Molecular Characterization, Expression Analysis, and ......Rhodnius prolixus, the principal Chagas...

83
Molecular Characterization, Expression Analysis, and Physiological Roles of Allatotropin in Rhodnius prolixus by Maryam Masood A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Cell and Systems Biology University of Toronto © Copyright by Maryam Masood 2013

Transcript of Molecular Characterization, Expression Analysis, and ......Rhodnius prolixus, the principal Chagas...

  • Molecular Characterization, Expression Analysis, and Physiological Roles of

    Allatotropin in Rhodnius prolixus

    by

    Maryam Masood

    A thesis submitted in conformity with the requirements

    for the degree of Master of Science

    Department of Cell and Systems Biology

    University of Toronto

    © Copyright by Maryam Masood 2013

  • ii

    Molecular Characterization, Expression Analysis, and Physiological Roles of Allatotropin

    in Rhodnius prolixus

    Maryam Masood

    Master of Science

    Department of Cell and Systems Biology

    University of Toronto

    2013

    ABSTRACT

    Rhodnius prolixus, the principal Chagas disease vector, requires a blood meal to complete its

    moult cycle into the next stage. Allatotropins (ATs), a family of peptides first isolated from

    Manduca sexta, have been shown to regulate the biosynthesis of juvenile hormone, an insect

    growth and development hormone; however, ATs, being multimodal peptides, also exhibit

    myotropic effects on some insect visceral muscles. Here, this AT family of peptides has been

    examined in R. prolixus. Genomic analysis revealed a cDNA fragment of 973bp encoding one

    mature amidated AT tridecapeptide (Rhopr-AT) with high transcript levels observed, via RT-

    PCR, in the central nervous system (CNS) and pool of fat body and trachea. AT-like

    immunoreactive neurons were found throughout the CNS and AT-like immunoreactive

    processes were present on some peripheral tissues. Bioassays using hindgut and dorsal vessel

    contraction, however, failed to demonstrate any effect of Rhopr-AT on these tissues. Future

    work will examine the effects of Rhopr-AT on JH production.

  • iii

    Acknowledgments

    I would like to express a heartfelt gratitude to my supervisor, Dr. Orchard, whose expertise,

    understanding, patience, and vast knowledge added considerably to my graduate experience. I

    appreciate his ongoing guidance and advice that enabled me to stay on track and work at a

    smooth pace throughout the Master’s program. I am also extremely thankful to Dr. Angela

    Lange and Dr. Joel Levine for providing me with invaluable input and feedback that helped

    shape me into a better research student. Furthermore, I am indebted to my colleagues without

    whom this journey would be incomplete. Meet, Dohee, Lisa, Nikki, Laura, Himali all the

    undergraduates and work-study students, thank you all for enriching my graduate experience!

    I feel special thanks is in order for Garima and Nigam for their continual guidance, support and

    mere presence with which they brought the lab to life and made it such a wonderful place to be

    in! Thank you amigos!

    On a final note, I’d like to thank my family for putting up with my “princess” behaviour over

    the last two years. Your support was absolutely fundamental to my research and I am extremely

    appreciative!

  • iv

    Table of Contents ABSTRACT ................................................................................................................................... ii

    Acknowledgments ......................................................................................................................... iii

    List of Tables ................................................................................................................................... v

    List of Figures ................................................................................................................................. vi

    List of In-text Abbreviations ........................................................................................................ vii

    Molecular Characterization, Expression Analysis and Physiological Roles of Allatotropin

    in Rhodnius prolixus ...................................................................................................................... 1

    INTRODUCTION ........................................................................................................................... 1

    Rhodnius prolixus ........................................................................................................................ 1

    Feeding-related physiological processes in Rhodnius prolixus .................................................. 2

    Neuropeptides and Families ........................................................................................................ 3

    G-protein coupled receptors (GPCRs) ........................................................................................ 5

    Allatoregulatory Peptides ............................................................................................................ 6

    Allatostatins ................................................................................................................................. 7

    Allatotropins ................................................................................................................................ 9

    THESIS OBJECTIVE ................................................................................................................... 16

    MATERIALS AND METHODS .................................................................................................. 17

    RESULTS ...................................................................................................................................... 27

    DISCUSSION ................................................................................................................................ 54

    FUTURE DIRECTIONS ............................................................................................................... 62

    REFERENCES .............................................................................................................................. 64

  • v

    List of Tables

    Table 1. Sequences of known or predicted allatotropin peptides.. ................................................ 10

    Table 2. Sequence similarities between allatotropin-like peptides of different lepidopteran

    species ............................................................................................................................................ 14

    Table 3. Gene-specific primers used for Rhopr-AT prepropeptide cDNA amplification ............. 19

    Table 4. Amino acid sequence and purity of peptides used for the bioassays............................... 25

  • vi

    List of Figures

    Figure 1. A schematic representing the possible targets of allatoregulatory peptides..................... 8

    Figure 2. A) Nucleotide sequence of allatotropin (AT) precursor peptide cDNA in

    R. prolixus. B) Schematic representation of Rhopr-AT gene organization. C) Northern

    blot of Rhopr-AT transcript ........................................................................................................... 28

    Figure 3. ClustalO alignment of Rhopr-AT prepropeptide sequence with allatotropin

    prepropeptide sequences from other arthropods. ........................................................................... 31

    Figure 4. Phylogenetic relationship of allatotropin prepropeptide sequence with

    other insect sequences.................................................................................................................... 33

    Figure 5. RT-PCR representing the expression profile of the Rhopr-AT transcript in

    R. prolixus ...................................................................................................................................... 35

    Figure 6. Confocal images showing AT-like immunostaining in cells of R. prolixus

    CNS, corpus cardiacum and dorsal vessel ..................................................................................... 39

    Figure 7. Confocal images showing AT-like immunostaining in cells of R. prolixus

    CNS and peripheral nerves ............................................................................................................ 41

    Figure 8. Camera lucida map depicting distribution of AT-like immunoreactive cells

    within the CNS. A) Dorsal view. B) Ventral view of the brain..................................................... 43

    Figure 9. Confocal images showing AT-like immunoreactivity in peripheral tissues

    of R. prolixus ................................................................................................................................. 45

    Figure 10. Sample traces depicting effects of Rhopr-AT and Rhopr- Kinin 2 on IVth

    (A) and Vth

    (B) instar hindguts ...................................................................................................... 48

    Figure 11. Dose-response curves representing the change in hindgut contractions of

    IVth

    instar R. prolixus hindguts ...................................................................................................... 50

    Figure 12. A) Dose-response curve for Rhopr-AT activity on heart-beat of R. prolixus .............. 52

  • vii

    List of In-text Abbreviations

    Note: Only abbreviated scientific terms used repeatedly have been listed here. Initial use of an

    in-text abbreviation is preceded by the full form.

    AST: Allatostatin

    AT: Allatotropin

    AT-like-ir: allatotropin-like immunoreactive

    cAMP: 3’-5’-cyclic adenosine monophosphate

    CC/ CA: corpora cardiaca/ corpora allata

    CNS: central nervous system

    CRF: corticotropin releasing factor

    DH: diuretic hormone

    DV: Dorsal vessel

    GPCR: G protein-coupled receptor

    HG: Hindgut

    JH: Juvenile Hormone

    MALDI-TOF MS/MS: matrix assisted laser desorption ionization time-of-flight tandem

    mass spectrometry

    MTGM: mesothoracic ganglionic mass

    MTs: Malpighian tubules

    ORF: open reading frame

    PRO: prothoracic ganglion

    SG: Salivary gland

    SOG: suboesophageal ganglion

    SP: Sex peptide

    Rhopr-kinin 2: Rhodnius prolixus kinin- 2

    UTR: Untranslated region

  • 1

    Molecular Characterization, Expression Analysis and Physiological Roles of

    Allatotropin in Rhodnius prolixus

    INTRODUCTION

    Rhodnius prolixus

    American trypanosomiasis also referred to as the Chagas disease is a widespread pandemic in

    Central and South America. Approximately 7-8 million people are infected worldwide, though

    mostly in Latin America, with an estimated report of 50,000 deaths each year (Lima et al., 2010;

    Rassi et al., 2010; World Health Organization, 2012). Rhodnius prolixus, one of the principal

    vectors for Trypanosoma cruzi - the parasite responsible for Chagas disease, is an obligate

    feeder whereby each blood meal initiates growth and development into the next instar stage.

    Since the lifecycle of R. prolixus is dependent on the availability of a blood meal, each instar,

    when provided the opportunity, is capable of gorging up to ten times its original body weight.

    The considerable intake of fluid limits mobility, and increases the risk of predation; therefore,

    within 2-3 minutes of blood feeding R. prolixus urinates and it is during this post-prandial

    diuresis that the parasite, T. cruzi, is transmitted from the vector to the host (Orchard, 2006;

    Rassi et al., 2010). Transmitted T. cruzi leads to either an acute symptomatic phase, expressed

    by 1-2 % of the population, or a chronic asymptomatic phase, expressed by about 70% of the

    infected population (World Health Organization, 2012). The acute phase is treatable with

    antiparastic drugs such as benznidazole (Rassi et al., 2010); however, since the symptoms are

    mild they usually go unnoticed and the disease develops into a fatal phase leading to chronic

    cardiomyopathy, gastrointestinal and neurological problems (Rassi et al., 2010; World Health

    Organization, 2012). Since the chronic stage of Chagas disease is incurable, the only other

    treatment currently available is prevention. Triatomes such as R. prolixus are commonly found

    in thatched roofs along with nooks and crevices associated with poor housing conditions (World

    Health Organization, 2008). Therefore, Chagas disease has been seen to be strongly associated

    with socio-economic conditions, with people at the poverty line at higher risk of infection.

    Control programs implemented in regions of Central and South America have been successful in

    diminishing the number of disease cases since the late 1990s (Rassi et al., 2010); however,

  • 2

    complete eradication can only be possible by inhibition of the vector host and information

    pertinent to their lifecycles can be aided by relevant molecular and physiological research.

