Network


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

Hotspot


Dive into the research topics where Shireen A. Davies is active.

Publication


Featured researches published by Shireen A. Davies.


Genome Biology | 2004

Function-informed transcriptome analysis of Drosophila renal tubule

Jing-jing Wang; Laura Kean; Jingli Yang; Adrian K. Allan; Shireen A. Davies; Pawel Herzyk; Julian A. T. Dow

BackgroundComprehensive, tissue-specific, microarray analysis is a potent tool for the identification of tightly defined expression patterns that might be missed in whole-organism scans. We applied such an analysis to Drosophila melanogaster Malpighian (renal) tubule, a defined differentiated tissue.ResultsThe transcriptome of the D. melanogaster Malpighian tubule is highly reproducible and significantly different from that obtained from whole-organism arrays. More than 200 genes are more than 10-fold enriched and over 1,000 are significantly enriched. Of the top 200 genes, only 18 have previously been named, and only 45% have even estimates of function. In addition, 30 transcription factors, not previously implicated in tubule development, are shown to be enriched in adult tubule, and their expression patterns respect precisely the domains and cell types previously identified by enhancer trapping. Of Drosophila genes with close human disease homologs, 50 are enriched threefold or more, and eight enriched 10-fold or more, in tubule. Intriguingly, several of these diseases have human renal phenotypes, implying close conservation of renal function across 400 million years of divergent evolution.ConclusionsFrom those genes that are identifiable, a radically new view of the function of the tubule, emphasizing solute transport rather than fluid secretion, can be obtained. The results illustrate the phenotype gap: historically, the effort expended on a model organism has tended to concentrate on a relatively small set of processes, rather than on the spread of genes in the genome.


Insect Biochemistry and Molecular Biology | 2000

Nitric oxide signalling in insects.

Shireen A. Davies

The gas, nitric oxide (NO), was first described as a second messenger molecule at least a decade ago; since then, research into mechanisms of intracellular generation of NO and of the functional role of the NO signalling pathway has generated thousands of research papers, many reviews and a Nobel Prize. This reflects the fundamental importance of NO and its signalling processes in biology and medicine. The family of enzymes which form NO froml-arginine are known as nitric oxide synthases (NOS). These are large complex haem-containing enzymes which contain binding sites for the co-factors flavin adenine dinucleotide (FAD), flavin mononucleotide (FMN) and nicotinamide adenine dinucleotide phosphate (NADP), (Marletta, 1989; Bredt et al., 1991; Mayer, 1994), and also contain the co-factor tetrahydrobiopterin (Mayer, 1994). Recent work has shown that the NOS complex consists of an oxygenase (catalytic) domain and a reductase domain. The catalytic domain, which is dimerised in biologically active NOS (Stuehr, 1999), contains the haem group and tetrahydrobiopterin, and binds the substrate, l-arginine (see Fig. 1). Ca2+/calmodulin binds at a regulatory site between the domains to aid the process of electron transfer between them (Crane et al., 1999). NO generation occurs in the presence of NADPH in a complex two-stage reaction (via hydroxyarginine) which results in oxidation of the guanadino nitrogen group of arginine, to form citrulline (Fig. 2). NOS enzymes are encoded by a large multigene family, and occur as several isoforms. So far, three genes have been identified in vertebrates and humans, NOS1, NOS2andNOS3. The conservation of exon/intron structure across humanNOS1, NOS2and NOS3genes sug-


Journal of Biological Chemistry | 1996

Analysis and inactivation of vha55, the gene encoding the vacuolar ATPase B-subunit in Drosophila melanogaster reveals a larval lethal phenotype.

Shireen A. Davies; Stephen F. Goodwin; D. C. Kelly; Z. Wang; M. A. Sozen; Kim Kaiser; Julian A. T. Dow

Vacuolar ATPases play major roles in endomembrane and plasma membrane proton transport in eukaryotes. A Drosophila melanogaster cDNA encoding vha55, the 55-kDa vacuolar ATPase (V-ATPase) regulatory B-subunit, was characterized and mapped to 87C2-4 on chromosome 3R. A fly line was identified that carried a single lethal P-element insertion within the coding portion of gene, and its LacZ reporter gene revealed elevated expression in Malpighian tubules, rectum, antennal palps, and oviduct, regions where V-ATPases are believed to play a plasma membrane, rather than an endomembrane, role. The P-element vha55 insertion was shown to be allelic to a known lethal complementation group l(3)SzA (= l(3)87Ca) at 87C, for which many alleles have been described previously. Deletions of the locus have been shown to be larval lethal, whereas point mutations show a range of phenotypes from subvital to embryonic lethal, implying that severe alleles confer a partial dominant negative phenotype. The P-element null allele of vha55 was shown also to suppress ectopic sex combs in Polycomb males, suggesting that transcriptional silencing may be modulated by genes other than those with known homeotic or DNA binding functions.


