021694] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 06/08/2011] [Indexed: 12/30/2022] Open
Abstract
Although mosquito genome projects have uncovered orthologues of many known developmental regulatory genes, extremely little is known about mosquito development. In this study, the role of semaphorin-1a (sema1a) was investigated during vector mosquito embryonic ventral nerve cord development. Expression of sema1a and the plexin A (plexA) receptor are detected in the embryonic ventral nerve cords of Aedes aegypti (dengue vector) and Anopheles gambiae (malaria vector), suggesting that Sema1a signaling may regulate mosquito nervous system development. Analysis of sema1a function was investigated through siRNA-mediated knockdown in A. aegypti embryos. Knockdown of sema1a during A. aegypti development results in a number of nerve cord phenotypes, including thinning, breakage, and occasional fusion of the longitudinal connectives, thin or absent commissures, and general distortion of the nerve cord. Although analysis of Drosophila melanogaster sema1a loss-of-function mutants uncovered many similar phenotypes, aspects of the longitudinal phenotypes differed between D. melanogaster and A. aegypti. The results of this investigation suggest that Sema1a is required for development of the insect ventral nerve cord, but that the developmental roles of this guidance molecule have diverged in dipteran insects.
Collapse
Affiliation(s)
- Morgan Haugen
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
| | - Ellen Flannery
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Michael Tomchaney
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Akio Mori
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Susanta K. Behura
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - David W. Severson
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Molly Duman-Scheel
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, South Bend, Indiana, United States of America
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
- * E-mail:
| |
Collapse
|