Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Werner Boll is active.

Publication


Featured researches published by Werner Boll.


Development | 2002

The Drosophila Pox neuro gene: control of male courtship behavior and fertility as revealed by a complete dissection of all enhancers.

Werner Boll; Markus Noll

We have dissected the entire cis-regulatory region of the Drosophila Pox neuro gene with regard to its enhancers, and have analyzed their functions by the selective addition to Pox neuro null mutant flies of one or several functions, each regulated by a complete or partial enhancer. We have identified at least 15 enhancers with an astounding complexity in arrangement and substructure that regulate Pox neuro functions required for the development of the peripheral and central nervous system and of most appendages. Many of these functions are essential for normal male courtship behavior and fertility. Two enhancers regulate the development of the penis, claspers and posterior lobes of male genitalia. Three enhancers, two of which overlap, control the development of chemosensory bristles in the labellum, legs and wings, some or all of which are required for the transmission of gustatory signals elicited by female pheromones. An additional enhancer regulates in the developing brain the connectivity of two specific neuronal clusters entrusted with processing olfactory pheromone signals from the antennal nerve. Finally, functions crucial for the ability of the male to copulate depend on an enhancer that activates Pox neuro expression in the embryonic ventral cord. In addition to these male courtship and fertility functions of Pox neuro, we have identified enhancers that regulate: (1) proper segmentation of tarsal segments in the leg disc and in homologous segments of the antennal disc; and (2) proper development of the wing hinge and hence the ability of the fly to fly.


PLOS ONE | 2009

Sensory integration regulating male courtship behavior in Drosophila.

Dimitrije Krstic; Werner Boll; Markus Noll

The courtship behavior of Drosophila melanogaster serves as an excellent model system to study how complex innate behaviors are controlled by the nervous system. To understand how the underlying neural network controls this behavior, it is not sufficient to unravel its architecture, but also crucial to decipher its logic. By systematic analysis of how variations in sensory inputs alter the courtship behavior of a naïve male in the single-choice courtship paradigm, we derive a model describing the logic of the network that integrates the various sensory stimuli and elicits this complex innate behavior. This approach and the model derived from it distinguish (i) between initiation and maintenance of courtship, (ii) between courtship in daylight and in the dark, where the male uses a scanning strategy to retrieve the decamping female, and (iii) between courtship towards receptive virgin females and mature males. The last distinction demonstrates that sexual orientation of the courting male, in the absence of discriminatory visual cues, depends on the integration of gustatory and behavioral feedback inputs, but not on olfactory signals from the courted animal. The model will complement studies on the connectivity and intrinsic properties of the neurons forming the circuitry that regulates male courtship behavior.


Developmental Biology | 2010

Origin of Pax and Six gene families in sponges: Single PaxB and Six1/2 orthologs in Chalinula loosanoffi.

April Hill; Werner Boll; Carolin Ries; Lisa Warner; Marisa Osswalt; Malcolm Hill; Markus Noll

Pax genes play an important role in networks of transcription factors that determine organogenesis, notably the development of sensory organs. Other members of this regulatory network include transcription factors encoded by the Six gene family. Sponges lack organs and a nervous system, possibly because they have not evolved a Pax/Six network. Here we show that the demosponge Chalinula loosanoffi encodes only one Pax and one Six gene, representatives of the PaxB and Six1/2 subfamilies. Analysis of their temporal transcription patterns during development shows no correlation of their mRNA levels while their spatial patterns show some overlap of expression in adult tissue, although cellular resolution was not achieved. These results do not suggest that these genes form a major network in this basal phylum, although its existence in a minor fraction of cells is not excluded. We further show that sponge PaxB can substitute for some of the Pax2, but not of the Pax6 functions in Drosophila. Finally, we have analyzed the phylogeny of Pax and Six genes and have derived a model of the evolution of the Pax gene subfamilies in metazoans. It illustrates a diversification of Pax genes into subfamilies mostly in triploblasts before the protostome-deuterostome split, whereas few subfamilies were lost in various phyla after the Cambrian explosion.


Developmental Cell | 2015

Co-option of an Ancestral Hox-Regulated Network Underlies a Recently Evolved Morphological Novelty

William J. Glassford; Winslow C. Johnson; Natalie R. Dall; Sarah Smith; Yang Liu; Werner Boll; Markus Noll; Mark Rebeiz

The evolutionary origins of complex morphological structures such as the vertebrate eye or insect wing remain one of the greatest mysteries of biology. Recent comparative studies of gene expression imply that new structures are not built from scratch, but rather form by co-opting preexisting gene networks. A key prediction of this model is that upstream factors within the network will activate their preexisting targets (i.e., enhancers) to form novel anatomies. Here, we show how a recently derived morphological novelty present in the genitalia of D. melanogaster employs an ancestral Hox-regulated network deployed in the embryo to generate the larval posterior spiracle. We demonstrate how transcriptional enhancers and constituent transcription factor binding sites are used in both ancestral and novel contexts. These results illustrate network co-option at the level of individual connections between regulatory genes and highlight how morphological novelty may originate through the co-option of networks controlling seemingly unrelated structures.