    Feeding related physiological processes in Rhodnius prolixus

    Diuresis

    Rhodnius prolixus is capable of engorging a blood meal up to 10-12 times its original unfed

    body weight. The substantial blood meal causes inflation of the abdomen leading to activation

    of stretch receptors that trigger a cascade of events regulated by neurohormones and hormones;

    diuresis being one of them. The imbibed meal is rich in water and ions which are consequently

    absorbed from the gut into the haemolymph and hence need to be sequentially secreted from the

    insect to maintain the internal osmotic balance of the haemolymph (Coast, 2009). The amount of

    water secreted in the urine depends on the pre-feeding state of the insect, with dessicated insects

    secreting less volume of urine compared to newly emerged fed instars (Maddrell, 1964). Ionic

    regulation is controlled by specialised Malpighian tubules (MTs) that are characterised by their

    location and relative epithelial conductance to ions; the upper tubules secrete excess Na+ ions

    while the lower tubules preferentially reabsorb K+ and Cl

    - ions (Coast, 2009). Thus, eventually,

    a hyposomotic urine is produced which travels to the hindgut to be released from the anus.

    Growth and development

    Another physiological response initiated by abdominal stretch due to feeding is initiation and

    completion of the moult cycle (Callier and Nijhout, 2013). Wigglesworth’s pioneering studies

    (see 1934, 1948) regarding ecdysis identified the presence of a moulting hormone, later referred

    to as “juvenile hormone” (JH) that was released from the corpora allata (CA) of R. prolixus.

    Regulated production and release of this moulting hormone during critical time periods was

    shown to result in either ecdysis into the next instar or metamorphosis into adults. However,

    since then it has been determined that ecdysis is not an isolated event controlled by a single

    hormone, rather a cocktail of hormones orchestrate growth and development and ecdysis

    behaviour. Some of the neurohormones and hormones known to be involved in other insects

    regulating this process are ecdysteroids, corazonin, preecdysis-triggering hormone (PETH),

    ecdysis triggering hormone (ETH), eclosion hormone (EH) and crustacean cardioactive peptide

    (CCAP) (Kim et al., 2006a; Lee do and Lange, 2011; Roller et al., 2010; Vafopoulou et al.,

  • 3

    2012; Zitnan and Adams, 2005). Ecdysis related hormones are highly conserved among insect

    species and orthologues are seen in Diptera and Lepidopterans (Kim et al., 2004).

    Neuropeptides and Families

    Physiological processes including foraging, feeding, growth, development, metabolite

    regulation, digestion, reproduction and cardiovascular control etc are modulated by numerous

    neuropeptides that are released in a developmental stage-specific manner (for specific roles of

    neuropeptides on aforementioned physiological processes, see: Audsley and Weaver, 2009;

    Coast, 1996, 2009; Mykles et al., 2010; Wielendaele et al., 2013). Neuropeptides are short

    chains of amino acids that offer great flexibility in function through structure and mode of

    action; they may be neuromodulators, neurohormones or neurotransmitters (Orchard et al.,

    2001). Neurohormones are neuropeptides (or amines) released into the haemolymph, from

    neurosecretory cells, that subsequently circulate throughout the body; hence targeting multiple

    tissues simultaneously. A neurohormone-mediated response is elicited only by those tissues that

    possess appropriate ligand-specific receptors. Neurotransmitters, on the other hand, are released

    at synapses, and result in discreet, private communication; while neuromodulators are released

    locally in a paracrine manner and diffuse locally towards target tissues (Nässel, 2002; Orchard

    et al., 2001). Due to the relative modes of transfer the duration of the message also differs

    between neurotransmitters and neurohormones, with neurohormones relaying persistent, long-

    term messages and neurotransmitters conveying short-term messages (Nässel, 2002; Orchard et

    al., 2001). The multiplicity of neuropeptides and their diverse physiological roles make them

    excellent cell-cell communication molecules; hence neuropeptides are well-represented among

    various taxa, with even simple organisms such as coelenterates and nematodes producing

    several neuropeptides that work in concert with one another.

    Neuropeptides act through multiple receptor types, though most interact with G-protein coupled

    receptors (GPCRs), and functional flexibility is further enhanced by tissue receptivity, i.e.

    receptors for neuropeptides are expressed in varying amounts in different tissues in a stage-

    specific manner. In Aedes aegypti, for example, the expression levels of allatotropin receptor

    vary between brain, gut and reproductive tissues and receptor levels fluctuate according to the

    developmental and feeding state of the organism (Nouzova et al., 2012). Hence, a single

    neuropeptide can conduct a coordinated physiological response by affecting multiple tissues

  • 4

    simultaneously. Moreover, multiplicity can be achieved by the presence of several isoforms of a

    neuropeptide that are in turn released or expressed in a time and tissue-specific manner e.g.

    allatotropins, allatostatins, leucokinins, adipokinetic hormones and tachykinins can all have

    multiple isoforms within single species (Nässel, 2002). Additionally, multiple neuropeptides

    may be co-localised and co-released, or released as sequential peptidergic ensembles from

    neurosecretory cells, resulting in increased complexity and efficacy in regulation of

    physiological processes. Examples of such coordinated biological behaviours include feeding,

    ecdysis and reproduction (Audsley and Weaver, 2009; Kim et al., 2006b; Truman, 2005;

    Wielendaele et al., 2013).

    Familial Organization

    Neuropeptide nomenclature stems from grouping peptides with similar biological functions or

    sequence similarities under one family. However, sometimes, structurally unrelated peptides are

    also grouped together based on their preliminary identification e.g. members of the allatostatin

    family: allatostatin A, B and C. These allatostatins are derived from three different genes, have

    different peptide sequences and not all have allatoregulatory function across species;

    nevertheless they are collectively classified as allatostatins. Hence, while most neuropeptides

    are organized into respective functional or structural families, some discrepancies do exist and

    these will probably increase as additional (related or unrelated) functions of neuropeptides are

    discovered.

    Identification

    Traditionally neuropeptides were identified via mass tissue extraction followed by peptide

    sequencing through Edman degradation. Matrix-assisted laser desorption/ionization time-of-

    flight mass spectrometry (MALDI-TOF MS) and immunohistochemistry with antibodies

    specific to the peptide have aided in determining peptide sequence and distribution within

    peripheral tissues and central nervous systems. However, with the recent availability of genomic

    databases for various arthropod species, prediction and sequencing of genes encoding

    neuropeptides and candidate receptors has become possible. Therefore, specialized techniques

    such as in situ hybridisation, using transcript specific probes, can now be deployed to localize

    neuropeptide gene transcripts in cells. Moreover, with molecular techniques such as qRT-PCR

  • 5

    one can quantify the neuropeptide transcript levels in various tissues at different developmental

    time points. Additionally, specific biological roles of neuropeptides in a given process can be

    assigned by altering neuropeptide expression levels via gene silencing through RNA

    interference (RNAi) and GAL4/UAS systems. Receptor identification and characterisation has

    further fuelled neuropeptidomic research and thus vital information about specific neuropeptide-

    receptor interactions can be attained along with an insight into possible second messenger

    pathways involved. Although genomic sequencing and annotation have accelerated

    neuropeptide research, deficiencies still exist in our comprehension of post-translational

    modifications and the structure of the mature neuropeptides eventually produced; hence, these

    new methods of genetic sequencing and bioinformatics need to be complemented with

    traditional techniques of mass spectrometry for determining the complete biochemical nature of

    the mature neuropeptides (Caers et al., 2012; Nässel and Winther, 2010).

    Synthesis and Release

    Neuropeptides are secretory proteins transcribed with signal sequences guiding them to the

    endoplasmic reticulum for post-translational modifications. They are synthesised as large

    precursor molecules that can encode one or many structurally-related or unrelated mature

    neuropeptides, each subsequently cleaved at specific dibasic cleavage sites (Caers et al., 2012;

    Veenstra, 2000). Following post-translational modifications such as glycosylation, N-terminal

    pyroglutamate addition, C-terminal amidation etc the neuropeptides are packaged into secretory

    storage granules and channelled to axon terminals where they are stored and can undergo

    exocytic release when the membrane is depolarized (Nässel, 2002; Zupanc, 1996).

    G-protein coupled receptors (GPCRs)

    G-protein coupled receptors (GPCRs), as the name suggests, are transmembrane receptors

    associated with heterotrimeric GTP binding proteins, G proteins, at their cytosolic C-terminal

    (Caers et al., 2012). The structure of GPCRs is conserved and defined by the presence of 7 α-

    helical chains that span the membrane thus linking the outside of the cell to the inside. These

    helical chains are referred to as the transmembrane domains (Meeusen et al., 2003). Messages

    are transduced when specific ligands such as neuropeptides or biogenic amines (Hauser et al.,

  • 6

    2008) interact with sequences at the extracellular, usually glycosylated, N-terminus. Ligand-

    binding initiates a cascade of intracellular events that begin with activation and dissociation of

    the heterotrimeric GTP molecule via dephosphorylation. The dissociated units subsequently

    interact with various host molecules, such as phospholipase Cβ or adenyl cyclase, resulting in

    either an increase of intracellular Ca2+

    levels or synthesis of 3’-5’-cyclic adenosine

    monophosphate (cAMP). Moreover, the subunits can interact directly with ion channels; Ca2+

    or

    K+

    ion channels, resulting in an influx of extracellular calcium and potassium (Caers et al.,

    2012; Meeusen et al., 2003). Receptor activation by certain ligands can be determined by

    measuring these intracellular Ca2+

    or cAMP levels with the help of cell lines cells expressing

    apoaequorin proteins or the luciferase gene respectively (Caers et al., 2012; Hauser et al., 2006).

    Additionally, receptor activation can also be used to identify receptor-specific ligands from a

    library of neuropeptides, and hence isolated, putative receptors can be deorphanized (reverse

    pharmacology). The entire signal transduction pathway culminates in ligand degradation or

    receptor desensitization/internalization due to prolonged stimulation. Receptor-ligand

    interactions are an important avenue to explore due to their manifestation as mediators of critical

    biological roles such as reproduction, feeding, growth and behaviour. Biosynthetic agents

    engineered towards altering such interactions could be pivotal in designing species-specific

    pesticides.

    Allatoregulatory Peptides

    Allatoregulatory peptides are a class of neuropeptides that have been identified as regulators of

    JH biosynthesis from the CA of insects. These include stimulatory allatotropins and inhibitory

    allatostatins. Juvenile hormones are sesquesterpenoids produced by the endocrine CA

    (Goodman et al, 2005) that govern key biological processes in insects including development

    and metamorphosis (Tobe and Bendena, 1999), reproduction including vitellogenesis and egg

    production (Davey, 2007), and behaviour (Nijhout and Wheeler, 1982; Riddiford, 2008; Zhu et

    al., 2008). Evolutionary analyses reveal ‘acquired roles’ for JH regulation in developmental

    processes (ecdysis and metamorphosis) and subsequent modulation of this hormone by

    allatoregulatory peptides (Tobe and Bendena, 1999). Hence, despite the recent revelation of

    stage, species and dose-dependent regulation of allatoregulatory peptides on JH titres (Audsley

  • 7

    et al., 2008; Hoffmann et al., 1999; Weaver and Audsley, 2009), the possible additional

    ancestral roles that may still be modulated by these allatoregulatory neuropeptides must also be

    taken into consideration. Figure 1 represents a schematic adapted from McNeil and Tobe (2001)

    that summarises some of the main physiological roles regulated by allatoregulatory peptides;

    however, it must be noted that allatoregulatory peptides are pleiotropic in nature and only a

    subset from the plethora of physiological functions mediated by these neuropeptides has been

    represented in Figure 1.

    Allatostatins

    Allatostatins (ASTs) belong to a family of structurally-unrelated peptides that were originally

    isolated as inhibitory neuropeptides affecting JH biosynthesis (Nässel, 2002). Due to their

    structural disparity ASTs have been grouped into three distinct families: allatostatin-A (AST-A),

    allatostatin-B (AST-B) and allatostatin-C (AST-C); each family having been identified from

    different species and each possessing multimodal characteristics. The AST-A, also called the

    FGLamides due to their characteristic C terminal Y/FXFGL/I/V sequence and AST-C, also

    known as PISCF-ASTs due to their C terminal motif, were originally isolated as inhibitors of JH

    from Diploptera unipuncta and Manduca sexta respectively (Kramer et al., 1991; Woodhead et

    al., 1989). Based on these findings, they are alternatively also referred to as the Diploptera type

    or Manduca sexta type ASTs; however, since then both AST-A and AST-C have been identified

    in numerous other species where they are found to express allatostatic properties along with

    inhibitory roles in a variety of physiological processes e.g. feeding (Weaver and Audsley, 2009).

    In addition, AST-B type neuropeptides (W2W

    9) were first isolated as myoinhibitory peptides

    (MIPs) from L. migratoria (Nässel, 2002; Schoofs et al., 1991) and later identified as inhibitors

    of JH biosynthesis in M. sexta and G. bimaculatus (Nässel, 2002); hence, the AST-B family is

    also acknowledged as Lom-MIP or locust-type AST. Therefore, it is evident that allatostatins

    exert a species specific inhibition on JH biosynthesis and are not universal in their allatostatic

    function as initially considered.

  • 8

    Figure 1. A schematic representing the possible targets of allatoregulatory peptides (AT and

    AST). Modified from McNeil and Tobe (2001).

  • 9

    Allatotropins

    Identification and Characterization

    Another set of neuropeptides that regulate JH biosynthesis from the CA are the family of

    allatotropins. Allatotropins (ATs) were first isolated as amidated tridecapeptides from the heads

    of Manduca sexta pharate adults and subsequently shown to have stimulatory effects on JH

    biosynthesis from the CA (Katoaka et.al., 1989). Since then peptides similar to AT have been

    characterised or predicted in various holometabolous and hemimetabolous insects (Table 1).

    Surprisingly though, ATs are also present in non-insect species such as arachnids (Horodyski,

    2013), annelids (Ukena et al., 1995), crustaceans (Christie et al., 2011) and even molluscs;

    species that lack JH (Jing et al., 2010; Veenstra, 2010).

    The mature, functional peptide is usually 13-14 residues long, with a highly conserved

    pentapeptide TARGF-amide C-terminal motif which constitutes the biologically-active domain

    (Table 1; Audsley et al., 2008). Hitherto, only two structurally-unrelated ATs have been

    identified; the classic TARGF-amide AT and AT-2, a unique allatotropin isolated from S.

    frugiperda with a C-terminal PISCF sequence (Abdel-latief et al., 2004a). AT-2 has a C-

    terminal sequence similarity with the traditional AST-C and was expected to be allatostatic in

    nature; however, due to its stimulatory effect on JH biosynthesis it was termed AT-2 (Abdel-

    latief et al., 2004a). To date, this novel AT is limited to S. frugiperda and its presence has not

    been identified in any other organism.

    Allatotropin and Juvenile Hormone

    Allatotropins, as implied by their name, are potent stimulators of JH biosynthesis from the CA

    of some insects. They were first identified as allatoregulatory peptides in adult M. sexta were

    they were able to induce an 80% increase in JH biosynthesis. Subsequently, ATs have been

    found to regulate JH titres in a dose–dependent and stage specific manner in several insect

    species including lepidopterans: S. frugiperda, L. oleracea, H. virescens, (Audsley et al., 2000;

    Oeh et al., 2000; Rachinsky et al., 2003); dipterans: A. aegypti and P. regina (Li et al., 2003; Tu

    et al., 2001), a hymenopteran: A. mellifera (Rachinsky and Feldlaufer, 2000) and a

    dermapteran: E. annulipes (Rankin et al., 2005). It has been confirmed by in-vitro and receptor-

  • 10

    Table 1. Sequences of known or predicted allatotropin peptides. Amino acids shared by all

    sequences are highlighted in black, while conserved amino acids are shaded grey.

    Species Peptide Sequence References

    Lepidopterans Manduca sexta GFK-NVEMMTARGF (Taylor et al., 1996)

    Bombyx mori GFK-NVEMMTARGF (Nagata et al., 2012)

    Spodoptera frugiperda GFK-NVEMMTARGF (Abdel-latief et al., 2003)

    Samia cynthia ricini GFK-NVEMMTARGF (Sheng et al., 2007)

    Helicoverpa armigera GFK-NVEMMTARGF (Yin et al., 2005)

    Pseudaletia unipuncta GFK-NVEMMTARGF (Truesdell et al., 2000)

    Diptera

    Aedes aegypti APFR-NSEMMTARGF (Veenstra and Costes, 1999)

    Anopheles gambiea APFR-NSEMMTARGF (Riehle et al., 2002)

    Hemiptera

    Rhodnius prolixus GFK-NVQLSTARGF

    (Ons et al., 2009; Ons et al.,

    2011)

    Ixodida

    Ixodes scapularis GFR-KMKISTARGF (Horodyski, 2013)

    Hymenopetra

    Nasonia vitripennis GFQ-PEYISTAYGF (Hauser et al., 2010)

    Cladocera

    Daphnia pulex GFK-TVGLATARGF (Christie et al., 2011)

    Coleoptera

    Tribolium castaneum GIEALKYH-NMDLGTARGY (Weaver and Audsley, 2008)

    Orthoptera

    Locusta migratoria GFK-NVALSTARGF (Paemen et al., 1991)

    Blattodea

    Periplaneta americana GFK-NVALSTARGF (Neupert et al., 2009b)

    Leucophaea maderae GFK-NVALSTARGF (Neupert et al., 2009b)

    Mollusca

    Aplysia californica GFRLNSASRVAHGY (Jing et al., 2010)

    Lottia gigantea GFKANSASRVAHGY (Veenstra, 2010)

    Haplotaxida

    Eisenia foetida GFKDGAADRISHGF (Elekonich and Horodyski,

    2003; Ukena et al., 1995)

  • 11

    mediated assays that the mode of action of AT upon JH biosynthesis stimulation is through Ca2+

    and cAMP dependent-pathways, whereby an increase in the intracellular Ca2+

    levels of the CA

    cells results in an increased JH production (Horodyski et al., 2011; Rachinsky et al., 2003;

    Vuerinckx et al., 2011).

    Multimodal Roles of Allatotropin

    Allatotropins are pleiotropic peptides that have been found to regulate diverse physiological

    processes and are hence not just limited to stimulation of JH biosynthesis from the CA of

    insects. Veenstra (1994) identified a cardioacceleratory role of AT in the tobacco hornworm, M.

    sexta, and since then ATs have been found as cardioacceleratory peptides stimulating

    contractions in a dose-dependent manner in various species including L. maderae, P. americana

    (Rudwall et al., 2000) and T. infestans (Sterkel et al., 2010). Moreover, ATs have also been

    identified as myostimulatory peptides inducing gut contractions (foregut, whole gut or hindgut)

    in multiple lepidopteran and blattodea species, along with a coleopteran, T.castaneum,

    dermapteran: E. annulipes and hemipteran: T. infestans (Duve et al., 1999; Oeh et al., 2003;

    Rankin et al., 2005; Santini and Ronderos, 2007; Vuerinckx et al., 2011). Additionally, in M.

    sexta ATs have been found to be potent inhibitors of ion transport across the posterior midgut

    epithelium; however, this is a stage and species-specific response that has only been observed in

    feeding fourth instars and day 2 fifth instars, and is completely absent in other lepidopterans

    (Lee et al., 1998).

    Allatotropin-related peptides (ATRPs) have also been identified from the male accessory glands

    of L. migratoria (Lom-AG-MT) and head extracts of Leptinotarsa decemlineata, (Led-OVM)

    (Paemen et al., 1991; Spittaels et al., 1996). These ATRPs are found to have a preferential

    myostimulatory effect on female oviducts than hindguts of both aforementioned species as well

    as Leucophaea maderae. Myotropic regulation of reproductive tissues is another novel role of

    ATs that opens avenues for research into other reproductive processes (vitellogenesis, sexual

    receptivity, oviposition rate, egg deposition) that are modulated by AT either directly,

    myostimulation, or indirectly via JH regulation; a role similar to the sex peptides of Drosophila

    (Chen et al., 1988; Elekonich and Horodyski, 2003).

  • 12

    A possible role in photic entrainment of the circadian clock has also been suggested for AT. In

    monarch butterflies, JH levels are up-regulated in summer butterflies compared to migratory

    butterflies. The increased JH levels in suitable environmental conditions allow for rapid

    completion of female ovarian cycle, vitellogenesis, as well as production of sex pheromones -

    all factors leading to an onset of sexual maturity in recently emerged females. Concurrently AT

    levels are also up-regulated in summer monarch butterflies vs migratory butterflies suggesting a

    possible allatoregulatory role of the neuropeptide upon JH production (Zhu et al., 2008). Similar

    JH up-regulation was also observed in summer vs migratory individuals of the true armyworm,

    P. unipuncta (McNeil and Tobe, 2001); however, since AT levels were not quantified in the two

    behavioural phenotypes, it is only speculated that a similar JH stimulation and up-regulation of

    AT levels occurs within the sedentary summer moths. Moreover, AT has been found to be

    associated with the circadian rhythms of L. maderae, where time-dependent injections cause a

    phase shift in the circadian rhythms of the innate locomotor activity (Petri et al., 2002; Petri et

    al., 1995).

    The presence of ATs in non-insect phyla such as annelids and molluscs, species that lack JH,

    suggests an alternative role for AT. Indeed, it has been found that the allatotropin-related

    peptide present in Aplysia regulates muscle contractions in the protraction behaviour (Jing et al.,

    2010). Moreover, in the Turbellaria and Catenulida AT-like immunoreactivity has been found

    associated with muscle tissues suggesting a possible myotropic activity in these taxa (Adami et

    al., 2011). Considering these findings, it has been suggested that the ancestral role of AT is

    myotropic, with allatoregulation of JH titres being an acquired function in arthropods over time

    (Elekonich and Horodyski, 2003; Tobe and Bendena, 1999).

    Allatotropin Gene

    The family of ATs, including allatotropin-like (ATL) and allatotropin related peptides (ATRPs),

    have been identified and extensively characterised among the invertebrate phyla including

    insects, molluscs, annelids, arachnids and crustaceans (Christie et al., 2011; Jing et al., 2010;

    Ukena et al., 1995; Veenstra, 2010); however, genomic characterisation has been largely limited

    to lepidopterans (Abdel-latief et al., 2003; Nagata et al., 2012; Sheng et al., 2007; Taylor et al.,

    1996; Truesdell et al., 2000; Yin et al., 2005) with the only species outside this order from

    which the AT gene has been isolated and cloned being the dipteran, A. aegypti (Veenstra and

  • 13

    Costes, 1999). The AT neuropeptide is encoded by a single gene as a single transcript, except in

    lepidopterans where multiple splice variants are present. The number of variants and hence the

    number of isoforms expressing AT and ATL peptides are species-specific and range from 3-4

    isoforms per species. Though ATL peptides do not share a sequence homology to AT, they are

    relatively well conserved amongst themselves (Table 2). Allatotropin and ATL peptides are

    preferentially expressed in certain tissues of the organism in a time- and developmental-stage

    specific manner (Horodyski et al., 2001) thereby adding to the multiplicity and hormonal

    complexity of allatotropin and its functions. Besides the lepidopterans, the only other species

    predicted to have multiple isoforms is Ixodes scapularis (Horodyski, 2013). With the

    commencement of the peptidomic era, ATs have also been predicted or determined in other

    insects (Table 1) using in silico bioinformatic tools to screen the genomes and/or mass

    spectrometric analyses. Initially thought to be absent from the parasitic wasp: Nasonia

    vitrepennis (Hauser et al., 2010), a subsequent in silico genomic analysis has predicted the

    presence of a hypothetical AT protein (Ac: XP_003427574.1); however, despite the presence of

    this genomic toolkit, no AT or ATRPs have been identified in Drosophila species (Adams et al.,

    2000; Nässel, 2002; Nässel and Winther, 2010).

    Allatotropin Receptor

    The allatotropin receptor (ATR) has been identified from the yellow fever mosquito, A. aegypti

    (Nouzova et al., 2012); Tribolium castaneum (Vuerinckx et al., 2011); and two lepidopterans:

    Bombyx mori (Yamanaka et al., 2008) and M. sexta (Horodyski et al., 2011). The receptors

    share a high sequence similarity with mammalian orexin receptors, suggesting a possible

    evolutionary link between the two (Caers et al., 2012). Analysis of the Drosophila genome

    again yielded no results for the presence of ATRs (Caers et al., 2012).

  • 14

    Table 2. Sequence similarities, highlighted in black, between allatotropin-like peptides of

    different lepidopteran species

    Species Sequence References

    Manduca sexta_I GTFKPNSNILIARGY (Horodyski et al., 2001)

    Samia cynthia ricini_I GTFKPNSNILIARGY (Sheng et al., 2007)

    Helicoverpa armigera_I GVFRPNSNVLIARGY (Yin et al., 2005)

    Manduca sexta_II GTPTFKSPTVGIARDF (Horodyski et al., 2001)

    Samia cynthia ricini_II GTPIFKSPTVGIARDF (Sheng et al., 2007)

    Helicoverpa armigera_II GTPTFKSPTVGIARDF (Yin et al., 2005)

    Manduca sexta_III PWFNPKSKLLVSTRF (Horodyski et al., 2001)

    Samia cynthia ricini_III FNPKNSLMVAYDF (Sheng et al., 2007)

    Helicoverpa armigera_III FNPKSNLMVAYDF (Yin et al., 2005)

  • 15

    Allatotropin receptors are expressed in a tissue specific manner with high transcript levels in the

    central nervous systems and reproductive tissues, with the only exception of high transcript

    levels in the guts of M. sexta. The receptor transcript exhibits sex specific expression in T.

    castaneum (Vuerinckx et al., 2011), while in the lepidopterans and dipterans, transcript levels

    are regulated according to the developmental stage and feeding state of the organism e.g. a

    decrease in receptor expression is observed in A. aegypti following a blood meal (Nouzova et

    al., 2012). The receptor expression levels follow endogenous JH titres through different

    developmental stages. In lepidopterans, allatotropin receptors exhibit a preferential binding to

    AT and ATL peptides in a dose-dependent manner, whereby ATL-I peptide, with a semi-

    conserved C-terminal domain, is more potent than AT itself (Horodyski et al., 2011). The ATRs

    are typical GPCRs that activate the phospholipase Cβ and adenyl cyclase pathway leading to an

    increase in intracellular Ca2+

    levels and cAMP levels. It has been found in H. virescens that an

    increased Ca2+

    concentration leads to an increased sensitivity of the CA cells (Rachinsky et al.,

    2003). Moreover, in the same year Oeh et al. (2003) discovered that the signal transduction

    pathway for the myostimulatory effects of AT on visceral muscles of H. virescens was also

    mediated by an increase in intracellular Ca2+

    levels through the PKC pathway. Hence, signal

    transduction through Ca2+

    mediated pathways seems to be the primary route of action for ATs.

    Allatotropins and Rhodnius prolixus

    In 2009, Ons et al. isolated three putative, truncated isoforms of AT from R. prolixus brains

    following a neuropeptidomic search using mass spectrometry. In a subsequent study, however,

    Sterkel et al. (2011) determined that the truncated isoforms were probably structurally-

    redundant and only one mature amidated tridecapeptide was thought to be the functional AT

    (Rhopr-AT). Subsequent research by Ons et al. (2011) led to the discovery, isolation and

    characterisation of the gene encoding AT in R. prolixus; however, these few aforementioned

    experiments encapsulate the extent of our knowledge regarding ATs in R. prolixus, and aspects

    such as gene expression, distribution and peptide function are still untouched.

    Since some research has been done in a closely related triatomine bug, Triatoma infestans, an

    initial comparative approach can be taken to study the distribution and roles of AT in R.

    prolixus. In T. infestans, allatotropin-like immunoreactive (AT-like-ir) processes have been

    observed extending over the dorsal vessel and retrocerebral complex; while AT-like-ir cells are

  • 16

    also seen in the optic lobes of the brain and in the anterior midgut as open type endocrine cells

    (Riccillo and Ronderos, 2010). Surprisingly, however, AT-like immunoreactivity is also

    observed in the Malpighian tubules (MTs) and the AT-like-ir content is regulated depending

    upon the feeding state of the insect (Santini and Ronderos, 2009). T. infestans, like R. prolixus,

    are blood gorging hemipterans that imbibe a large blood meal and are hence faced with the same

    short-term physiological challenges as R. prolixus; they need to rapidly undergo diuresis. Hence,

    based on the hypothesis that AT produced from the endocrine cells of the MTs regulates diuretic

    behaviour it was found that AT exerted a myotropic effect on hindgut contractions in a dose-

    dependent manner (Santini and Ronderos, 2007). Moreover, AT was a sexually dimorphic

    cardioacceleratory peptide at the level of the dorsal vessel and myostimulatory peptide upon the

    anterior midgut (Sterkel et al., 2010).

    THESIS OBJECTIVE

    The goal of this thesis was to identify and confirm the gene transcript, its relative tissue

    distribution and to determine any potential roles of AT in the blood-gorging hempiteran, R.

    prolixus. These objectives were achieved through in silico bioinformatics, in vitro molecular

    techniques supplemented with immunohistochemical analysis, and tissue bioassays. Moreover,

    much of the work stemmed from comparative analysis with previous literature that determined

    novel and peculiar roles of AT in T. infestans.

    Originally isolated as stimulators of JH, ATs have since been identified as multimodal

    neuropeptides that are well-represented among invertebrates. Allatotropins regulate a plethora of

    physiological functions in complex combinations of species, tissue, sex or developmental time,

    hence offering great complexity and multiplicity in function. Therefore, owing to its

    fundamental role in an array of biological process it becomes critical to obtain a comprehensive

    understanding of this family of neuropeptides that were once thought to be mere stimulators of

    JH biosynthesis.

  • 17

    MATERIALS AND METHODS

    Animals and tissues

    Rhodnius prolixus were obtained from a long-standing colony maintained at high relative

    humidity in 25oC incubators at the University of Toronto Mississauga. Insects were fed

    defibrinated rabbits’ blood (Cedarlane Laboratories Inc., Burlington, ON, Canada) at all instar

    stages. The blood meal initiates growth and development and the insect emerges into the next

    instar approximately 1-2 weeks post-feeding. Unfed (approx. 4-6 weeks) IVth

    instars were used

    for conducting all the experiments with the exception of hindgut assays where both unfed IVth

    and Vth instars were used. Additionally, the heart assays and RT-PCRs were conducted using

    only unfed Vth

    instars. All physiological assays were carried out in R. prolixus saline modified

    from Lane et al (1975); namely150mM NaCl, 8.6mM KCl, 2.0mM CaCl2, 4.0mM NaHCO3,

    8.5mM MgCl2, 5mM HEPES and 34mM glucose; pH 7.0. Tissues for molecular biology

    experiments were dissected in nuclease free phosphate buffered saline (PBS) (Sigma-Aldrich,

    Oakville, ON, Canada) and immediately placed in RNAse-free microcentrifuge tubes containing

    RNA laterTM

    RNA stabilization reagent (Qiagen Inc., Mississauga, ON, Canada).

    Construction of CNS cDNA

    Fresh IVth

    instar central nervous system (CNS) cDNA was used as a template for PCR reactions.

    Total CNS RNA was extracted using Trizol® reagent (Life Technologies Corporation,

    Carlsbad, CA, USA) and 400ng of extracted RNA was used for cDNA synthesis using the

    oligo(dT)20 primer provided by iScript™ Select cDNA Synthesis kit (Bio-Rad Laboratories

    Ltd., Mississauga, ON, Canada). The cDNA synthesis reaction was carried out using the

    following thermal cycler conditions: incubation at 25oC for 5 minutes, 42

    oC for 30 minutes and

    final 85°C for 5 minutes to heat-inactivate the reverse transcriptase. The final product was

    diluted 1:2 and stored at -20oC, an aliquot being used for subsequent PCR reactions as needed.

  • 18

    In-silico and in-vitro search of cDNA sequence encoding allatotropin prepropeptide

    Predicted allatotropin prepropeptide sequence (Ons et al., 2011) was used to tBLASTn search

    the Rhodnius genome using Geneious software 4.7.6 (Biomatters, Ltd., Aukland, New Zealand).

    The tBLASTn search yielded positive hits against contig 3631 and contig 64. The results

    obtained were then used to predict possible promoter regions and splice sites via online software

    provided by Berkeley Drosophila Genome Project. The possible signal peptide sequence was

    predicted using SignalP 4.1 software (Petersen et al., 2011).

    IVth

    instar CNS cDNA was screened for R. prolixus allatotropin (Rhopr-AT) gene using gene

    specific forward and reverse primers: Allatrp-For1, Allatrp-Rev5 (Table 3) in a Polymerase

    Chain Reaction (PCR). These primers successfully amplified the entire open reading frame

    (ORF) as well as segments of the 5’ and 3’ untranslated regions (UTRs). All PCRs were

    conducted using the s1000 Thermal Cycler (Bio-Rad Laboratories, Mississauga, ON, Canada),

    and the temperature cycling conditions were as follows: initial denaturation at 94oC for 3

    minutes, followed by 35 cycles of denaturation at 94oC for 30 seconds, annealing at 62

    oC for 30

    seconds, extension at 72o C for 1 minute, followed by 10 minute final extension at 72

    o C.

    Products obtained from the PCR were column purified using the AxyPrep™ PCR Clean-up Kit

    (Axygen Biosciences, Union City, CA, USA) and subsequently cloned into the pGEM®-T

    vector (Promega Corporation, Madison, WI, USA). Mach1™-T1R Chemically Competent E.

    coli cells (Life Technologies Corporation, Carlsbad, CA, USA) were transformed with the

    recombinant vector and grown on LB/Ampicillin/XGal medium for 12-16 hours. Successfully

    transformed colonies were selected; their plasmids purified and sent for sequencing at the Sick

    Kids DNA Sequencing Facility (The Centre for Applied Genomics, Hospital for Sick Children,

    Toronto, ON, Canada).

    The molecular techniques remained identical for the cloning of PCR products that were

    subsequently used for synthesis of RNA probes for Northern blot hybridisation, except for gene

    specific primers, annealing and extension temperatures and times. The primers used for

    amplification of this product were Allatrp-For1-Exon3 and Allatrp-Rev5 at an annealing

    temperature of 51oC for 30 seconds and an extension step at 72

    oC for 30 seconds. The rest of the

    thermal cycler conditions remained unchanged.

  • 19

    Table 3. Gene-specific primers used for Rhopr-AT prepropeptide cDNA amplification

    Oligonucleotide Name Oligonucleotide sequence (5’-3’)

    Allatrp-For1 GCATCAGTCGGAGTACAGA

    Allatrp-For1-Exon3 TGTCCATCGTCAGCATAC

    Allatrp-Rev4 AGTATGCTGACGATGGACAC

    Allatrp-Rev5 CAGATGAAATGACTTTTTAACAACAC

    rp49for1 (positive control) GTGAAACTCAGGAGAAATTGGC

    rp49Rev1(positive control) AGGACACACCATGCGCTATC

  • 20

    Phylogenetic Analysis of Rhopr-AT prepropeptide

    In order to identify homologous allatotropin sequences from other insect species a protein

    BLAST against other genomic databases was performed using Rhopr-AT prepropeptide

    sequence (Ac: ACS45387.1) as a query. The prepropeptide sequences were selected from the

    following species: Manduca sexta (Ac: AAB08757.1), Mythimna separata (Ac: ABA61322.1),

    Mythimna unipuncta (Ac: AAF23016.1), Spodoptera frugiperda (Ac: CAD32495.1),

    Spodoptera exigua (Ac: AEO27700.1), Helicoverpa armigera (Ac: AAR32789.1), Samia ricini

    (Ac: AAZ30065.1), Daphnia pulex (Ac: EFX71302.1), Clostera anastomosis (Ac:

    AAV59461.2), Aedes aegypti (Ac: AAB06179.1), Anopheles gambiae (Ac: Xm_320402.3),

    Bombyx mori (Ac: AAY40838.1), Tribolium castaneum (Ac: NP_001137204.1), Ixodes

    scapularis (Ac: EEC06620.1), predicted hypothetical protein: Nasonia vitripennis (Ac:

    XP_003427574.1), predicted hypothetical protein: Culex quinquefasciatus (Ac: EDS30439.1),

    predicted hypothetical protein: Acyrthosiphon pisum (Ac: XP_001942912.1), predicted

    hypothetical protein: Bombus impatiens (Ac: XP_003488345.1) and predicted hypothetical

    protein: Bombus terrestris (Ac: XP_003398476.1). Aforementioned sequences were used to

    conduct a subsequent alignment using ClustalOmega (http://www.ebi.ac.uk/Tools/msa/clustalo/)

    following which, the sequences were imported to BOXSHADE

    (http://www.ch.embnet.org/software/BOX_form.html) to construct the final image depicting

    conserved regions between amino acid sequences. The sequences along with splice variants, if

    any, were imported into MEGA 5.0 (Tamura et al., 2011) and a maximum parsimony

    phylogenetic tree was constructed using the Close-Neighbor-Interchange (CNI) analysis with a

    bootstrap value of 1000 iterations.

    Reverse Transcription-Polymerase Chain Reaction (RT-PCR) to elucidate spatial distribution

    of transcript

    Insects were dissected and the tissues pooled into 11 separate groups: 1) CNS, 2) dorsal vessel

    (DV), 3) foregut (FG), 4) salivary glands (SG), 5) fat body and trachea (FbTr), 6) anterior

    midgut (AM), 7) posterior midgut (PM), 8) Malpighian tubules (MT), 9) hindgut (HG), 10)

    immature testes (TT) and 11) immature ovaries (Ov). Total RNA from these tissues was

    extracted using the Trizol® reagent (Life Technologies Corporation, Carlsbad, CA, USA) and

    http://www.ebi.ac.uk/Tools/msa/clustalo/http://www.ch.embnet.org/software/BOX_form.html

  • 21

    cDNA was synthesised as described earlier. To elucidate the presence of Rhopr-AT mRNA

    transcript a 553bp fragment spanning all three exons was chosen. This region was amplified

    using the following primers: Allatrp-For1 and Allatrp-Rev4, while a positive control with the

    amplification of a 229bp ribosomal protein 49 (Rp49) was done using rp49for1 and rp49Rev1

    primers (Table 3). The thermal cycle conditions for the ensuing PCR reaction were as follows:

    initial denaturation at 94°C for 3 minutes, 35 cycles of denaturation at 94°C for 30 seconds,

    annealing at 57°C for 30 seconds, extension at 72°C for 30 seconds and final extension at 72°C

    for 10 minutes. Three independent biological replicates were done for the RT-PCR.

    Northern Blotting to determine transcript size

    Total CNS RNA from 100 insects was extracted using Trizol® Reagent (Life Technologies

    Corporation, Carlsbad, CA, USA). 2µg of RNA along with 2x RNA loading dye (Fermentas

    Canada Inc., Burlington, ON, Canada) was denatured at 75oC for 5 minutes and immediately

    chilled on ice. 30ng of RiboRuler™ High Range RNA Ladder (Fermentas Canada Inc.,

    Burlington, ON, Canada) with 2x RNA loading dye (Fermentas Canada Inc., Burlington, ON,

    Canada) was also prepared under simultaneous conditions. Samples were loaded on a 1% RNase

    free agarose gel containing formaldehyde and electrophoresed at 70V for 140 minutes. To

    ensure the RNA samples were separated sufficiently the gel was briefly viewed under UV light.

    Following UV analysis, a 20 minute wash with diethylpyrocarbonate (DEPC)-treated distilled

    water was done, and the RNA was transferred from the gel onto a positively charged nylon

    membrane (Roche, Mannheim, Germany) in 20x saline-sodium citrate (SSC) via overnight

    downward capillary transfer. The gel was briefly viewed under UV light again to ensure no

    residual RNA remained. The nylon membrane was briefly washed (4-5 seconds) in DEPC

    treated water to remove excess 20xSSC that could result in high background of the blot. Finally,

    the RNA was fixed to the membrane by UV cross-linking at 30, 000µJ/cm2 (UVP CL-1000,

    Upland, CA) and drying the membrane at room temperature for 2-3 hours. The blot was stored

    at 4oC until further use.

    Hybridization was conducted using the DIG Northern Starter Kit (Roche, Mannheim, Germany)

    with some modifications to the manufacturer’s protocol. Specifically, a digoxigenin (DIG)-

    labelled anti-sense RNA probe complementary to the 3’ UTR of Rhopr-AT mRNA transcript

    was synthesised for hybridisation with the membrane. Overnight linearization at 37oC of a 20µg

  • 22

    pGEM®-T vector, containing 426bp Rhopr-AT fragment, was done using SacII Restriction

    Enzyme. The reaction was then quenched with a water bath treatment at 65oC for 20 minutes

    and the linearized plasmid was PCR purified using the AxyPrep™ PCR Clean-up Kit (Axygen

    Biosciences, Union City, CA, USA). In vitro transcription of the 1µg of linearized plasmid

    using Sp6 polymerase from the DIG RNA labelling kit SP6/T7 (Roche Applied Science,

    Mannheim, Germany) followed. The DNA template was removed by incubating the reaction

    with 2µl of deoxyribonuclease I for 15 minutes at 37oC. The synthesised antisense RNA probe

    was stored at -20oC until further use. A DNA DIG-labelled RNA ladder probe was also

    synthesised according to the manufacturer’s protocol and stored at -20oC.

    The RNA-bound nylon membrane was pre-hybridized for 30 minutes at 68oC with pre-warmed

    hybridisation solution. Following pre-hybridisation, the membrane was hybridised for 16-18

    hours at 68oC with hybridisation solution containing100ng/mL SacII linearized RNA probe. To

    remove unbound probe and decrease background noise, the membrane was subjected to

    stringent washes: two washes with 2x SSC, 0.1% SDS at room temperature, followed by two

    washes with pre-warmed 0.5x SSC, 0.1% SDS at 68oC. Immunological detection of the

    membrane was done by exposing the Bioflex Scientific Imaging Films (Clonex Corporation,

    Markham, ON, Canada) to the chemiluminescence substance 15-30 minutes post application.

    Transcript size of the Rhopr-AT prepropeptide was determined by aligning it to the RNA ladder.

    Three independent biological replicates were performed.

    Immunohistochemistry to detect peptide localisation

    The dorsal cuticle and diaphragm of unfed IVth

    instar R. prolixus nymphs were removed to

    expose the digestive tract and CNS. Tissues were subsequently fixed in-situ by submerging the

    bug in 2% paraformaldehyde fixative (pH 7.0) for 18-24 hours at 4˚C. Excess fixative was

    washed off using phosphate buffered saline (PBS) - washes were done every 10 minutes for 2

    hours. The tissues (gut, Malpighian tubules, salivary glands and CNS along with peripheral and

    abdominal nerves) were dissected out and placed into respective 1.5 ml microcentrifuge tubes

    containing PBS. To increase permeability of tissues to the antiserum they were treated with 4%

    Triton X-100 in PBS with 2% bovine serum albumin (BSA) and 10% normal goat serum (NGS)

  • 23

    for 1 h at room temperature. Excess Triton X-100 was washed off with PBS and the tissues were

    incubated on a shaker for 48 hours at 4oC with antiserum made by rabbits against Manduca

    sexta AT (Manse-AT: GFKNVEMMTARGF-NH2, a gift from J. Veenstra, France). The

    antiserum had been pre-incubated for 18-24 hours at 4oC with a 0.4% solution of Triton X-100

    in PBS with 2% BSA and 2% NGS to obtain a 1:1000 dilution and to minimise non-specific

    binding. Following primary antiserum incubation, tissues were washed at room temperature for

    4-6 hours in PBS and then incubated on a shaker for 18 hours at 4oC with a secondary antibody

    of Cy3-labelled goat anti-rabbit diluted 1:600 with 10% NGS in PBS. Tissues were then washed

    with PBS repeatedly for 8 hours and finally mounted on microscope slides in 100% glycerol.

    Allatotropin-like immunoreactivity was observed and images were obtained using a Zeiss

    confocal laser microscope (Carl Zeiss, Jena, Germany) and Zen 2009 viewing software.

    Allatotropin-like immunoreactivity within the CNS was mapped using an epifluorescence

    microscope with a drawing tube attached. Controls whereby primary antiserum was pre-

    incubated with 10-5

    M Rhopr-AT for 24 hours yielded no staining.

    Physiological Roles of Allatotropin: Hindgut Contraction Assay

    A horizontal incision along the thoracic segment and two lateral incisions along the dorsal

    cuticle of R. prolixus were done to expose the tissues. Tissues were bathed in 100µl R. prolixus

    saline. Dorsal diaphragm, dorsal vessel, Malpighian tubules, fat body and trachea were removed

    sequentially in an effort to obtain a clean preparation consisting of just the gut. Then, a silk

    thread was used to tie a double knot at the anterior end of the hindgut (at the junction of

    posterior midgut and hindgut) and the rest of the anterior digestive tract removed. The hindgut,

    attached to the ventral cuticle at the anus, was removed and pinned in a well containing 200µl of

    saline in a Sylgard-coated Petri dish. The free end of the silk thread was used to tie a double

    knot around the hook of the force transducer (Aksjeselskapet Mikroelektronikk, Horten,

    Norway). The knots and the resulting thread were tied at a 90o angle with the hook to increase

    efficiency of the force transducer. The hindgut was stretched slightly to mimic its natural

    physiological state. Longitudinal contractions exhibited by the tissue were measured using

    PicoScope 2202 and recorded with PicoLog (Pico Technology, Cambridgeshire, UK). The

    hindgut was washed with saline and left to bathe and equilibrate in saline for about 20 minutes

  • 24

    prior to starting the experiment. Synthetic Rhopr-allatotropin (Table 4) was stored at -20 oC as

    5µl aliquots of 10-3

    M and serially diluted with saline to obtain the desired applied dose for the

    tissue. All doses were made and kept on ice at the beginning of the experiment, and brought

    back to room temperature prior to application. Allatotropin doses ranged from 1x 10-16

    M to

    1x10-6

    M and were applied in a random order. For application of a dose, 100µl of saline was

    removed and 100µl of saline solution containing twice the desired concentration of peptide was

    added simultaneously to maintain the total volume of 200µl in the well. The change in basal

    tonus from saline incubation to peptide application was measured and then the dose was washed

    off with saline until the basal tonus returned to pre-dose conditions. The experiment was

    repeated with at least 5 independent hindgut preparations. Data were analysed using GraphPad

    Prism.

    To verify the viability of the preparation and to determine the relative effects of allatotropin

    compared to a known stimulator of hindgut contractions, Rhopr-kinin 2 (Table 4) (dose range:

    10-11

    M to 10-6

    M) was also applied on the same prep. Furthermore, the same doses of Rhopr-AT

    and Rhopr-kinin were also tested on Vth

    instar hindguts to deduce if any differences in effect

    existed between the two instar stages.

  • 25

    Table 4. Amino acid sequence and purity of peptides used for the bioassays

    Peptide Name Peptide Sequence Peptide Purity

    Rhopr-AT NVQLSTARGF-amide >98%

    Rhopr-Kinin 2 AKFSSWG-amide >95%

    Proctolin RYLPT >83%

  • 26

    Physiological Roles of Allatotropin: Heart Impedance Assay

    The legs were removed and the bug was pinned dorsal side down in Sylgard-coated Petri dish.

    Lateral incisions along the ventral cuticle and a single horizontal incision along the ventral

    thoracic segment enabled smooth removal of the ventral cuticle. Following this, the ventral

    diaphragm and digestive tract were also removed to expose the dorsal vessel and associated

    alary muscles which were attached to the dorsal cuticle and diaphragm. Tissues were bathed in

    200µl of saline. The experimental set up consisted of minuten pin electrodes on either side of

    the heart at the junction of the 5th

    and 6th

    abdominal segments. The electrodes measured the

    change in resistivity caused by every beat of the heart. This change in resistivity resulting in

    impedance of electric flow between the electrodes was amplified using an impedance converter

    (UFI model 2991, Morro Bay, CA, USA), measured with a PicoScope 2202 and recorded with

    PicoLog Software (Pico Technology, Cambridgeshire, UK). Since the change in impedance is

    due to the heart beat, every deflection upon the PicoScope chart is indicative of a heartbeat. The

    heart was allowed to acclimatize to the experimental set up for about 15 minutes before starting

    the experiment. Allatotropin doses (10-12

    to 10-6

    M) were made as described for the hindgut assay

    and tubes were kept on ice until application upon the preparation. Doses were applied in a

    random order, removing and adding the same volumes as mentioned earlier for the hindgut

    assays. Positive controls using the myotropic pentapeptide, proctolin (Table 4) (10-10

    and 10-9

    M)

    were also done to confirm viability of the preparation.

  • 27

    RESULTS

    Characterisation of cDNA and determination of Rhopr-AT transcript length

    An in-silico BLAST search followed by cloning of the Rhopr-AT gene using gene specific

    primers revealed a cDNA region of 973bp (Figure 2A). The cloned cDNA was identical to the

    predicted sequence by Ons et al. (2011; Ac: GQ162783). The cDNA spans over three exons:

    83bp, 211bp and 680bp in length. The first exon, situated on contig 3631 contains the

    transcription start site at the 5’ UTR; while the latter two exons, on contig 64, contain the ORF

    encoding an allatotropin prepropeptide of 119 amino acid residues and fragments of the 3’ UTR

    (Figure 2B). The mature AT-peptide is 13 amino acids long, between residues 45 and 58 and is

    identical to the sequence determined by MALDI-TOF mass spectrometry of extracts of R.

    prolixus brains (Ons et.al., 2011; Sterkel et.al., 2011). Possible endoproteolytic cleavage sites:

    Arg44

    , Lys59

    Arg60

    , flank the mature AT-peptide that ends with a glycine residue; the signal for

    carboxyterminal amidation by peptidyl-glycine-α-amidating monooxygenase (Eipper et. al.,

    1992). Another possible endoproteolytic cleavage site: Arg93

    and Arg94

    downstream the mature

    peptide has also been identified but the resulting predicted peptide has not been identified in

    extracts of the CNS, and so it is not known if the cleavage site is functional. Analysis of

    transcript size via a Northern blot yielded a single band of approximately 1.4kb (Figure 2C).

    Based on this analysis and in-silico search, it is predicted that approximately 427bp of the

    3’UTR is missing, which must also contain the polyadenylation signal (AATAAA).

  • 28

    Figure 2. A) Nucleotide sequence of allatotropin (AT) precursor peptide cDNA in R. prolixus.

    The entire open reading frame along with the deduced amino acid sequence is shown directly

    below the codons. Initial methionine start codon is capitalized, bolded and double underlined

    while the stop codon is bolded. Nucleotides preceding the initial start codon and following the

    stop codon represent the 5’ and 3’ untranslated regions (UTRs) respectively. Dashed line

    represents the 5’ transcription start site, while the possible signal peptide sequence is double

    underlined with the possible cleavage site indicated by a bolded asterisk. The boxed region

    depicts the mature AT peptide, with the possible dibasic proteolytic cleavage sites shaded in

    grey. C terminal glycine residue (Gly58) required for amidation is single underlined. B)

    Schematic representation of Rhopr-AT gene organization. The gene is comprised of three

    exons- depicted by boxes representing number of nucleotides contained in each; and two

    introns- depicted by solid lines. The start (ATG) and stop (TGA) codons are represented by

    upward arrows. C) Northern blot of Rhopr-AT transcript hybridised with a complementary

    digoxigenin-labelled anti-sense RNA probe 426bp in length. The approximate size of the

    transcript was determined using markers shown on the right.

  • 29

  • 30

    Rhopr-AT prepropeptide and phylogenetic relationship

    The ClustalO alignment represents a high degree of conservation for the 13 residue mature

    allatotropin peptide sequence. The Rhopr-AT sequence is approximately 75% identical, with a

    shared identity of 10 out of 13 residues, to the lepidopteran species - all of which share an

    identical mature allatotropin sequence (Table 1, Figure 3). The mature allatotropin sequence

    terminates with the motif TARGF which is also highly conserved among insects; exceptions

    being, N. vitripennis, B.impatiens and B. terrestris predicted sequences where the amino acid R

    is replaced by Y and T. castaneum where terminal F is replaced by Y. The C-terminal motifs of

    the prepropeptides also have a high degree of conservation among species of different taxa

    (Figure 3). Species such as R. prolixus, D. pulex and A. aegypti have a single prepropeptide

    encoding the mature allatotropin, while lepidopteran species generally have multiple

    prepropeptides encoding the mature AT sequence along with Allatotropin-like peptides (Table 1

    and 2).

    Moreover, lepidopteran species also form a monophyletic group when placed on a phylogenetic

    tree. The hypothetical and currently unidentified allatotropin prepropeptide sequences of the

    Bombus species form a monophyletic group with the predicted N. vitrepennis allatotropin

    sequence, while R. prolixus allatotropin prepropeptide forms a monophyletic group with

    Culicidae (Figure 4). The arthropod T. castaneum and mollusc A. california form separate

    clades rooting the tree.

    RT-PCR

    CNS and peripheral tissues were dissected from 10 Vth

    instar R. prolixus. RNA was extracted

    using Trizol Reagant and cDNA for individual tissues was synthesised. An aliquot of this

    single-stranded cDNA was used as a template in a subsequent PCR reaction used to amplify a

    553bp segment of Rhopr-AT using gene specific primers. Rp49 was used as the reference gene.

    Highest level of expression was observed in the CNS and pool of fat body trachea (Figure 5);

    the latter suggesting the presence of peripheral neuroendocrine cells expressing allatotropin. In

    only one trial, faint bands were also seen in the immature testes.

  • 31

    Figure 3. ClustalO alignment of Rhopr-AT prepropeptide sequence with allatotropin

    prepropeptide sequences from other arthropods. Identical amino acid sequences are shaded in

    black, while similar amino acids are highlighted in grey. The mature allatotropin sequence is

    denoted by a red bar above. The following sequences were used to create the alignment:

    Manduca sexta I, Mythimna separata, Mythimna unipuncta, Spodoptera frugiperda I,

    Spodoptera exigua, Helicoverpa armigera I, Samia ricini I, Daphnia pulex, Clostera

    anastomosis, Aedes aegypti, Anopheles gambiae, Tribolium castaneum, Bombyx mori,

    predicted hypothetical protein: Nasonia vitripennis, predicted hypothetical protein: Culex

    quinquefasciatus, predicted hypothetical protein: Acyrthosiphon pisum, predicted hypothetical

    protein: Bombus impatiens, predicted hypothetical protein: Bombus terrestris.

  • 32

  • 33

    Figure 4. Phylogenetic relationship of allatotropin prepropeptide sequence with other insect

    sequences. The maximum parsimonius tree was constructed using Close-Neighbor-Interchange

    (CNI) analysis where each node represents a percentage corresponding to 1000 bootstrap

    replicates. The analysis involved 27 amino acid sequences and was conducted in MEGA5.

  • 34

  • 35

    Figure 5. RT-PCR representing the expression profile of the Rhopr-AT transcript in unfed Vth

    -

    instar R. prolixus. N = 3. Tissues were dissected and the total RNA extracted from 10 insects.1)

    CNS, 2) dorsal vessel, 3) foregut, 4) salivary glands, 5) fat body and trachea, 6) anterior midgut,

    7) posterior midgut, 8) Malpighian tubules, 9) hindgut, 10) immature testes, 11) immature

    ovaries, 12) No template control. A 229bp ribosomal protein 49 (Rp49) was also amplified as a

    positive control to test the quality of the cDNA.

  • 36

  • 37

    Immunohistochemistry - Phenotypic expression of allatotropin-like immunoreactivity in the

    CNS and peripheral tissues

    CNS

    Intense allatotropin-like immunoreactivity was observed throughout the CNS and some

    peripheral tissues of R. prolixus, with the greatest number of intensely-stained cell bodies being

    present on the dorsal surface of the brain (Figure 6A, 8A). A cluster of approximately 30-35

    cells along the dorsal lateral (dl) regions of each sub-optic lobe (ol) was observed. Varicose

    axon terminals were seen in the optic lobes, though the origin of these projections could not be

    traced. Moreover, a cluster of 10 bilaterally-paired cells were seen along the dorsal midline (dm)

    of the protocerebrum and 3 bilaterally-paired cells were seen along the dorsal anterior midline

    (dam) of the protocerebrum (Figure 6A, 8A). The dm cells are likely median neurosecretory

    cells. Furthermore, 3 bilaterally-paired dorsal posterior medial cells (dpm) and 2 bilaterally-

    paired dorsal posterior lateral cells (dpl) were seen (Figure 6A, 8A). Most of the cells were seen

    in the protocerebrum with a few in the tritocerebrum. The ventral surface of the brain had fewer

    cells in comparison (Figure 8B). These include clusters of 8 bilaterally-paired cells along the

    ventral medial region (vm) in close proximity to 6 bilaterally-paired cells anterior to the

    tritocerebrum surrounding the oesophageal canal. Three bilaterally-paired posterior-lateral cells

    (pl) were also present in the tritocerebrum. Finally, a cluster of 8 cell bodies at the base of the

    optic lobes (ol) were observed in each hemisphere (Figure 8B).

    Characteristic bilaterally-paired cell bodies were also observed in each ganglion: the

    suboesophageal ganglion (SOG), prothoracic ganglion (PRO) and mesothoracic ganglionic mass

    (MTGM). Cells in the SOG were concentrated along the midline; however some cell clusters

    were also seen along the lateral surfaces (Figure 6B, 8A). Particularly, 2 distinct pairs of dorsal-

    medial (dm) intensely-stained cells (whose projections travelled anteriorly and then laterally

    before being lost in the neuropil) were seen dotted by approximately 7 posterior smaller cells at

    the dorsal posterior medial region (pm) of the SOG (Figure 6B, 8A). Along the ventral surface 2

    brightly-stained cells, surrounded by smaller lightly stained cells were seen anterior to the

    neuropil; while 4 pairs of dorso-ventral (dv) bilaterally-paired cells bordered the dorsal medial

    intense staining cells (Figure 6B, 8A). Clusters of cells were also seen just posterior to the brain

    and a pair of bilaterally-paired cells was seen at the posterior medial region (pm) of the SOG

  • 38

    (Figure 8A). Furthermore, varicose axonal processes originating from the posterior SOG

    traverse through the PRO and MTGM where they bifurcate and give rise to dense neuropil

    regions (Figure 6D, 7A, 8A). These projections are contained within the CNS in the PRO and

    SOG as they could not be traced into peripheral nerves, hence suggesting they are interneurons.

    In the PRO, 6 anterior-lateral cells (al) and 2 pairs of anterior-medial cells (am) were seen on the

    dorsal surface (Figure 6D, 8A), and 2 pairs of anterior-medial cells (am) on the ventral surface

    (Figure 8A). Bilaterally-paired ventral medial (vm) and posterior ventral medial (pvm) cells

    were also observed in the PRO in close proximity of the axonal projections (Figure 6D, 8A).

    The MTGM was more densely populated with cells than the PRO with cell clusters seen around

    the axonal processes, which again terminated in dense neuropil regions within the ganglion

    (Figure 7A, 8A). Bilaterally-paired cell bodies were observed along the anterior region of the

    MTGM. Moreover, a cluster of 8 intensely staining bilaterally-paired cells was seen at the

    posterior dorsal surface (pd) of the MTGM and 4 pairs of 3 cells each were also seen at the

    posterior of the MTGM (Figure 7A, 7B, 8A).

    Periphery

    Axonal projections exited the brain and arborized over the retrocerebral complex (CC/CA)

    before entering the dorsal vessel (DV) (Figure 6C, 7C). Neurohaemal processes were also

    observed on the 2nd

    and 3rd

    abdominal nerve of the MTGM (Figure 7B, 7E, 8A), as well as on

    the trunk nerves (Figure 7D, 8A). The neurohaemal processes observed on the CC/CA, DV

    (Figure 7C), abdominal and trunk nerves (Fig. 7B, 7E, 7D) suggest possible neurohormonal

    roles for AT in R. prolixus. The accessory duct of the salivary gland showed staining for AT-

    like-ir processes across its length, which then proliferated over the principal gland (Figure 9E).

    Furthermore, some faint staining was observed over the hindgut (Figure 9A, 9B). However, a

    lack of staining in the Malpighian tubules (Fig. 9A, 9C, 9D) challenges the recent report of AT-

    like peptide production by the tubules and staining as reported in another triatomine bug,

    Triatoma infestans (Santini et. al., 2007, 2009; and see later under Discussion).

  • 39

    Figure 6. Confocal images showing AT-like immunostaining in cells of IVth

    instar Rhodnius

    prolixus CNS, corpus cardiacum and dorsal vessel. A) Brain and anterior suboesophageal

    ganglion (SOG). B) SOG. C) Corpus Cardiacum (CC) and dorsal vessel (DV). D) Prothoracic

    ganglion (PRO). Arrows point to CC and DV. Scale bars represent 100µm.

  • 40

  • 41

    Figure 7. Confocal images showing AT-like immunostaining in cells of IVth

    instar Rhodnius

    prolixus CNS and peripheral nerves. A) Mesothoracic ganglionic mass (MTGM). B) Abdominal

    nerves exiting the MTGM. C) Dorsal vessel (DV). D) Trunk nerve. E) Abdominal nerves. AT-

    like immunoreactivity along the CC, DV and nerves is depicted by arrows. N = 60. Scale bars

    represent 100µm.

  • 42

  • 43

    Figure 8. Camera lucida map depicting distribution of AT-like immunoreactive cells within the

    CNS. Open cells are best seen from the ventral surface, while filled cells are best seen from the

    dorsal surface. Scale bar represents 200µm. A) Dorsal view. B) Ventral view of the brain. Some

    groups have been labelled for clarity in text description. dam = dorsal anterior midline, dm =

    dorsal midline, dl = dorsal lateral, ol = optic lobe, dpl = dorsal posterior lateral, dpm= dorsal

    posterior medial, dv = dorso-ventral, pm= posterior medial, al = anterior lateral, am = anterior

    medial, pd = posterior dorsal, pl = posterior lateral, vm = ventral medial, pvm = posterior

    ventral medial.

  • 44

  • 45

    Figure 9. Confocal images showing AT-like immunoreactivity in peripheral tissues of IVth

    instar

    Rhodnius prolixus. A) Posterior midgut (PMG), Malpighian tubules (MT) and hindgut (HG). B)

    HG. C) PMG and MT. D) Magnified MT. E) Salivary gland (SG). AT-like immunoreactive

    processes are depicted by arrows while areas devoid of staining (notably the MTs) are depicted

    by arrowheads. Scale bar represents 100 µm. N= 60.

  • 46

  • 47

    Hindgut Assay via Force Transducer

    The physiological role of Rhopr-AT upon the hindgut was tested by measuring changes in

    longitudinal contractions of the preparation using a force transducer. This tissue was chosen

    since there are some faint AT-like immunoreactive processes along its surface. Application of

    Rhopr-AT at doses ranging from 10-15

    M to 10-6

    M resulted in no significant change from the

    basal conditions (Figure 10, 11). To test that the bioassay was viable and robust, and the

    negative results valid, a positive control was performed. The application of another myoactive

    peptide, Rhopr-kinin 2 (10-11

    M to 10-6

    M), was able to induce forceful contractions of the

    hindgut (Figure 10, 11), with maximum effect at 10-8

    M Rhopr-kinin 2. Rhopr-AT was also

    tested on Vth

    instar hindgut to confirm the negative results obtained from IVth

    instar hindguts.

    Doses of Rhopr-AT also failed to elicit any significant change in contractions. The negative

    results obtained were again validated using Rhopr-Kinin 2 (10-11

    M to 10-6

    M). A sample trace for

    both peptides at the same dose is shown in Figure 10.

    Heart Impedance Assay

    Rhopr-AT (10-12

    M to 10-6

    M) had no significant effect on heart-beat frequency. No notable

    change in frequency was seen after the application of a Rhopr-AT (Figure 12). The myotropic

    pentapetide proctolin, however, did yield a large increase in frequency, as seen in Figure 12B.

  • 48

    Figure 10. Sample traces depicting effects of different peptide doses on IVth

    and Vth

    instar

    hindguts. Upward arrowhead represents the application of a dose, while the downward arrow

    represents saline wash. A and B represent the effect of 10-7

    M Rhopr-AT and 10-7

    M Rhopr-Kinin

    2 upon IVth

    instar hindgut respectively. C and D represent the effect of 10-7

    M Rhopr-AT and 10-

    7M Rhopr-Kinin 2 upon V

    th instar hindguts. Large deflections after wash off indicate additional

    aggressive washing. N=5.

  • 49

  • 50

    Figure 11. Dose-response curves representing the change in longitudinal contractions/tension of

    IVth

    instar R. prolixus hindgut after application of different doses of Rhopr-AT and Rhopr-kinin

    2. N=5.

  • 51

    10- 1 6 10- 1 4 10- 1 2 10- 1 0 10- 8 10- 6 10- 40

    5

    10

    15

    20

    25Rhopr-AT

    Rhopr-Kinin 2

    Dose (M)

    Ten

    sio

    n (

    mg

    )

  • 52

    Figure 12. A) Dose-response curve for Rhopr-AT activity on heart-beat of R. prolixus. No

    significant change in frequency was observed. N=5. B) Sample traces of spontaneous heart-beat

    in saline, Rhopr-AT and the myoactive pentapeptide, proctolin. Scale bar represents 30s.

  • 53

    A)

    B)

  • 54

    DISCUSSION

    Recent availability of vector genomes and EST databases has facilitated the process of

    identification and isolation of genes that encode neuropeptides, neurohormones and

    neuromodulators within species. Using an in-silico approach the allatotropin gene for the

    triatomine bug, Rhodnius prolixus, was identified and subsequently cloned to reveal a cDNA

    fragment of 973bp encoding one mature amidated allatotropin peptide. The amidated

    tridecapeptide is identical to the Rhopr-AT sequence determined by MALDI-TOF mass

    spectrometry of extracts of R. prolixus brains (Ons et.al., 2011; Ons et.al., 2009). A Northern

    blot further certified the presence of a single mRNA encoding a single mature allatotropin

    peptide, 1.4kb in length. Absence of multiple bands in the Northern blot, along with single hits

    from the BLAST results also confirms that Rhopr-AT is a single transcript encoded by one gene

    without splice variants.

    The expression profile for Rhopr-AT via RT-PCR revealed high transcript levels in the CNS and

    pool of fat body and trachea alluding to the presence of possible peripheral neuroendocrine cells

    that also express allatotropin. Furthermore, immunohistochemical analysis of R. prolixus tissues

    displayed AT-like-ir cells well-dispersed throughout the CNS along with AT-like-ir processes

    projecting to peripheral tissues. Faint staining was observed over the posterior midgut and

    hindgut, while no staining was observed in the Malpighian tubules. Allatotropin-like

    immunoreactive axons in the salivary nerve traverse along the accessory duct before forming a

    nerve plexus over the principal gland of the salivary gland.

    Contraction assays with the force transducer, to determine the effect of Rhopr-AT upon the

    hindgut yielded negative results for both IVth

    and Vth

    instars. To verify that the negative results

    obtained were due to inactivity of Rhopr-AT and no other artefact, a positive control using

    Rhopr-kinin 2 was performed. The myotropic peptide, Rhopr-kinin 2 was able to induce robust

    longitudinal contractions in the hindguts of both IVth

    and Vth

    instars. Considering the elaborate

    nerve plexus over the dorsal vessel, it was postulated that Rhopr-AT might stimulate

    contractions of the heart. Interestingly though, no such effect was observed. The negative results

    yielded from the heart impedance assay were validated with the myotropic pentapeptide

    proctolin, which was able to dramatically increase the frequency of contractions. The

  • 55

    allatotropic activity of Rhopr-AT on JH production cannot be tested at this time since the R.

    prolixus JH is yet to be chemically identified.

    Allatotropins (ATs) first isolated from the heads of adult Manduca sexta (Kataoka et al., 1989)

    have since been isolated and characterised extensively in several other lepidopteran species. The

    AT gene is transcribed as multiple mRNA isoforms within most lepidopterans; each isoform

    differing from another as a consequence of alternative splicing and encoding the mature

    amidated tridecapeptide AT along with allatotropin-like peptides (ATL) (Abdel-latief et al.,

    2003; Nagata et al., 2012; Sheng et al., 2007; Taylor et al., 1996; Truesdell et al., 2000; Yin et

    al., 2005). The phenomenon of multiple AT mRNA isoforms is limited to the lepidopteran order

    and characterization and prediction of AT in various oth