Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Entomol ; 57(6): 1920-1929, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32574364

ABSTRACT

Mosquito-borne pathogens contribute significantly to the global burden of infectious diseases and are a continuing public health concern in the United States. Blood feeding by vector mosquitoes is a critical step in the transmission of human pathogens. Continuous surveillance of mosquito feeding patterns, especially in major population centers, is necessary for sustainable, effective control strategies. To better understand female feeding habits in Harris County, TX, we trapped mosquitoes from various locations, distributed among urban and semi-urban environments. Bloodmeal hosts were determined using a cytochrome C oxidase I DNA barcoding strategy. We identified a diverse array of vertebrate hosts with a high degree of avian host utilization, most surprisingly from anthropophilic species like Aedes aegypti (L.). We also detected sequences from two different vertebrate hosts in about half of specimens examined, suggesting that multiple bloodmeals had been acquired in the same feeding cycle by a sizable fraction of females in both urban and semi-urban locations. The high proportion of feeding on domestic chickens may indicate that a significant number of homeowners are rearing chickens within close proximity to study trap sites. As non-amplifying hosts, chickens may have a diluting effect on West Nile virus, as well as a zooprophylactic effect in their immediate vicinities. Ultimately, spatial and temporal host utilization patterns add insight into potential disease transmission dynamics, thereby informing vector control strategies in Harris County and other metropolitan areas.


Subject(s)
Culicidae/physiology , Mosquito Vectors/physiology , Aedes/physiology , Animals , Culex/physiology , Feeding Behavior , Female , Texas
2.
Article in English | MEDLINE | ID: mdl-19005656

ABSTRACT

Bursicon was identified in 1965 as a peptide neurohormone that initiates the tanning of the insect cuticle immediately after the shedding of the old one during the final stages of the molting process. Its molecular identity as an approximately 30 kDa bioactive heterodimer consisting of two cystine knot proteins resisted elucidation for 43 years. The sequence of the two bursicon subunits is highly conserved among arthropods, and this conservation extends even to echinoderms. We review the efforts leading to bursicon's characterization, the identification of its leucine-rich repeat-containing, G protein-coupled receptor (LGR2), and the progress towards revealing its various functions. It is now clear that bursicon regulates different aspects of wing inflation in Drosophila melanogaster besides being involved at various points in the cuticle tanning process in different insects. We also describe the current knowledge of the expression of bursicon in the central nervous system of different insects in large homologous neurosecretory cells, and the changes in its expression during the development of Manduca sexta and D. melanogaster. Although much remains to be learned, the elucidation of its molecular identity and that of its receptor has provided the breakthrough needed for investigating the diverse actions of this critical insect neurohormone.


Subject(s)
Insecta/genetics , Insecta/physiology , Invertebrate Hormones/chemistry , Invertebrate Hormones/physiology , Amino Acid Sequence , Animals , Arthropods/genetics , Arthropods/growth & development , Arthropods/physiology , Brain/metabolism , Cystine Knot Motifs , Drosophila/genetics , Drosophila/growth & development , Drosophila/physiology , Evolution, Molecular , Ganglia, Invertebrate/metabolism , Insecta/growth & development , Invertebrate Hormones/genetics , Invertebrate Hormones/isolation & purification , Molecular Sequence Data , Neuropeptides/chemistry , Neuropeptides/genetics , Neuropeptides/isolation & purification , Neuropeptides/physiology , Receptors, Neuropeptide/genetics , Wings, Animal/growth & development
3.
J Neurosci ; 28(53): 14379-91, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-19118171

ABSTRACT

Hormones are often responsible for synchronizing somatic physiological changes with changes in behavior. Ecdysis (i.e., the shedding of the exoskeleton) in insects has served as a useful model for elucidating the molecular and cellular mechanisms of this synchronization, and has provided numerous insights into the hormonal coordination of body and behavior. An example in which the mechanisms have remained enigmatic is the neurohormone bursicon, which, after the final molt, coordinates the plasticization and tanning of the initially folded wings with behaviors that drive wing expansion. The somatic effects of the hormone are governed by bursicon that is released into the blood from neurons in the abdominal ganglion (the B(AG)), which die after wing expansion. How bursicon induces the behavioral programs required for wing expansion, however, has remained unknown. Here we show by targeted suppression of excitability that a pair of bursicon-immunoreactive neurons distinct from the B(AG) and located within the subesophageal ganglion in Drosophila (the B(SEG)) is involved in controlling wing expansion behaviors. Unlike the B(AG), the B(SEG) arborize widely in the nervous system, including within the abdominal neuromeres, suggesting that, in addition to governing behavior, they also may modulate the B(AG.) Indeed, we show that animals lacking bursicon receptor function have deficits both in the humoral release of bursicon and in posteclosion apoptosis of the B(AG). Our results reveal novel neuromodulatory functions for bursicon and support the hypothesis that the B(SEG) are essential for orchestrating both the behavioral and somatic processes underlying wing expansion.


Subject(s)
Central Nervous System/metabolism , Insect Hormones/metabolism , Invertebrate Hormones/physiology , Metamorphosis, Biological/physiology , Wings, Animal/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , CD8 Antigens/metabolism , Calcitonin/metabolism , Cell Death/genetics , Cell Death/physiology , Central Nervous System/growth & development , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Ganglia, Invertebrate/growth & development , Ganglia, Invertebrate/metabolism , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , In Situ Nick-End Labeling/methods , Insect Hormones/genetics , Invertebrate Hormones/genetics , Larva , Metamorphosis, Biological/genetics , Neural Pathways/metabolism , Neurons/metabolism , Peptide Fragments/metabolism
4.
J Comp Neurol ; 506(5): 759-74, 2008 Feb 10.
Article in English | MEDLINE | ID: mdl-18076057

ABSTRACT

During posteclosion, insects undergo sequential processes of wing expansion and cuticle tanning. Bursicon, a highly conserved neurohormone implicated in regulation of these processes, was characterized recently as a heterodimeric cystine knot protein in Drosophila melanogaster. Here we report the predicted precursor sequences of bursicon subunits (Masburs and Maspburs) in the moth Manduca sexta. Distinct developmental patterns of mRNA transcript and subunit-specific protein labeling of burs and pburs as well as crustacean cardioactive peptide in neurons of the ventral nervous system were observed in pharate larval, pupal, and adult stages. A subset of bursicon neurons located in thoracic ganglia of larvae expresses ecdysis-triggering hormone (ETH) receptors, suggesting that they are direct targets of ETH. Projections of bursicon neurons within the CNS and to neurohemal secretory sites are consistent with both central signaling and circulatory hormone functions. Intrinsic cells of the corpora cardiaca contain pburs transcripts and pburs-like immunoreactivity, whereas burs transcripts and burs-like immunoreactivity were absent in these cells. Recombinant bursicon induces both wing expansion and tanning, whereas synthetic eclosion hormone induces only wing expansion.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Insect Proteins/genetics , Invertebrate Hormones/genetics , Manduca/genetics , Molting/genetics , Amino Acid Sequence , Animals , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/metabolism , Immunohistochemistry , Insect Proteins/metabolism , Invertebrate Hormones/metabolism , Manduca/growth & development , Manduca/metabolism , Metamorphosis, Biological/genetics , Metamorphosis, Biological/physiology , Molecular Sequence Data , Molting/physiology , Neurons/cytology , Neurons/metabolism , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/analysis , Receptors, Peptide/metabolism , Sequence Alignment , Tissue Distribution , Wings, Animal/growth & development , Wings, Animal/metabolism
5.
Proc Natl Acad Sci U S A ; 102(8): 2820-5, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15703293

ABSTRACT

All arthropods periodically molt to replace their exoskeleton (cuticle). Immediately after shedding the old cuticle, the neurohormone bursicon causes the hardening and darkening of the new cuticle. Here we show that bursicon, to our knowledge the first heterodimeric cystine knot hormone found in insects, consists of two proteins encoded by the genes burs and pburs (partner of burs). The pburs/burs heterodimer from Drosophila melanogaster binds with high affinity and specificity to activate the G protein-coupled receptor DLGR2, leading to the stimulation of cAMP signaling in vitro and tanning in neck-ligated blowflies. Native bursicon from Periplaneta americana is also a heterodimer. In D. melanogaster the levels of pburs, burs, and DLGR2 transcripts are increased before ecdysis, consistent with their role in postecdysial cuticle changes. Immunohistochemical analyses in diverse insect species revealed the colocalization of pburs- and burs-immunoreactivity in some of the neurosecretory neurons that also express crustacean cardioactive peptide. Forty-three years after its initial description, the elucidation of the molecular identity of bursicon and the verification of its receptor allow for studies of bursicon actions in regulating cuticle tanning, wing expansion, and as yet unknown functions. Because bursicon subunit genes are homologous to the vertebrate bone morphogenetic protein antagonists, our findings also facilitate investigation on the function of these proteins during vertebrate development.


Subject(s)
Drosophila Proteins/metabolism , Invertebrate Hormones/physiology , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Cystine/chemistry , Dimerization , Drosophila melanogaster , Invertebrate Hormones/chemistry , Molecular Sequence Data , Neuropeptides/analysis
6.
Acta Biol Hung ; 55(1-4): 91-102, 2004.
Article in English | MEDLINE | ID: mdl-15270222

ABSTRACT

We describe the molecular analysis and cellular expression of the insect peptide neurohormone, bursicon. Bursicon triggers the sclerotization of the soft insect cuticle after ecdysis. Using protein elution analyses from SDS gels, we determined the molecular weight of bursicon from different insects to be approximately 30 kDa. Four partial peptide sequences of Periplaneta americana bursicon were obtained from purified nerve cord homogenates separated on two-dimensional gels. Antibodies produced against one of the sequences identified the cellular location of bursicon in different insects and showed that bursicon is co-produced with crustacean cardioactive peptide (CCAP) in the same neurons in all insects tested so far. Additionally, using the partial peptide sequences, we successfully searched the Drosophila genome project for the gene encoding bursicon. With Drosophila as a tool, we can now verify the function of the sequence using transgenic flies. Sequence comparisons also allowed us to verify that bursicon is conserved, corroborating the older data from bioassays and immunohistochemical analyses. The sequence of bursicon will enable further analysis of its function, release, and evolution.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Invertebrate Hormones/physiology , Animals , Biological Assay , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Immunohistochemistry , In Situ Hybridization , Insecta , Invertebrate Hormones/chemistry , Neurons/metabolism , Peptides/chemistry , Periplaneta , Sodium Dodecyl Sulfate/chemistry , Species Specificity , Superoxide Dismutase/metabolism , Tenebrio , Time Factors
7.
Curr Biol ; 14(13): 1208-13, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15242619

ABSTRACT

To accommodate growth, insects must periodically replace their exoskeletons. After shedding the old cuticle, the new soft cuticle must sclerotize. Sclerotization has long been known to be controlled by the neuropeptide hormone bursicon, but its large size of 30 kDa has frustrated attempts to determine its sequence and structure. Using partial sequences obtained from purified cockroach bursicon, we identified the Drosophila melanogaster gene CG13419 as a candidate bursicon gene. CG13419 encodes a peptide with a predicted final molecular weight of 15 kDa, which likely functions as a dimer. This predicted bursicon protein belongs to the cystine knot family, which includes vertebrate transforming growth factor-beta (TGF-beta) and glycoprotein hormones. Point mutations in the bursicon gene cause defects in cuticle sclerotization and wing expansion behavior. Bioassays show that these mutants have decreased bursicon bioactivity. In situ hybridization and immunocytochemistry revealed that bursicon is co-expressed with crustacean cardioactive peptide (CCAP). Transgenic flies that lack CCAP neurons also lacked bursicon bioactivity. Our results indicate that CG13419 encodes bursicon, the last of the classic set of insect developmental hormones. It is the first member of the cystine knot family to have a defined function in invertebrates. Mutants show that the spectrum of bursicon actions is broader than formerly demonstrated.


Subject(s)
Drosophila melanogaster/genetics , Invertebrate Hormones/genetics , Molting/genetics , Phenotype , RNA, Messenger/metabolism , Amino Acid Sequence , Animals , Biological Assay , DNA Primers , Immunohistochemistry , In Situ Hybridization , Invertebrate Hormones/metabolism , Molecular Sequence Data , Point Mutation/genetics , RNA, Messenger/genetics , Sequence Alignment , Sequence Analysis, DNA
8.
J Comp Neurol ; 452(2): 163-77, 2002 Oct 14.
Article in English | MEDLINE | ID: mdl-12271490

ABSTRACT

Bursicon is the final neurohormone released at the end of the molting cycle. It triggers the sclerotization (tanning) of the insect cuticle. Until now, its existence has been verified only by bioassays. In an attempt to identify this important neurohormone, bursicon was purified from homogenates of 2,850 nerve cords of the cockroach Periplaneta americana by using high performance liquid chromatography technology and two-dimensional gel electrophoresis. Bursicon bioactivity was found in four distinct protein spots at approximately 30 kDa between pH 5.3 and 5.9. The protein of one of these spots at pH 5.7 was subsequently microsequenced, and five partial amino acid sequences were retrieved. Evidence is presented that two of these sequences are derived from bursicon. Antibodies raised against the two sequences labeled bursicon-containing neurons in the central nervous systems of P. americana. One of these antisera labeled bursicon-containing neurons in the crickets Teleogryllus commodus and Gryllus bimaculatus, and the moth Manduca sexta. A cluster of four bilaterally paired neurons in the brain of Drososphila melanogaster was also labeled. In addition, this antiserum detected three spots corresponding to bursicon in Western blots of two-dimensional gels. The 12-amino acid sequence detected by this antiserum, thus, seems to be conserved even among species that are distantly related.


Subject(s)
Central Nervous System/metabolism , Invertebrate Hormones/analysis , Invertebrate Hormones/metabolism , Amino Acid Sequence , Animals , Central Nervous System/chemistry , Chromatography, High Pressure Liquid , Drosophila , Electrophoresis, Gel, Two-Dimensional , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/metabolism , Gryllidae , Immune Sera , Immunohistochemistry , Invertebrate Hormones/isolation & purification , Larva , Manduca , Neuropeptides/metabolism , Periplaneta , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...