Advances in Insect Physiology | 2001

The Drosophila melanogaster Malpighian tubule.

Julian A. T. Dow; Shireen A. Davies

Publisher Summary This chapter reviews the morphology, function, and development of malpighian tubule of Drosophila melanogaster . Recent results have provided a reference point for tubule studies for insect physiologists and a unique resource for developmental and genetic analysis of fundamental questions of differentiated cell function. It is noted that the formation of the Drosophila melanogaster malpighian tubule is better understood than that of any other insect and it provides a model for epithelial development in general. In addition to the standard Drosophila techniques of close microscopic examination, genetic screens, enhancer trapping, and reporter gene expression, there are some specific tools that have made progress much easier.


The Journal of Experimental Biology | 2004

Conservation of capa peptide-induced nitric oxide signalling in Diptera

Valerie P. Pollock; James McGettigan; Pablo Cabrero; Ian M. Maudlin; Julian A. T. Dow; Shireen A. Davies

SUMMARY In D. melanogaster Malpighian (renal) tubules, the capa peptides stimulate production of nitric oxide (NO) and guanosine 3′, 5′-cyclic monophosphate (cGMP), resulting in increased fluid transport. The roles of NO synthase (NOS), NO and cGMP in capa peptide signalling were tested in several other insect species of medical relevance within the Diptera (Aedes aegypti, Anopheles stephensi and Glossina morsitans) and in one orthopteran out-group, Schistocerca gregaria. NOS immunoreactivity was detectable by immunocytochemistry in tubules from all species studied. D. melanogaster, A. aegypti and A. stephensi express NOS in only principal cells, whereas G. morsitans and S. gregaria show more general NOS expression in the tubule. Measurement of associated NOS activity (NADPH diaphorase) shows that both D. melanogaster capa-1 and the two capa peptides encoded in the A. gambiae genome, QGLVPFPRVamide (AngCAPA-QGL) and GPTVGLFAFPRVamide (AngCAPA-GPT), all stimulate NOS activity in D. melanogaster, A. aegypti, A. stephensi and G. morsitans tubules but not in S. gregaria. Furthermore, capa-stimulated NOS activity in all the Diptera was inhibited by the NOS inhibitor l-NAME. All capa peptides stimulate an increase in cGMP content across the dipteran species, but not in the orthopteran S. gregaria. Similarly, all capa peptides tested stimulate fluid secretion in D. melanogaster, A. aegypti, A. stephensi and G. morsitans tubules but are either without effect or are inhibitory on S. gregaria. Consistent with these results, the Drosophila capa receptor was shown to be expressed in Drosophila tubules, and its closest Anopheles homologue was shown to be expressed in Anopheles tubules. Thus, we provide the first demonstration of physiological roles for two putative A. gambiae neuropeptides. We also demonstrate neuropeptide modulation of fluid secretion in tsetse tubule for the first time. Finally, we show the generality of capa peptide action, to stimulate NO/cGMP signalling and increase fluid transport, across the Diptera, but not in the more primitive Orthoptera.


Journal of Molecular and Cellular Cardiology | 2009

Changes of intra-mitochondrial Ca2+ in adult ventricular cardiomyocytes examined using a novel fluorescent Ca2+ indicator targeted to mitochondria

Sarah Kettlewell; Pablo Cabrero; Stuart A. Nicklin; Julian A. T. Dow; Shireen A. Davies; Godfrey L. Smith

In this study a Ca(2+) sensitive protein was targeted to the mitochondria of adult rabbit ventricular cardiomyocytes using an adenovirus transfection technique. The probe (Mitycam) was a Ca(2+)-sensitive inverse pericam fused to subunit VIII of human cytochrome c oxidase. Mitycam expression pattern and Ca(2+) sensitivity was characterized in HeLa cells and isolated adult rabbit cardiomyocytes. Cardiomyocytes expressing Mitycam were voltage-clamped and depolarized at regular intervals to elicit a Ca(2+) transient. Cytoplasmic (Fura-2) and mitochondrial Ca(2+) (Mitycam) fluorescence were measured simultaneously under a range of cellular Ca(2+) loads. After 48 h post-adenoviral transfection, Mitycam expression showed a characteristic localization pattern in HeLa cells and cardiomyocytes. The Ca(2+) sensitive component of Mitycam fluorescence was 12% of total fluorescence in HeLa cells with a K(d) of approximately 220 nM. In cardiomyocytes, basal and beat-to-beat changes in Mitycam fluorescence were detected on initiation of a train of depolarizations. Time to peak of the mitochondrial Ca(2+) transient was slower, but the rate of decay was faster than the cytoplasmic signal. During spontaneous Ca(2+) release the relative amplitude and the time course of the mitochondrial and cytoplasmic signals were comparable. Inhibition of mitochondrial respiration decreased the mitochondrial transient amplitude by approximately 65% and increased the time to 50% decay, whilst cytosolic Ca(2+) transients were unchanged. The mitochondrial Ca(2+) uniporter (mCU) inhibitor Ru360 prevented both the basal and transient components of the rise in mitochondrial Ca(2+). The mitochondrial-targeted Ca(2+) probe indicates sustained and transient phases of mitochondrial Ca(2+) signal, which are dependent on cytoplasmic Ca(2+) levels and require a functional mCU.


Journal of Cell Science | 2008

Identification of two partners from the bacterial Kef exchanger family for the apical plasma membrane V-ATPase of Metazoa

Jonathan P. Day; Susan Wan; Adrian K. Allan; Laura Kean; Shireen A. Davies; Joseph V. Gray; Julian A. T. Dow

The vital task of vectorial solute transport is often energised by a plasma membrane, proton-motive V-ATPase. However, its proposed partner, an apical alkali-metal/proton exchanger, has remained elusive. Here, both FlyAtlas microarray data and in situ analyses demonstrate that the bacterial kefB and kefC (members of the CPA2 family) homologues in Drosophila, CG10806 and CG31052, respectively, are both co-expressed with V-ATPase genes in transporting epithelia. Immunocytochemistry localises endogenous CG10806 and CG31052 to the apical plasma membrane of the Malpighian (renal) tubule. YFP-tagged CG10806 and CG31052 both localise to the plasma membrane of Drosophila S2 cells, and when driven in principal cells of the Malpighian tubule, they localise specifically to the apical plasma membrane. V-ATPase-energised fluid secretion is affected by overexpression of CG10806, but not CG31052; in the former case, overexpression causes higher basal rates, but lower stimulated rates, of fluid secretion compared with parental controls. Overexpression also impacts levels of secreted Na+ and K+. Both genes rescue exchanger-deficient (nha1 nhx1) yeast, but act differently; CG10806 is driven predominantly to the plasma membrane and confers protection against excess K+, whereas CG31052 is expressed predominantly on the vacuolar membrane and protects against excess Na+. Thus, both CG10806 and CG31052 are functionally members of the CPA2 gene family, colocalise to the same apical membrane as the plasma membrane V-ATPase and show distinct ion specificities, as expected for the Wieczorek exchanger.


The Journal of Experimental Biology | 2005

Sulphonylurea sensitivity and enriched expression implicate inward rectifier K+ channels in Drosophila melanogaster renal function.

Jennifer M. Evans; Adrian K. Allan; Shireen A. Davies; Julian A. T. Dow

SUMMARY Insect Malpighian (renal) tubules are capable of transporting fluid at remarkable rates. Secondary active transport of potassium at the apical surface of the principal cell must be matched by a high-capacity basolateral potassium entry route. A recent microarray analysis of Drosophila tubule identified three extremely abundant and enriched K+ channel genes encoding the three inward rectifier channels of Drosophila: ir, irk2 and irk3. Enriched expression of inward rectifier channels in tubule was verified by quantitative RT-PCR, and all three IRKs localised to principal cells of the main segment (and ir and irk3 to the lower tubule) by in situ hybridisation, suggesting roles both in primary secretion and reabsorption. A new splice form of irk2 was also identified. The role of inward rectifiers in fluid secretion was assessed with a panel of selective inhibitors of inward rectifier channels, the antidiabetic sulphonylureas. All completely inhibited fluid secretion, with IC50s of 0.78 mmol l-1 for glibenclamide and approximately 5 mmol l-1 for tolbutamide, 0.01 mmol l-1 for minoxidil and 0.1 mmol l-1 for diazoxide. This pharmacology is consistent with a lower-affinity class of inward rectifier channel that does not form an obligate multimer with the sulphonylurea receptor (SUR), although effects on non-IRK targets cannot be excluded. Glibenclamide inhibited fluid secretion similarly to basolateral K+-free saline. Radiolabelled glibenclamide is both potently transported and metabolised by tubule. Furthermore, glibenclamide is capable of blocking transport of the organic dye amaranth (azorubin S), at concentrations of glibenclamide much lower than required to impact on fluid secretion. Glibenclamide thus interacts with tubule in three separate ways; as a potent inhibitor of fluid secretion, as an inhibitor (possibly competitive) of an organic solute transporter and as a substrate for excretion and metabolism.