PLOS ONE | 2013

Influence of the White Locus on the Courtship Behavior of Drosophila Males

Dimitrije Krstic; Werner Boll; Markus Noll

Since its discovery by Morgan, the Drosophila white gene has become one of the most intensely studied genes and has been widely used as a genetic marker. Earlier reports that over- and misexpression of White protein in Drosophila males leads to male-male courtship implicated white in courtship control. While previous studies suggested that it is the mislocalization of White protein within cells that causes the courtship phenotype, we demonstrate here that also the lack of extra-retinal White can cause very similar behavioral changes. Moreover, we provide evidence that the lack of White function increases the sexual arousal of males in general, of which the enhanced male-male courtship might be an indirect effect. We further show that white mutant flies are not only optomotor blind but also dazzled by the over-flow of light in daylight. Implications of these findings for the proper interpretation of behavioral studies with white mutant flies are discussed.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Positive and negative gustatory inputs affect Drosophila lifespan partly in parallel to dFOXO signaling

Ivan Ostojic; Werner Boll; Michael J. Waterson; Tammy Chan; Rashmi Chandra; Scott D. Pletcher; Joy Alcedo

Significance The ability of the worm Caenorhabditis elegans to taste or smell can influence its lifespan, and the effect of odors on lifespan has also been shown to exist in the fruit fly Drosophila. Now we provide evidence that fly lifespan is also affected by its ability to taste: some taste inputs shorten lifespan, whereas others increase it. In flies, the lifespan-shortening taste inputs act via an insulin-like signaling pathway and its downstream transcription factor dFOXO, whereas the lifespan-lengthening taste inputs can act independently of this pathway. The taste influence on lifespan is also unlikely linked to changes in food intake levels. Thus, different taste inputs will affect lifespan through more than one mechanism. In Caenorhabditis elegans, a subset of gustatory neurons, as well as olfactory neurons, shortens lifespan, whereas a different subset of gustatory neurons lengthens it. Recently, the lifespan-shortening effect of olfactory neurons has been reported to be conserved in Drosophila. Here we show that the Drosophila gustatory system also affects lifespan in a bidirectional manner. We find that taste inputs shorten lifespan through inhibition of the insulin pathway effector dFOXO, whereas other taste inputs lengthen lifespan in parallel to this pathway. We also note that the gustatory influence on lifespan does not necessarily depend on food intake levels. Finally, we identify the nature of some of the taste inputs that could shorten versus lengthen lifespan. Together our data suggest that different gustatory cues can modulate the activities of distinct signaling pathways, including different insulin-like peptides, to promote physiological changes that ultimately affect lifespan.


Gene | 1986

A new approach to high sensitivity differential hybridization

Werner Boll; Jun-ichi Fujisawa; Jarmo Niemi; Charles Weissmann

We describe a new approach to differential hybridization, designed to identify cDNA clones representing rare mRNA species. Duplicate filters carrying a library of cDNA from phorbolmyristate acetate (PMA)-induced EL-4 cells in lambda gt11 were hybridized with high concentrations of unlabeled, cloned, single-stranded cDNA from induced and control EL-4 cells, respectively. Plaques binding single-stranded cDNA were revealed by a second round of hybridization with 35S-labeled DNA complementary to the vector moiety of the single-stranded cDNA. Plaques corresponding to PMA-induced mRNAs occurring at a level of about 1 part in 15,000 were isolated. We believe the method is at least ten times more sensitive than conventional differential hybridization.


From Gene to Protein: Information Transfer in Normal and Abnormal Cells | 1979

EXPRESSION OF CLONED VIRAL AND CHROMOSOMAL PLASMID-LINKED DNA IN COGNATE HOST CELLS

Charles Weissmann; Ned Mantei; Werner Boll; Robert F. Weaver; Neil Wilkie; Barklie Clements; Tadatsugu Taniguchi; Albert van Ooyen; Johan Van Den Berg; Michael Fried; Kenneth Murray

Publisher Summary This chapter focuses on expression of cloned viral and chromosomal plasmid-linked DNA in cognate host cells. The possible hazards inherent to hybrid DNA technology are usually speculated. An important question in this regard is whether a viral genome linked to a prokaryotic vector can be expressed in a host cell. This is shown to be the case both for prokaryotic viral DNA in a prokaryotic host and for eukaryotic viral DNA in a eukaryotic host. It is found that an intact copy of Qβ DNA joined to pCRI elicits formation of Qβ in E. coli, a hybrid consisting of a head-to-tail polyoma DNA dimer and pBR322 causes polyoma formation in cultured mouse fibroblasts, and the cloned thymidine kinase (TK) gene of herpes simplex type I linked to pBR322 can transform TK− mouse L cells. The chapter discusses the properties of E. coli HB101 containing Qβ DNA plasmids. It also presents physical and biological characterization of a hybrid plasmid containing Qβ DNA and discusses the characterization of the Qβ RNA formed following infection with Qβ DNA-containing plasmids.


PLOS ONE | 2017

Crucial roles of Pox neuro in the developing ellipsoid body and antennal lobes of the Drosophila brain

Shilpi Minocha; Werner Boll; Markus Noll

The paired box gene Pox neuro (Poxn) is expressed in two bilaterally symmetric neuronal clusters of the developing adult Drosophila brain, a protocerebral dorsal cluster (DC) and a deutocerebral ventral cluster (VC). We show that all cells that express Poxn in the developing brain are postmitotic neurons. During embryogenesis, the DC and VC consist of only 20 and 12 neurons that express Poxn, designated embryonic Poxn-neurons. The number of Poxn-neurons increases only during the third larval instar, when the DC and VC increase dramatically to about 242 and 109 Poxn-neurons, respectively, virtually all of which survive to the adult stage, while no new Poxn-neurons are added during metamorphosis. Although the vast majority of Poxn-neurons express Poxn only during third instar, about half of them are born by the end of embryogenesis, as demonstrated by the absence of BrdU incorporation during larval stages. At late third instar, embryonic Poxn-neurons, which begin to express Poxn during embryogenesis, can be easily distinguished from embryonic-born and larval-born Poxn-neurons, which begin to express Poxn only during third instar, (i) by the absence of Pros, (ii) their overt differentiation of axons and neurites, and (iii) the strikingly larger diameter of their cell bodies still apparent in the adult brain. The embryonic Poxn-neurons are primary neurons that lay out the pioneering tracts for the secondary Poxn-neurons, which differentiate projections and axons that follow those of the primary neurons during metamorphosis. The DC and the VC participate only in two neuropils of the adult brain. The DC forms most, if not all, of the neurons that connect the bulb (lateral triangle) with the ellipsoid body, a prominent neuropil of the central complex, while the VC forms most of the ventral projection neurons of the antennal lobe, which connect it ipsilaterally to the lateral horn, bypassing the mushroom bodies. In addition, Poxn-neurons of the VC are ventral local interneurons of the antennal lobe. In the absence of Poxn protein in the developing brain, embryonic Poxn-neurons stall their projections and cannot find their proper target neuropils, the bulb and ellipsoid body in the case of the DC, or the antennal lobe and lateral horn in the case of the VC, whereby the absence of the ellipsoid body neuropil is particularly striking. Poxn is thus crucial for pathfinding both in the DC and VC. Additional implications of our results are discussed.


Developmental Biology | 2015

Pox neuro control of cell lineages that give rise to larval poly-innervated external sensory organs in Drosophila

Yanrui Jiang; Werner Boll; Markus Noll

The Pox neuro (Poxn) gene of Drosophila plays a crucial role in the development of poly-innervated external sensory (p-es) organs. However, how Poxn exerts this role has remained elusive. In this study, we have analyzed the cell lineages of all larval p-es organs, namely of the kölbchen, papilla 6, and hair 3. Surprisingly, these lineages are distinct from any previously reported cell lineages of sensory organs. Unlike the well-established lineage of mono-innervated external sensory (m-es) organs and a previously proposed model of the p-es lineage, we demonstrate that all wild-type p-es lineages exhibit the following features: the secondary precursor, pIIa, gives rise to all three support cells-socket, shaft, and sheath, whereas the other secondary precursor, pIIb, is neuronal and gives rise to all neurons. We further show that in one of the p-es lineages, that of papilla 6, one cell undergoes apoptosis. By contrast in Poxn null mutants, all p-es lineages have a reduced number of cells and their pattern of cell divisions is changed to that of an m-es organ, with the exception of a lineage in a minority of mutant kölbchen that retains a second bipolar neuron. Indeed, the role of Poxn in p-es lineages is consistent with the specification of the developmental potential of secondary precursors and the regulation of cell division but not apoptosis.

Collaboration


Dive into the Werner Boll's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A Hall

University of Zurich

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge