+ All Categories
Home > Documents > The drosophila angiotensin-converting enzyme homologue Ance is required for spermiogenesis

The drosophila angiotensin-converting enzyme homologue Ance is required for spermiogenesis

Date post: 08-Dec-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
10
The Drosophila angiotensin-converting enzyme homologue Ance is required for spermiogenesis Debra Hurst, a Caroline M. Rylett, b R. Elwyn Isaac, b and Alan D. Shirras a, * a Department of Biological Sciences, Lancaster University, Lancaster, LA 1 4YQ, UK b Department of Biology, University of Leeds, Leeds, LS 2 9JT, UK Received for publication 3 October 2002, revised 12 November 2002, accepted 13 November 2002 Abstract The Angiotensin-converting enzyme (Ance) gene of Drosophila melanogaster is a homologue of mammalian angiotensin-converting enzyme (ACE), a peptidyl dipeptidase implicated in regulation of blood pressure and male fertility. In Drosophila, Ance protein is present in vesicular structures within spermatocytes and immature spermatids. It is also present within the lumen of the testis and the waste bag, and is associated with the surface of elongated spermatid bundles. Ance mRNA is found mainly in large primary spermatocytes and is not detectable in cyst cells. Testes lacking germ cells have reduced levels of ACE activity, and no Ance protein is detectable by immunocy- tochemistry, indicating that the germ cells are the major site of Ance synthesis. Ance mutant testes lack individualised sperm and have very few actin-based individualisation complexes. Spermatid nuclei undergo scattering along the cyst and have abnormal morphology, similar to other individualisation mutants. Mutant spermatids also have abnormal ultrastructure with grossly defective mitochondrial derivatives. The failure of Ance mutant testes to form individualisation complexes may be due to a failure in correct spermatid differentiation. Taken together, the expression pattern and mutant phenotype suggest that Ance is required for spermatid differentiation, probably through the processing of a regulatory peptide synthesised within the developing cyst. © 2003 Elsevier Science (USA). All rights reserved. Keywords: Ance; Metallopeptidase; Spermatogenesis; ACE; Spermatid individualisation Introduction The testes of Drosophila offer an ideal system for the study of spermatogenesis. All stages are present simulta- neously and can be observed in simple squashes. For re- views of Drosophila spermatogenesis and its genetic con- trol, see Fuller (1993, 1998). Germ line stem cells at the apex of the testis undergo mitotic divisions. Cells which enter the spermatogenic pathway first undergo 4 incomplete mitotic divisions to generate a cyst of 16 cells connected by cytoplasmic bridges. This cyst becomes associated with two somatically derived cyst cells, which envelop the cyst and accompany it throughout spermatogenesis. The germ cells of the cyst enlarge to become primary spermatocytes, which undergo meiosis and pass down the testis to generate a cyst of 64 spermatids. As the spermatids move towards the base of the testes, they undergo a series of complex differentia- tion events. The head end of the cyst becomes embedded in the testis wall at the base of the testis, and the spermatids undergo a dramatic elongation to become approximately 2 mm in length, extending almost the entire length of the testis. Spermatids then undergo individualisation, whereby each becomes invested in its own plasma membrane and excess cytoplasm is removed. This process is carried out by a cytoskeletal–membrane structure known as the individu- alisation complex (IC). Each IC is composed of 64 “invest- ment cones” which form at the head end of the cyst and then move as a unit towards the tail end, encasing spermatids in their own plasma membrane as they move. Excess cyto- plasm is removed to a waste bag, which is situated at the tail end of the cyst when individualisation is complete. Cysts, containing mature sperm, then coil towards the base of the testis, along with the waste bag. Sperm are released from the * Corresponding author. Fax: 44-1524-843854. E-mail address: [email protected] (A.D. Shirras). R Available online at www.sciencedirect.com Developmental Biology 254 (2003) 238 –247 www.elsevier.com/locate/ydbio 0012-1606/03/$ – see front matter © 2003 Elsevier Science (USA). All rights reserved. doi:10.1016/S0012-1606(02)00082-9
Transcript

The Drosophila angiotensin-converting enzyme homologue Anceis required for spermiogenesis

Debra Hurst,a Caroline M. Rylett,b R. Elwyn Isaac,b and Alan D. Shirrasa,*a Department of Biological Sciences, Lancaster University, Lancaster, LA 1 4YQ, UK

b Department of Biology, University of Leeds, Leeds, LS 2 9JT, UK

Received for publication 3 October 2002, revised 12 November 2002, accepted 13 November 2002

Abstract

The Angiotensin-converting enzyme (Ance) gene of Drosophila melanogaster is a homologue of mammalian angiotensin-convertingenzyme (ACE), a peptidyl dipeptidase implicated in regulation of blood pressure and male fertility. In Drosophila, Ance protein is presentin vesicular structures within spermatocytes and immature spermatids. It is also present within the lumen of the testis and the waste bag,and is associated with the surface of elongated spermatid bundles. Ance mRNA is found mainly in large primary spermatocytes and is notdetectable in cyst cells. Testes lacking germ cells have reduced levels of ACE activity, and no Ance protein is detectable by immunocy-tochemistry, indicating that the germ cells are the major site of Ance synthesis. Ance mutant testes lack individualised sperm and have veryfew actin-based individualisation complexes. Spermatid nuclei undergo scattering along the cyst and have abnormal morphology, similarto other individualisation mutants. Mutant spermatids also have abnormal ultrastructure with grossly defective mitochondrial derivatives.The failure of Ance mutant testes to form individualisation complexes may be due to a failure in correct spermatid differentiation. Takentogether, the expression pattern and mutant phenotype suggest that Ance is required for spermatid differentiation, probably through theprocessing of a regulatory peptide synthesised within the developing cyst.© 2003 Elsevier Science (USA). All rights reserved.

Keywords: Ance; Metallopeptidase; Spermatogenesis; ACE; Spermatid individualisation

Introduction

The testes of Drosophila offer an ideal system for thestudy of spermatogenesis. All stages are present simulta-neously and can be observed in simple squashes. For re-views of Drosophila spermatogenesis and its genetic con-trol, see Fuller (1993, 1998). Germ line stem cells at theapex of the testis undergo mitotic divisions. Cells whichenter the spermatogenic pathway first undergo 4 incompletemitotic divisions to generate a cyst of 16 cells connected bycytoplasmic bridges. This cyst becomes associated with twosomatically derived cyst cells, which envelop the cyst andaccompany it throughout spermatogenesis. The germ cellsof the cyst enlarge to become primary spermatocytes, whichundergo meiosis and pass down the testis to generate a cyst

of 64 spermatids. As the spermatids move towards the baseof the testes, they undergo a series of complex differentia-tion events. The head end of the cyst becomes embedded inthe testis wall at the base of the testis, and the spermatidsundergo a dramatic elongation to become approximately 2mm in length, extending almost the entire length of thetestis. Spermatids then undergo individualisation, wherebyeach becomes invested in its own plasma membrane andexcess cytoplasm is removed. This process is carried out bya cytoskeletal–membrane structure known as the individu-alisation complex (IC). Each IC is composed of 64 “invest-ment cones” which form at the head end of the cyst and thenmove as a unit towards the tail end, encasing spermatids intheir own plasma membrane as they move. Excess cyto-plasm is removed to a waste bag, which is situated at the tailend of the cyst when individualisation is complete. Cysts,containing mature sperm, then coil towards the base of thetestis, along with the waste bag. Sperm are released from the

* Corresponding author. Fax: �44-1524-843854.E-mail address: [email protected] (A.D. Shirras).

R

Available online at www.sciencedirect.com

Developmental Biology 254 (2003) 238–247 www.elsevier.com/locate/ydbio

0012-1606/03/$ – see front matter © 2003 Elsevier Science (USA). All rights reserved.doi:10.1016/S0012-1606(02)00082-9

encasing cyst cells and enter the seminal vesicle. The cystcells and contents of the waste bag are broken down.

Hormonal control of spermatogenesis has been exten-sively studied in mammals and is relatively well under-stood. Spermatogenesis in insects is also known to be underhormonal control, but much less is known about paracrineand autocrine signalling processes within the testes them-selves. In both mammals and insects, spermatogenesis issupported by somatic cells, which surround the developinggerm cells—Sertoli cells in mammals and cyst cells ininsects. There is evidence for two-way paracrine communi-cation between Sertoli cells and germ cells, which is re-quired for Sertoli cell function and sperm maturation. Someof the components of this signalling process have beenidentified. For example, nerve growth factor is secreted byrat round spermatids and is required for Sertoli cell viability(Chen et al., 1997). Peptidases known to be important forthe processing of physiologically active small peptides havebeen shown to be synthesised by Sertoli cells, implicatingthese molecules in the regulation of spermatogenesis (Mon-sees et al., 1998). Furthermore, elements of the tissue kal-likrein–kinin system have been identified in rat testes (Mon-sees et al., 1999). Kallikrein, the serine protease responsiblefor converting kininogen to bradykinin, is present in sper-matids, but absent from Sertoli cells. A number of pepti-dases which will cleave, and inactivate, bradykinin are as-sociated with Sertoli cell membranes (Monsees et al., 1999).These include somatic angiotensin-converting enzyme(ACE), neutral endopeptidase (NEP), and metallocar-boxypeptidase. A novel, soluble NEP, NL1, has been iden-tified in the round and elongated spermatids of mouse testes(Ghaddar et al., 2000). Two testes-specific peptidases (ger-minal ACE and PC4, a prohormone convertase) have beenshown to be vital for normal male fertility from gene knock-out experiments in mice (Krege et al., 1995; Hagaman et al.,1998; Esther et al., 1996; Mbikay et al., 1997). A thirdpeptide processing enzyme (N-arginine dibasic convertase,NRD, or nadrilysin) accumulates in the cytoplasm of elon-gated mammalian spermatids, where it is believed to have arole in germ cell differentiation (Chesneau et al., 1996).Taken together, these findings suggest that small bioactivepeptides play a paracrine or autocrine role in regulatingmammalian spermatogenesis. The substrates for mamma-lian germ cell peptidases are not, however, known, andtherefore the precise roles of these enzymes and their sub-strates in mammalian spermatogenesis remain to be eluci-dated.

The discovery of ACE activity in the testes of manyinsect species from different orders suggests that regulationof spermatogenesis by peptide processing may be wide-spread and evolutionarily conserved (Isaac et al., 1999).Drosophila melanogaster has six ACE-like genes, two ofwhich (Ance and Acer) code for active enzymes (Tatei et al.,1995; Cornell et al., 1995; Taylor et al., 1996; Houard et al.,1998). These differ from the somatic form of the mamma-lian enzyme in that they consist of a single catalytic domain

and do not possess a C-terminal hydrophobic membraneanchor. Ance mutants were previously reported to be lethalwith escapers showing male sterility (Tatei et al., 1995). Wereport here that Ance is expressed in Drosophila testes andthat it is required for spermatid differentiation and individu-alisation.

Materials and methods

Drosophila strains

Wild type Oregon R and mutant strains were maintainedon oatmeal/molasses/agar medium at 25°C. Ance34Eb1 andAnce34Eb2 alleles were originally isolated in a saturationmutagenesis of the Adh region (Woodruff and Ashburner,1979) and were obtained from John Roote, Department ofGenetics, University of Cambridge (Eb1) and Michael Le-vine, Department of Molecular and Cellular Biology, Uni-versity of California, Berkeley (Eb2). Df(2L)b88f32 andDf(2L)AdhnBR55 deficiencies both uncover Ance and wereobtained from John Roote. tud1 bw1 flies were obtainedfrom the Bloomington Stock Center. Male survivors fromtud1/tud1 females are male sterile with testes that lack germcells.

Immunohistochemistry and in situ hybridisation

Antibodies were raised against recombinant Ance ex-pressed in Pichia pastoris. Anti-Ance antiserum is specificfor Ance and does not cross-react with Acer (Houard et al.,1998.). Testes were dissected in PBS (10 mM Na phosphatebuffer, pH 7.4, 150 mM NaCl) and treated with 2% (v/v)hydrogen peroxide in methanol for 5 min, then washed threetimes in PBS prior to fixing for 20 min in 4% (w/v) para-formaldehyde in PBS. Tissues were blocked in 1% (w/v)bovine serum albumin (BSA), 10% (v/v) normal goat serumin PBST [PBS � 0.03% (v/v) Triton X-100] for 1 h at 25°Cbefore incubation in a 1:2000 dilution of primary antibodyin PBST overnight at 4°C. Testes were washed three timesin PBST before treatment with the Vectastain ABC Kit(Vector Laboratories Ltd., Peterborough, UK) according tothe manufacturer’s instructions. For immunofluorescence,testes were dissected, fixed in 4% (v/v) paraformaldehyde,blocked, treated with primary antibody, and washed inPBST as described above. Testes were then incubated in a1:2000 dilution of FITC-conjugated goat anti-rat IgG(Sigma-Aldrich Co. Poole, Dorset, UK) in PBST for 1 h at25°C. After three washes in PBST, testes were mounted inVectashield (Vector Laboratories) and viewed with a Bio-Rad Radiance 2000 confocal microscope.

For in situ hybridisations, testes were dissected in PBSand squashed beneath a siliconised coverslip on a poly-L-lysine-coated slide. Slides were frozen in liquid nitrogen,and the coverslip was flipped off. Slides were dehydrated in70% ethanol, washed in PBS, then fixed for 20 min in 5%

239D. Hurst et al. / Developmental Biology 254 (2003) 238–247

(v/v) formaldehyde in PBST. In situ hybridisation was car-ried out by using Ance RNA probes according to the methoddescribed in Siviter et al. (2000).

Measurement of ACE activity with the substrate hippuryl-L-histidyl-L-leucine (Hip-His-Leu)

ACE activity was assayed by measuring the amount ofhippuric acid released from Hip-His-Leu (Sigma-AldrichCo.). Oregon R and tud testes extracts (2 �l) were incubatedwith 5 mM Hip-His-Leu in 0.1 M Tris–HCl, pH 8.3, 0.3 MNaCl, 10 �M ZnSO4 for 4 h at 37°C in a total volume of 20�l. The enzyme activity was stopped by reducing the pH to2.0 with the addition of 8% (v/v) trifluoracetic acid. Thefinal volume was made up to 260 �l by the addition of 0.1%(v/v) trifluoracetic acid, and the resulting hippuric acid was

resolved by HPLC and detected at 214 nm, as describedpreviously (Lamango et al., 1996).

Immunoelectrophoresis

Proteins from Oregon R or tud testes (13 pairs) wereseparated on a 10% SDS–PAGE gel according to standardprocedures (Laemeli, 1970), transferred to a PVDF mem-brane, and then incubated with anti-Ance antibody at a1:5000 dilution in PBS containing 0.05% (v/v) Tween 20and 5% (w/v) non-fat dried milk powder (Houard et al.,1998). Bound anti-Ance antibody was detected by using ahorseradish peroxidase-conjugated sheep anti-rabbit Fc an-tibody and the Enhanced Chemi-Luminescence (ECL) De-tection Kit (Amersham Pharmacia Biotech Ltd., UK) asdescribed in the manufacturer’s instructions.

Fig. 1. Immunodetection of Ance protein in Drosophila testes. Whole testes were treated with an Ance-specific primary antiserum raised in rat. Boundantibodies were detected by using ABC kit (Vector Laboratories) (A, B, D–F) or a FITC-conjugated secondary antibody (C). (A) Whole testis. Staining isapparent in a narrow band close to the tip of the testis, in the waste bags, and is also associated with elongated cysts, strongest at the tail end. (B) Wholetestis treated with normal rat serum. Some faint staining of elongated cysts can be seen. (C) Optical section of sub-apical region, apical end uppermost. Strongstaining is present in vesicles of large primary spermatocytes. (D) Waste bag at higher magnification. (E) Apical band at higher magnification. (F) Vesicularstructure in large primary spermatocyte. wb, waste bag; ab, apical band; v, vesicles. Scale bars: (A, B) 100 �m; (C–E) 10 �m.

240 D. Hurst et al. / Developmental Biology 254 (2003) 238–247

Phalloidin staining

Individualisation complexes were stained by using rho-damine-conjugated phalloidin as described in Fabrizio et al.(1998).

Electron microscopy

Testes were dissected from newly eclosed Oregon R orAnce mutant males and fixed in 2.5% (v/v) gluteraldehydein 0.1 M phosphate buffer, pH 6.9, for 3 h. The specimenswere then washed twice in 0.1 M phosphate buffer, pH 6.9,before postfixation for 1 h in 1% (w/v) osmium tetroxide in0.1 M phosphate buffer, pH 6.9. After washing in twochanges of 0.1 M phosphate buffer, pH 6.9, the specimenswere dehydrated by using an ascending series of five etha-nol in water solutions (20–100%), each step taking 20 min.After one additional change of 100% ethanol, the specimenswere embedded in araldite (Luft, 1961). Sections (80–90nm) were cut from the araldite blocks by using an Ultrami-crotome (Reichert-Jung Ultracut-E, Leica, Milton Keynes,UK) and were stained with uranyl acetate and Reynolds’lead citrate solution (Reynolds, 1963). Sections were exam-ined by using a Jeol 1200EX transmission electron micro-scope (Jeol UK Ltd., Welwyn Garden City, UK) at an80-kV accelerating voltage.

Results

Expression of Ance in testes

Previous work by Tatei et al. (1995) suggested that Ancemay play some role in Drosophila testes. As a first step toelucidating this role, the distribution of Ance protein in wildtype testes was investigated by immunocytochemistry usingan Ance-specific antibody. A complex pattern of proteindistribution was revealed (Fig. 1A). Antibody staining wasobserved throughout the testes sheath in a punctate pattern.Towards the distal end, a diffuse band of staining on thesurface of the testis was observed (Fig. 1E). Although neverobserved in controls using nonimmune serum, this band ofstaining was not always observed with anti-Ance serum.Intense staining was observed associated with elongatedcysts, strongest at the tail end. Confocal microscopy did notreveal Ance inside elongated spermatid bundles (Fig. 1C),so this staining is associated with the surface of cysts,probably because Ance is adhering to the surface of the cystcells. Staining was also observed in the waste bags of cyststhat have undergone individualisation, as they coil towardsthe base of the testis (Fig. 1D). At higher magnification,Ance protein was observed in vesicular structures withinspermatocytes and early spermatids (Fig. 1C and F). These

Fig. 2. Distribution of Ance mRNA in Drosophila testes. In situ hybridisation was carried out on testes squashes by using a digoxygenin-labelled Anceantisense riboprobe (A) or a sense strand control (B). Highest levels of mRNA were detected in large primary spermatocytes (ps). Scale bars, 100 �m.Fig. 3. Immunodetection of Ance protein in testes lacking germ cells. (A, B) Immunocytochemistry. Testes from progeny of tud1/tud1 females were treatedwith anti- Ance antiserum (A) or normal rat serum (B), and bound antibodies were detected with an ABC Kit (Vector). t, testis; sv, seminal vesicle. Scalebar, 100 �m. (C) Immunoelectrophoresis. Each track contains protein extracts from 13 pairs of wild type (track 1) or tud (track 2) testes. The membrane wasprobed with Ance-specific anti-Ance antiserum and bound antibody detected by enhanced chemiluminescence.

241D. Hurst et al. / Developmental Biology 254 (2003) 238–247

vesicles tended to form a distinct vesicular body in primaryspermatocytes. In later stages, vesicles were more dispersed.

In situ hybridisation using an Ance probe showed thathighest levels of RNA are found in large primary spermato-cytes. Lower levels were observed in germ cells at laterstages of spermatogenesis, but no Ance mRNA could bedetected in cyst cells or in elongated cysts (Fig. 2A).

Ance in tudor testes

The results of immunocytochemistry and in situ hybri-disation suggested that the germ cells are the main sites ofAnce synthesis. To determine the relative contribution ofsomatic and germ line tissues to Ance expression, immuno-cytochemistry, immunoelectrophoresis, and ACE enzymeassays were carried out on agametic testes from malesderived from tud1/tud1 females (Boswell and Mahowald,1985). Immunocytochemistry using an Ance-specific anti-serum on tud testes did not yield significantly higher levelsof staining than with the control nonimmune serum (Fig. 3Aand B). The amount of Ance protein, detected by immuno-electrophoresis, was substantially reduced in tud testes (Fig.3C), and ACE enzyme activity, expressed on a per pair oftestes basis, was 28% of the wild type (Table 1).

Characterisation of Ance mutants

Two extant Ance mutant alleles were obtained. Thesealleles were derived from the same EMS mutagenesis andwere previously designated l(2)34Eb1 and l(2)34Eb2. Pre-vious reports had suggested that both these alleles are lethal(Tatei et al., 1995) but, under our culture conditions,Ance34Eb2 homozygotes were viable although males weresterile. Tatei et al. (1995) had previously confirmedl(2)34Eb2 as an allele of Ance by rescuing the lethality witha wild type copy of the gene. Ance34Eb1/Ance34Eb2 trans-heterozygotes had reduced viability, and males were alsosterile. The survival of Ance34Eb1 and Ance34Eb2 was ap-proximately 37 and 54%, respectively, when combined witha deficiency of the Ance region. These data suggest thatAnce34Eb1 is the stronger allele. For both alleles, Ance/Dfsurvivors were male sterile.

Ance mutant testes phenotype

Testes of Ance34Eb2 homozyogotes or Ance34Eb1/Ance34Eb2 individuals were extremely variable in appear-

ance. Most were small with few elongated spermatids, noindividualised sperm, and degeneration of variable extent.Testes from Ance/Df(2L)AdhnBR55 or Ance/Df(2L)b88f32

mutants, however, were much more consistent in appear-ance. These individuals had full-length testes containingelongated spermatid bundles, but there was no evidence ofspermatid individualisation (Fig. 4).

To investigate further the individualisation phenotype,mutant testes were stained with rhodamine–phalloidin andDAPI to reveal individualisation complexes and nuclei re-spectively. Wild type testes squashes contained around 12strongly staining individualisation complexes (Fig. 5). Ap-proximately half of these were associated with needle-shaped spermatid nuclei and therefore represent ICs whichhave formed but have not yet begun the individualisationprocess. In contrast, very few ICs were observed inAnce34Eb1/Df or Ance34Eb2/Df testes, and these were usuallyassociated with spermatid nuclei (Fig. 5). Mutant spermatidnuclei were often scattered along the cyst (Fig. 6), as isobserved with other individualisation mutants (Fabrizio et

Table 1ACE activity in the testes of Oregon R males and agametic testes ofmales derived from tudor females

ACE activity (units/animal)

OregonR testes 12.25 � 0.75tudor testes 3.46 � 0.66

Note. A unit of enzyme activity is 1 pmole of Hip-His-Leu hydrolysedper minute. Data are presented as the mean � s.e.m. of nine experiments.

Fig. 4. Phenotype of Oregon R wild type (A) and Ance34Eb1/Df(2L)AdhnBR55 mutant (B) testes. Testes were squashed beneath a cover-slip and viewed under phase contrast optics. Individualised, coiled motilesperm are visible in wild type (arrows). Ance mutant testes contain elon-gated spermatid cysts (ec) but no individualised sperm. Scale bar, 100 �m.

242 D. Hurst et al. / Developmental Biology 254 (2003) 238–247

al., 1998), and many nuclei had spherical instead of thenormal needle-shaped morphology (Fig. 6).

Ultrastructural analysis was carried out to determinewhether any other morphological abnormalities werepresent in mutant testes. Extended cysts never contained theordered array of spermatid tails observed in wild type cysts,and all elongated cysts contained about the same amount ofcytoplasm between sperm tails as is found in nonindividu-alised wild type cysts (Fig. 7A and B). Transverse sectionsof mutant testes revealed abnormal mitochondrial deriva-tives (Fig. 7D and E). The minor derivatives were largerthan those from mature, individualised wild type cysts,though usually smaller than major derivatives in the samecyst. The major derivative was variable in morphology,often distended and appearing dumb-bell shaped in crosssection (Fig. 7D and E). This was also observed less fre-quently with the minor derivative (Fig. 7E). Paracrystallinematrix was present in most major derivatives but was re-stricted to a small zone adjacent to the attachment with theaxoneme (Fig. 7D), a distribution normally found in imma-ture, elongating cysts in wild type testes (Fig. 7C; Fuller,

1993). These abnormalities were more frequent and pro-nounced in Ance34Eb1/Df than in Ance34Eb2/Df testes.

Discussion

The discovery that the testis and epididymis of sexuallymature rats are the richest source of ACE, and that the highlevels of activity were not attained in hypophysectomisedanimals, was the first piece of evidence for a possible rolefor ACE in mammalian reproduction (Cushman andCheung, 1971). We now know that the major source ofmammalian testicular ACE is the developing germ cells.RNA transcripts of mammalian germinal ACE (gACE) areonly found in haploid germ cells, with maximum expressionoccurring in mature spermatids of mice and rats at stages 10and 11 (cap phase) (Sibony et al., 1994). Proof that gACEhas a role in fertility was obtained by studying mice carry-ing an insertional mutation that inactivated the ACE gene.Mice lacking gACE were infertile, despite producing nor-mal-looking sperm (Krege et al., 1995; Hagaman et al.,

Fig. 5. Individualisation complexes (ICs) in Oregon R (A, B) and Ance34Eb1/Df(2L)AdhnBR55 (C, D) testes. Testes squashes were treated with rhodamine-conjugated phalloidin to detect F-actin complexes (A, C) and DAPI to detect nuclei (B, D). Several brightly fluorescing ICs are visible in (A). Approximately50% of these colocalise with DAPI staining nuclei (arrows in A and B). Only one IC is visible in the Ance34Eb1/Df(2L)AdhnBR55 spread which colocaliseswith nuclei (arrow in C and D). Scale bar, 50 �m.

243D. Hurst et al. / Developmental Biology 254 (2003) 238–247

1998). Failure to make angiotensin II is not the explanationfor the male infertility in these mice, since angiotensinogenknock-out mice are fertile (Kim et al., 1995). Another pos-sible function of gACE might be nonenzymic in nature.Hagaman et al. (1998) suggest the possibility that mousegACE might be involved in an interaction between spermand oviductal membranes, which would explain why sper-matozoa of gACE knockout males are not found in thefemale oviduct, beyond the intramural uterotubal junction,following mating.

The discovery of ACE in the testes of Haematobia irri-tans exigua (Wijffels et al., 1996), Lymantria dispar (Loebet al., 1998), Neobellieria bullata, Leptinotarsa decemlin-eata, and Locusta migratoria (Schoofs, et al., 1998; Isaac etal., 1999) and the localisation of the enzyme to germ cells inseveral of these species suggested that the function of gACEmight have been conserved during the course of evolution.

In situ hybridisation, immunolocalisation, and analysisof tud testes suggest that germ cells are the main sites ofAnce synthesis in Drosophila testes. The residual proteinand enzyme activity in tud testes may be due to Ance fromthe haemolymph adhering to the testis sheath. ACE activityin tud testes may also be due to the presence of Acer, whichcleaves Hip-His- Leu, albeit less efficiently than Ance(Houard et al., 1998). Ance protein accumulates in vesiclesin spermatocytes, where it may be involved in processingpeptides which are subsequently secreted by these cells.gACE in homogenates of human spermatazoa is very effi-cient at trimming basic dipeptides from the C terminus ofpeptide intermediates generated by prohormone convertases(Isaac et al., 1997) and may therefore have a general phys-iological role in the maturation of signaling peptides. Thispenchant for peptide intermediates with either Arg or Lys inthe last two amino acid positions is also shared with Ance

Fig. 7. Ultrastructure of Oregon R wild type and Ance34Eb1/Df(2L)AdhnBR55 testes. Micrographs represent transverse sections of testes at approximatelymid-length. (A) Wild type cyst prior to and (inset, at same magnification) after individualisation. The cyst is well ordered and, after individualisation, themajor mitochondrial derivative contains an electron- dense paracrystalline matrix. (B) Ance34Eb1/Df cyst. The cyst lacks overall organisation when comparedwith wild type and contains abnormal, lobed mitochondrial derivatives. (C) Immature, elongated wild type cyst. Paracrystalline matrix is present in the majormitochondrial derivative, adjacent to the axoneme. (D, E) Higher magnification views of mitochondrial derivatives in an Ance34Eb1/Df cyst. In (D), only themajor derivative is lobed and there is paracrystalline matrix adjacent to the axoneme. In (D), both derivatives are lobed, and there is very little paracrystallinematrix visible in the major derivative. a, axoneme; ma, major mitochondrial derivative; mi, minor derivative. Scale bars: (A, B) 500 nm; (C–E) 100 nm.

Fig. 6. Nuclei scattering and abnormal morphology in Ance34Eb1/Df(2L)AdhnBR55 spermatid cysts. (A) Normal needle-shaped nuclear morphology and noscattering. (B) Partial loss of nuclear morphology and five scattered nuclei. (C) Nuclei are all spherical and scattered in this cyst. (D, E) Cyst with abnormalnuclei located at the head end of the cyst (E) and aberrant, colocalising phalloidin staining (D). Scale bar, 10 �m.

244 D. Hurst et al. / Developmental Biology 254 (2003) 238–247

245D. Hurst et al. / Developmental Biology 254 (2003) 238–247

(Isaac et al., 1998). Evidence which supports the idea thatthe testes are a site of metabolism of bioactive peptidescomes from the fact that the gene for the �-amidatingenzyme, peptidylglycine monooxygenase (CG12130), isrepresented in the Berkeley Drosophila Genome Projectadult testes EST set. However, cDNAs for any of the knownDrosophila prohormone convertases are not present amongthe 23,087 sequenced testes clones. Establishing a peptideprocessing role for Ance in the secretory pathway musttherefore await further studies. The Ance-containing vesi-cles are a similar size and have a similar morphology to themultivesicular bodies identified by ultrastructural analysis(see Fuller, 1993). However, a function for these vesicleshas not previously been reported. Ance may also play a rolein the lumen of the testes. Immunoreactive material ispresent in the lumen and it is likely that it associates withthe surface of elongated cysts. We have observed a tendencyfor Ance to adhere to the surface of other cell types, such asimaginal discs, gut, and embryonic epidermis (Siviter et al.,2002). This adherent property of Ance might have func-tional significance for cell interactions and signaling. In thiscontext, it is worth noting the suggestion made by Hagamanet al. (1998) that mouse gACE might be involved in thebinding of spermatozoa to female tissue.

In late stages of spermatogenesis, Ance is found in thewaste bag, where it may be involved in degrading peptidesderived from spermatid cytoplasm. This Ance is likely to bederived from the germ cell vesicles, which may release theircontents into the waste cytoplasm at this stage.

Analysis of male sterile Ance mutants suggests a role forthe protein in spermatid differentiation. Spermatogenesisappears to proceed normally in the mutants to the elongatedspermatid stage, but spermatids subsequently fail to indi-vidualise. This is accompanied by nuclei scattering andspherical nuclear morphology. This abnormal nuclear mor-phology is similar to that seen in thousand points of light(tho) and mozzarella (moz) mutants (Fabrizio et al., 1998),but these mutants often display fragmented ICs in additionto scattered nuclei, and no obvious defects at earlier stagesin spermatogenesis have been reported. Very few individu-alisation complexes, either intact or disrupted, were ob-served in Ance mutant testes, suggesting that ICs are unableto form in mutant cysts or that they form, but quickly breakdown. In this respect, Ance testes have similarity with the“early” mutants dud and nanking (Fabrizio et al., 1998).Electron microscopy revealed gross morphological defectsin the mitochondrial derivatives of Ance mutant testes. dudand nanking also have defects at earlier stages of spermat-ogenesis (Fabrizio et al., 1998), and it is possible thatspermatid defects prevent proper assembly of ICs. Supportfor this suggestion comes from observation of occasionalabnormal phalloidin-staining structures associated with ab-errant spermatid nuclei in Ance testes. When interpretingthese results it must be borne in mind that the Ance alleleswe have studied are hypomorphs and the effects on sper-matogenesis of an Ance null may be far more severe. One

possible explanation of our results is that Ance is responsi-ble for processing a peptide which regulates spermatid dif-ferentiation. This peptide is presumably synthesised withinthe cyst, either in the germ cells or the cyst cells, as isolatedcysts will undergo meiosis, differentiate normally, and un-dergo spermatid individualisation when under in vitro cul-ture (Cross and Shellenbarger, 1979).

Acknowledgments

We thank Adrian Hick, University of Leeds, for provid-ing the electron micrographs, John Roote, University ofCambridge, and Michael Levine, University of California,for flies. This work was supported by the BBSRC (24/SO9564 and 89/SO9563).

References

Boswell, R.E., Mahowald, A.P., 1985. tudor, a gene required for assemblyof the germ plasm in Drosophila melanogaster. Cell 43, 97–104.

Chen, Y., Dicou, E., Djakiew, D., 1997. Characterization of nerve growthfactor precursor protein expression in rat round spermatids and thetrophic effects of nerve growth factor in the maintenance of Sertoli cellviability. Mol. Cell. Endocrinol. 127, 129–136.

Chesneau, V., Prat, A., Segretain, D., Hospital, V., Dupaix, A., Foulon, T.,Jegou, B., Cohen, P., 1996. NRD convertase: a putative processingendoprotease associated with the axoneme and the manchette in latespermatids. J. Cell Sci. 109, 2737–2745.

Cornell, M.J., Williams, T.A., Lamango, N.S., Coates, D., Corvol, P.,Soubrier, F., Hoheisel, J., Lehrach, H., Isaac, R.E., 1995. Cloning andexpression of an evolutionary conserved single-domain angiotensin-converting enzyme from Drosophila melanogaster. J. Biol. Chem. 270,13613–13619.

Cross, D.P., Shellenbarger, D.L., 1979. The dynamics of Drosophila mela-nogaster spermatogenesis in in vitro cultures. J. Embryol. Exp. Mor-phol. 53, 345–351.

Cushman, D.W., Cheung, H.S., 1971. Concentrations of angiotensin-con-verting enzyme in tissues of the rat. Biochim. Biophys. Acta 250,261–265.

Esther Jr., C.R., Howard, T.E., Marino, E.M., Goddard, J.M., Capecchi,M.R., Bernstein, K.E., 1996. Mice lacking angiotensin-converting en-zyme have low blood pressure, renal pathology and reduced malefertility. Lab. Invest. 74, 953–965.

Fabrizio, J.J., Hime, G., Lemmon, S.K., Bazinet, C., 1998. Genetic dissec-tion of sperm individualization in Drosophila melanogaster. Develop-ment 125, 1833–1843.

Fuller, M.T. 1993. Spermatogenesis in Drosophila, in: Bate, M., Martinez-Arias, A. (Eds.), The Development of Drosophila melanogaster, ColdSpring Harbor Laboratory Press, New York, pp. 71–147.

Fuller, M.T., 1998. Genetic control of cell proliferation and differentiationin Drosophila spermatogenesis. Semin. Cell Dev. Biol. 9, 433–444.

Ghaddar, G., Ruchon, A.F., Carpentier, M., Marcinkiewicz, M., Seidah,N.G., Crine, P., Desgroseillers, L., Boileau, G., 2000. Molecular clon-ing and biochemical characterization of a new mouse testis soluble-zinc-metallopeptidase of the neprilysin family. Biochem. J. 347, 419–429.

Hagaman, J.R., Moyer, J.S., Bachman, E.S., Sibony, M., Magyar, P.L.,Welch, J.E., Smithies, O., Krege, J.H., O’Brien, D.A., 1998. Angio-tensin-converting enzyme and male fertility. Proc. Natl. Acad. Sci.USA 95, 2552–2557.

246 D. Hurst et al. / Developmental Biology 254 (2003) 238–247

Hicks, J.L., Deng, W.M., Rogat, A.D., Miller, K.G., Bownes, M., 1999.Class VI unconventional myosin is required for spermatogenesis inDrosophila. Mol. Biol. Cell. 10, 4341–4353.

Houard, X., Williams, T.A., Michaud, A., Dani, P., Isaac, R.E., Shirras,A.D., Coates, D., Corvol, P., 1998. The Drosophila melanogaster-related angiotensin-I-converting enzymes Acer and Ance: distinct en-zymic characteristics and alternative expression during pupal develop-ment. Eur. J. Biochem. 257, 599–606.

Isaac, R.E., Ekbote, U., Coates, D., Shirras, A.D., 1999. Insect angiotensin-converting enzyme. A processing enzyme with broad substrate speci-ficity and a role in reproduction. Ann. N. Y. Acad. Sci. 897, 342–347.

Isaac, R.E., Williams, T.A., Sajid, M., Corvol, P., Coates, D., 1997.Cleavage of arginyl-arginine and lysyl-arginine from the C-terminus ofpro-hormone peptides by human germinal angiotensin I-convertingenzyme (ACE) and the C-domain of human somatic ACE. Biochem. J.328, 587–591.

Isaac, R.E., Schoofs, L., Williams, T.A., Veelaert, D., Sajid, M., Corvol, P.,Coates, D., 1998. A novel peptide-processing activity of insect pepti-dyl-dipeptidase A (angiotensin 1-converting enzyme): the hydrolysis oflysyl-arginine and arginyl-arginine from the C-terminus of an insectprohormone peptide. Biochem. J. 330, 61–65.

Kim, H.S., Krege, J.H., Kluckman, K.D., Hagaman, J.R., Hodgin, J.B.,Best, C.F., Jennette, J.C., Coffman, T.M., Maeda, N., Smithies, O.,1995. Genetic control of blood pressure and the angiotensinogen locus.Proc. Natl. Acad. Sci. USA 92, 2735–2739.

Krege, J.H., John, S.W.M., Langenbach, L.L., Hodgin, J.B., Hagaman,J.R., Bachman, E. S., Jennette, J.C., O’Brien, D.A., Smithies, O., 1995.Male–female differences in fertility and blood pressure in ACE-defi-cient mice. Nature 375, 146–148.

Laemeli, U.K., 1970. Cleavage of structural proteins during the assemblyof the head of bacteriophage T4. Nature 227, 680–685.

Lamango, N.S., Sajid, M., Isaac, R.E., 1996. The endopeptidase activityand the activation by CT of angiotensin-converting enzyme is evolu-tionarily conserved: purification and properties of an an angiotensin-converting enzyme from the housefly, Musca domestica. Biochem. J.314, 639–646.

Loeb, M.J., De Loof, A., Schoofs, L., Isaac, E., 1998. Angiotensin II andangiotensin-converting enzyme as candidate compounds modulatingthe effects of testis ecdysiotropin in testes of the gypsy moth, Lyman-tria dispar. Gen. Comp. Endocrinol. 112, 232–239.

Luft, J.H., 1961. Improvements in epoxy resin embedding methods. J. Bio-phys. Biochem. Cytol. 9, 409–414.

Mbikay, M., Tadros, H., Ishida, N., Lerner, C.P., De Lamirande, E., Chen,A., El-Alfy, M., Clermont, Y., et al., 1997. Impaired fertility in micedeficient for the testicular germ-cell protease PC4. Proc. Natl. Acad.Sci. USA 94, 6842–6846.

Monsees, T.K., Gornig, M., Schill, W.-B., Miska, W., 1998. Possibleinvolvement of proteases in the regulation of spermatogenesis. Andro-logia 30, 185–191.

Monsees, T.K., Miska, W., Blocher, S., Schill, W.-B., Winkler, A., Siems,W.-E., 1999. Elements of the kallikrein-kinin system are present in ratseminiferous epithelium. Immunopharmacology 45, 107–114.

Reynolds, E.S., 1963. The use of lead citrate at high pH as an opaque stainin electron microscopy. J. Cell Biol. 17, 208–212.

Schoofs, L., Veelaert, D., De Loof, A., Huybrechts, R., Isaac, E., 1998.Immunocytochemical distribution of angiotensin I-converting enzyme-like immunoreactivity in the brain and testis of insects. Brain Res. 785,215–227.

Sibony, M., Segretain, D., Gasc, J.M., 1994. Angiotensin-converting en-zyme in murine testis: step-specific expression of the germinal isoformduring spermiogenesis. Biol. Reprod. 50, 1015–1026.

Siviter, R.J., Coast, G.M., Winther, A.M., Nachman, R.J., Taylor, C.A.,Shirras, A.D., Coates, D., Isaac, R.E., Nassel, R.R., 2000. Expressionand functional characterisation of a Drosophila neuropeptide precursorwith homology to mammalian preprotachykinin A. J. Biol. Chem. 275,23273–23280.

Siviter, R.J., Taylor, C.A.M., Cottam, D.M., Denton, A., Dani, M.P.,Milner, M.J., Shirras, A.D., Isaac, R.E., 2002. Ance, a Drosophilaangiotensin-converting enzyme homologue, is expressed in imaginalcells during metamorphosis and is regulated by the steroid, 20-hy-droxyecdysone. Biochem. J. 367, 187–193.

Tatei, K., Haini, C., Ip., T., Levine, M., 1995. Race: a Drosophila homo-logue of the angiotensin converting enzyme. Mech. Dev. 51, 157–168.

Taylor, C.A.M., Coates, D., Shirras, A.D., 1996. The Acer gene of Dro-sophila codes for an angiotensin-converting enzyme homologue. Gene181, 191–197.

Wijffels, G., Fitzgerald, C., Gough, J., Riding, G., Elvin, C., Kemp, D.,Willadsen, P., 1996. Cloning and characterization of angiotensin-con-verting enzyme from the dipteran species, Haematobia irritans exigua,and its expression in the maturing male reproductive system. Eur.J. Biochem./FEBS 237, 414–423.

Woodruff, R.C., Ashburner, M., 1979. The genetics of a small autosomalregion of Drosophila melanogaster containing the structural gene foralcohol dehydrogenase. Genetics 92, 133–149.

247D. Hurst et al. / Developmental Biology 254 (2003) 238–247


Recommended