Journal of Biological Chemistry | 2006

Differential gel electrophoresis and transgenic mitochondrial calcium reporters demonstrate spatiotemporal filtering in calcium control of mitochondria

Selim Terhzaz; Tony D. Southall; Kathryn S. Lilley; Laura Kean; Adrian K. Allan; Shireen A. Davies; Julian A. T. Dow

Mitochondria must adjust both their intracellular location and their metabolism in order to balance their output to the needs of the cell. Here we show by the proteomic technique of time series difference gel electrophoresis that a major result of neuroendocrine stimulation of the Drosophila renal tubule is an extensive remodeling of the mitochondrial matrix. By generating Drosophila that were transgenic for both luminescent and fluorescent mitochondrial calcium reporters, it was shown that mitochondrial calcium tracked the slow (minutes) but not the rapid (<1 s) changes in cytoplasmic calcium and that this resulted in both increased mitochondrial membrane polarization and elevated cellular ATP levels. The selective V-ATPase inhibitor, bafilomycin, further enhanced ATP levels, suggesting that the apical plasma membrane V-ATPase is a major consumer of ATP. Both the mitochondrial calcium signal and the increase in ATP were abolished by the mitochondrial calcium uniporter blocker Ru360. By using both mitochondrial calcium imaging and the potential sensing dye JC-1, the apical mitochondria of principal cells were found to be selectively responsive to neuropeptide signaling. As the ultimate target is the V-ATPase in the apical plasma membrane, this selective activation of mitochondria is clearly adaptive. The results highlight the dynamic nature and both spatial and temporal heterogeneity of calcium signaling possible in differentiated, organotypic cells and provide a new model for neuroendocrine control of V-ATPase.


The Journal of Experimental Biology | 2003

NorpA and itpr mutants reveal roles for phospholipase C and inositol (1,4,5)- trisphosphate receptor in Drosophila melanogaster renal function

Valerie P. Pollock; Jonathan C. Radford; Susan Pyne; Gaiti Hasan; Julian A. T. Dow; Shireen A. Davies

SUMMARY Mutants of norpA, encoding phospholipase Cβ (PLCβ), and itpr, encoding inositol (1,4,5)-trisphosphate receptor (IP3R), both attenuate response to diuretic peptides of Drosophila melanogaster renal (Malpighian) tubules. Intact tubules from norpA mutants severely reduced diuresis stimulated by the principal cell- and stellate cell-specific neuropeptides, CAP2b and Drosophila leucokinin (Drosokinin), respectively, suggesting a role for PLCβ in both these cell types. Measurement of IP3 production in wild-type tubules and in Drosokinin-receptor-transfected S2 cells stimulated with CAP2b and Drosokinin, respectively, confirmed that both neuropeptides elevate IP3 levels. In itpr hypomorphs, basal IP3 levels are lower, although CAP2b-stimulated IP3 levels are not significantly reduced compared with wild type. However, CAP2b-stimulated fluid transport is significantly reduced in itpr alleles. Rescue of the itpr90B.0 allele with wild-type itpr restores CAP2b-stimulated fluid transport levels to wild type. Drosokinin-stimulated fluid transport is also reduced in homozygous and heteroallelic itpr mutants. Measurements of cytosolic calcium levels in intact tubules of wild-type and itpr mutants using targeted expression of the calcium reporter, aequorin, show that mutations in itpr attenuated both CAP2b- and Drosokinin-stimulated calcium responses. The reductions in calcium signals are associated with corresponding reductions in fluid transport rates. Thus, we describe a role for norpA and itpr in renal epithelia and show that both CAP2b and Drosokinin are PLCβ-dependent, IP3-mobilising neuropeptides in Drosophila. IP3R contributes to the calcium signalling cascades initiated by these peptides in both principal and stellate cells.

Collaboration


Dive into the Shireen A. Davies'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge