You are on page 1of 12

Cell Tissue Res (1985) 242:145-156 ten

a n d 1"tssue
Research
9 Springer-Verlag1985

Selective destruction and regeneration


of rat Leydig cells in vivo
A new method for the study of seminiferous tubular-interstitial tissue interaction

J.B. Kerr, K. Donachie*, and F.F.G. Rommerts**


MRC Reproductive Biology Unit, and *Department of Obstetrics and Gynaecology, Centre for Reproductive Biology, Edinburgh, Scotland;
**Department of Biochemistry, Erasmus University, Rotterdam, The Netherlands

Summary. The effect of a single i.p. administration of eth- tion returning to the normal range during weeks 6-9 (Bu-
ane dimethanesulphonate (EDS) upon rat testicular histolo- 'Lock and Jackson 1975). These changes are accompanied
gy was studied by light microscopy and morphometry up by a temporary decline in testis, seminal vesicle and prostate
to 4 weeks after treatment. One day after injection the inter- weights and impairment but subsequent restoration of sper-
stitial tissue exhibited degenerating Leydig cells, abundant matogenesis and fertility (Cooper and Jackson 1970; Jack-
pyknotic interstitial cells, deposition of cellular debris and son and Morris 1977; Jackson et al. 1973). Serum concen-
extensive networks of fibrillar material. Macrophages con- trations of follicle-stimulating hormone (FSH) and luteiniz-
tained greatly increased numbers of cytoplasmic inclusion ing hormone (LH) are initially elevated following EDS
bodies. From 3 to 7 days morphometric analysis showed treatment but return to within normal limits with the resto-
that Leydig cells and cellular debris had disappeared from ration of spermatogenic function (Jackson and Morris
the interstitial tissue, leaving only macrophages, fibroblasts 1977), suggesting re-establishment of the feedback signals
and lymphatic endothelial tissue. A very small number of linking the testis and the anterior pituitary. Although these
new Leydig cells were seen on day 14, often located in numerous studies point to a direct cytotoxic effect of EDS
peritubular or perivascular positions. Regeneration of foe- upon Leydig cell function, correlation with the morphologi-
tal-like Leydig cells occurred by 4 weeks, their cytoplasm cal response of the Leydig cells has not been available.
containing large lipid inclusions and, numerous Leydig cells In view of much recent interest in the dynamic interactions
were often observed closely applied to the walls of the semi- between spermatogenesis and the interstitial tissue, which
niferous tubules. The observations suggest that, after exper- suggests a paracrine relationship between the Sertoli cells
imental destruction and depletion of Leydig cells, an inter- and Leydig cells (Kerr and Sharpe 1985a; Sharpe 1984),
stitial precursor cell, as yet unidentified, gives rise to a new the possibility that EDS might serve as a valuable tool to
Leydig cell population. EDS thus offers a valuable opportu- study experimental alterations to this intercellular axis
nity to study further the interactions between the seminifer- prompted the present study.
ous tubules and the interstitial tissue following the destruc-
tion and subsequent regeneration of the Leydig cells.
Materials and methods
Key words: Antifertility c o m p o u n d - Ethane dimethanesul-
phonate - Leydig cells - Destruction - Regeneration - Rat Treatments and tissue fixation
Adult male Sprague-Dawley rats, approximately 350g
body weight were used from our own laboratory colony.
Twenty five rats (5 per group) received a single intraperiton-
Ethane-l,2-dimethanesulphonate (EDS) has long been re- eal injection of EDS (7.5 mg/100 g body weight) in di-
cognised to exert an unusual pharmacological action upon methyl-sulphoxide-water (1:3 V/V). A control group of 5
the rat testis resulting in a temporary period of sterility rats received a single i.p. injection of the vehicle. EDS-
2-8 weeks after a single treatment (Cooper and Jackson injected rats were killed on days 1, 3, 7, 14 and 28 after
1970; Jackson 1964; Jackson and Jackson 1984). In con- treatment, and controls were killed on day 28. The testes
trast to the antifertility actions of other simple diesters of were fixed by vascular perfusion using methods described
methane sulphonic acid, the anti-spermatogenic effect of by Kerr et al. (1984). Briefly, under deep ether anaesthesia
EDS is not attributable to the arrest of germ cell prolifera- the thoracic aorta was cannulated, the right atrium severed
tion, but rather it appears to selectively impair the function and the testes briefly perfused with physiological saline,
of the Leydig cells (Jackson and Jackson 1984). In the rat then with a mixture of 5% glutaraldehyde, 3% formalde-
a single dose of EDS inhibits Leydig cell testosterone pro- hyde and 0.01% trinitrophenol buffered in 0.1 M sodium
duction and suppresses serum androgen levels 24 h after cacodylate, pH 7.4, for 45 rain. The hardened testes were
administration (Bu'Lock and Jones 1976; Morris and removed, decapsulated and, after measurement of their vol-
McCluckie 1979), both these markers of Leydig cell func- umes by water displacement (Elias and Hyde 1980), cut
into cubes approximately 2 m m on edge and fixed for 3 h
Send offprint requests to. Dr. J.B. Kerr, Dept. of Anatomy, Mon- in the same fixative. Blocks were post-fixed for 2 h in os-
ash University, Clayton, Victoria 3168, Australia mium tetroxide, stained en bloc with uranyl acetate, dehy-
146

drated in ethanol and embedded in a 1 : 1 mixture of Epon- ten showed a prominent nucleolus and dense patches of
Araldite. Blocks from all animals were cut at 1 ~tm using heterochromatin were associated with the nuclear mem-
a Reichert OmU3 ultramicrotome and then stained with brane. Leydig cell cytoplasm was basophilic after Tolui-
Toluidine blue. Sections were examined and photographed dine-blue staining, and many dense inclusion bodies were
with a Zeiss photomicroscope II using Plan Apo 40/1.0 representative of mitochondria. Cytoplasmic lipid inclu-
and 63/1.4 oil-immersion objectives. sions were not commonly noted, a feature characteristic
of adult rat Leydig cells. Macrophages, present in fewer
Morphometric analysis numbers than Leydig cells, were readily identified by their
pale-staining irregular nuclei together with their character-
Blocks from all testes were cut at 1 lam and examined by
istically abundant supplies of cytoplasmic granules and vesi-
light microscopy to exclude from quantitative analysis any
cles indicative of their well developed lysosomal compo-
tissues which exhibited unsatisfactory preservation using
nents fulfilling the phagocytic function of macrophages.
criteria similar to those previously described (Kerr and
Thin fusiform endothelial cells forming the walls of lym-
Sharpe 1985a). Tissues displaying artifactual spaces be-
phatic spaces together with fibroblasts associated with the
tween the seminiferous tubules and the interstitial tissue
loose interstitial connective tissue were also observed. The
which may arise from poorly fixed localised sites within
pale-staining interstitial tissue matrix represented the inter-
the tissue or from mechanical damage during the cutting
stitial fluid of the extensive lymphatic sinusoids characteris-
of glutaraldehyde-fixed blocks were not assigned for mor-
tic of the testis in rats and other rodent species. Twenty-four
phometry. Testes of 3 of the 5 animals in each group were
hours after EDS treatment, most of the Leydig cells exhib-
thus selected in which the preservation of the tissue was
ited various degrees of disintegration, few appeared mor-
judged satisfactory for quantitative analysis. A minimum
phologically unaltered and deposition of fibrillar-like mate-
of 10 one-micron sections (all from different blocks) of
rial was also observed (Figs. 2, 3). By light microscopy the
transversely-sectioned seminiferous tubules were selected
structural damage to Leydig cells was often so marked that
from each rat which on average provided an area containing
it became difficult to identify individual cells since their
15 20 seminiferous tubules in each section. Sections were
cytoplasm was fragmented and dispersed throughout large
examined using 10 x eyepieces and a 25 x objective lens.
areas of the intersitital spaces (Fig. 3). Clumps of frag-
The volumetric densities of the total interstitial tissue (all
mented Leydig cells commonly exhibited dense granular
cells, extracellular spaces and blood vessels), the Leydig
material and numerous cytoplasmic vesicles. Individual
cells and macrophages (identified as described below) were
pyknotic structures were often scattered amongst the exten-
measured by a point-counting technique using a graticule
sive cellular debris. Macrophages were always present and
of 121 points (11 x 11 squares) inserted into one eyepiece.
contained increased numbers of dense granules and cyto-
Volumetric densities of these components were determined
plasmic vesicles. Extensive networks of randomly oriented
on 6-10 random areas of each section and the scores were
fibre-like material was noted (Fig. 4), a feature not seen
expressed as a percentage of the total number of points
in the normal interstitial tissue. Since the endothelial lining
superimposed upon each section. Average volumetric densi-
of lymphatic spaces was not seen to partly surround the
ties were calculated for each testis and the means +s.d.
degenerating Leydig cells, the fibrillar material was possibly
for the 3 animals belonging to each group were determined.
derived from disorganisation and dispersal of these endo-
The volume of the testis occupied by each component was
thelial cells. Elongated or irregularly-shaped nuclei of endo-
obtained by multiplying the volumetric density of the com-
thelial and connective tissue cells were also observed, but
ponent by the volume of the whole testis from which the
their morphology did not appear different from control
sections were derived. Mean absolute volumes of the com-
tissues. The seminiferous epithelium appeared qualitatively
ponents were calculated for each treatment group. The pos-
normal. On day 3 after EDS treatment, Leydig cells had
sible alteration of volumetric density and absolute volume
disappeared from the testis (Fig. 5) although spermatogene-
of the measured components due to changes in the volume
sis appeared unaffected. Despite extensive analysis of the
of tissue blocks during their processing and embedding were
interstitial tissue, none of the interstitial cells could be classi-
not taken into account since the principal interest of the
fied as adult, immature or atrophied Leydig cells. The cellu-
study was to compare the morphology of the normal testis
lar debris and fibrillar-like material present on day 1 after
to testes from EDS-treated animals.
treatment was not seen on day 3. The interstitial spaces
were filled with interstitial fluid, within which were numer-
Statistics ous macrophages although in contrast to their appearance
Morphometric data are expressed as the mean _+S.D. Sta- on day 1, their cytoplasm did not usually contain especially
tistical significance of the results was determined by Dun- large granules and vesicles. Interstitial connective tissues
can's new multiple range test. and elongated cytoplasmic profiles of endothelial cells were
a common feature of the interstitial tissue. Occasional lym-
phocytes and plasma cells were noted within the interstitial
Results lymphatic sinusoids. Seven days after EDS treatment, the
morphology of the seminiferous epithelium was similar to
Histology day 3 although degenerating germ cells were occasionally
In vehicle-treated control rats, the testicular interstitial tis- present in the basal aspects of seminiferous tubules at stage
sue contained prominent clusters of Leydig cells often sur- VII and VIII of the spermatogenic cycle. Leydig cells were
rounding blood vessels or occupying positions more cen- not observed within the interstitial tissue (Fig. 6). Occasion-
trally placed within the interstitial tissue (Fig. 1). Leydig ally, focal areas of the interstitial tissue were filled with
cells exhibited a wide variety of shapes including ovoid, great numbers of mononucleated cells (Fig. 7) commonly
polygonal or irregularly elongated profiles. Their nuclei of- surrounding the interstitial capillaries and venules. In fa-
147

Fig. 1. Vehicle-treated control testis illustrating the typical morphology of the interstitial tissue containing numerous Leydig cells. Macro-
phages (M) and a monocyte (asterisk) are shown within the fluid-filled interstitial lymphatic sinusoid (LS). Nuclei of lymphatic endothelial
cells are shown (arrowheads). x 470

Fig. 2. 1 day after EDS treatment, showing early disruption of interstitial tissue organisation. Some Leydig cells appear normal while
others (arrowheads) show cytoplasmic damage. Fragments of pyknotic material are seen and macrophages (arrows) often exhibit dense
cytoplasmic inclusions, x 470
148

Fig. 3. 1 day after EDS treatment, illustrating more advanced cellular disintegration resulting in many scattered pyknotic fragments
(arrows). Some Leydig cells (L) appear intact but contain unusual cytoplasmic vesicles. Macrophages (arrowheads') are numerous and
contain many cytoplasmic granules and vesicles. • 470

Fig. 4. 1 day after EDS treatment, in which Leydig cells are absent but cellular debris (arrowhead) and pyknotic material possibly
represent degraded remnants of Leydig cells. Many mononucleated cells are present, including macrophages (asterisk). Fibrillar networks
(arrows) occupy the lymphatic sinusoidal spaces (LS). • 470
149

Fig. 5. 3 days after EDS treatment, illustrating disappearance of Leydig cells and cellular debris. The lymphatic sinusoid (LS) contains
cells with cytoplasmic filopodia, at times identified as macrophages (arrows). Note perivascular and peritubular nuclei (arrowheads)
of unknown identity. The nucleus of a lymphatic endothelial cell is shown (asterisk). x 470

Fig. 6. 7 days after EDS treatment, illustrating the lymphatic sinusoid (LS) containing macrophages and a monocyte (arrows), the
latter having a characteristic nucleus and granule-free cytoplasm. Nuclei of endothelial cells are indicated (arrowheads). x 470
150

Fig. 7. 7 days alter EDS treatment, illustrating focal infiltration of monocytes and tymphocytes into the interstitial tissue. A cell within
the venule is closely applied to the vessel endothelium (arrow). x 470 Inset." Possible migration of this cell through the endothelial
tissues, x 850

Fig. 8. 14 days after EDS treatment, showing Leydig cells (asterisks) with oval or spherical nuclei. Other interstitial cells (arrowheads)
possibly represent stages in the development of new Leydig cells. Macrophages and mononucleated cells are illustrated (arrows). x 470
151

Fig. 9. 28 days after EDS treatment, illustrating abundant foetal-like Leydig cells with round nuclei and cytoplasmic lipid inclusions
(L). Macrophages are also shown (arrows). x 470

vourable sections, these cells were also observed within the morphology of Leydig cells in the foetal testis. These Leydig
vessel lumina and at times appeared to be migrating cells contained less irregularly-shaped nuclei which were
through the endothelium of the vessels. These observations usually spherical or slightly ovoid in shape. In addition,
together with their morphology suggested their classifica- many of the Leydig cells exhibited large cytoplasmic lipid
tion as a mixture of monocytes and lymphocytes. Two inclusions, a feature rarely seen in normal adult rat Leydig
weeks following treatment, the majority of the interstitial cells. The location and morphology of macrophages, con-
tissue spaces presented a similar morphology and cell popu- nective tissue cells, endothelial cells and lymphatic sinusoids
lation to that previously seen on days 3 and 7, but occasion- of this treatment group was similar to their appearance
ally, small numbers of Leydig cells were observed (Fig. 8). within control testes. Although some seminiferous tubules
However, the structure of these Leydig cells was not identi- were severely depleted of germ cells most exhibited more
cal to Leydig cells seen in the adult testis. Their nuclei advanced spermatogenesis compared to the 2 week treat-
were larger and showed a spherical or oval shape, often ment group.
containing a single large nucleolus. Although the cytoplas-
mic area was well developed, granules representing mito- Morphometric analysis
chondria were not especially abundant and small lipid inclu- Testis volume was significantly (P<0.01) reduced 2 and
sions were often present. Interstitial cells with elongated 4 weeks after EDS treatment (Table l). The volumetric den-
or polygonal nuclei were occasionally seen close to the peri- sities of the measured interstitial tissue components showed
tubular tissue, and their position and morphology was simi- significant increases and decreases in EDS-treated rats com-
lar to the interstitial mesenchymal cells seen in foetal or pared to vehicle-injected control rats. A better appreciation
immature rat testes. The seminiferous tubules exhibited of these changes was demonstrated by the conversion of
patchy disruption of spermatogenesis. The most remarkable volumetric density data into absolute volumes per testis
feature of the interstitial tissue 28 days after EDS adminis- in the various treatment groups. The total volume of inter-
tration was the striking abundance of large Leydig cells stitial tissue (all interstitial cells, extracellular spaces and
(Fig. 9). Leydig cells often surrounded blood vessels or blood vessels) was significantly ( P < 0.05) increased I day
formed large clusters of cells, and of particular interest was post-EDS treatment compared to vehicle-treated testes, but
their frequent association with the peritubular tissues of at 7 and 14 day post-EDS treatment, it was significantly
the seminiferous tubules, thereby forming uninterrupted decreased (P<0.05 to 0.001). The significant increase in
rows of cells partly surrounding the tubules (Fig. 10). By total interstitial tissue volume 1 day after EDS reflected
comparison with adult Leydig cells from the vehicle-injected a major increase in the total interstitial fluid volume (mea-
control group, the Leydig cells of the 28 day group exhib- sured as extracellular space). The total volume of Leydig
ited a very different morphology and were similar to the cells within the testes of all EDS-treated groups was signifi-
152

Fig. 10. 28 days after EDS treatment, showing a seminiferous tubule partly surrounded by peritubular foetal-likc Leydig cells. The
lymphatic sinusoidal space (LS) is indicated, x 470
153

Table 1. Morphometric analysis of interstitial tissue (Means + S.D.; n =3 animals)

Vehicle Days after EDS treatment


control
1 3 7 14 28

Testis volume (cm3) 1.24 + 0.04 1.37+0.11 1.12_+0.12 1.04___0.05 0.89_+0.04* 0.75+0.07*
O

Volumetric density (%)


Total interstitial tissue l t.4 _ 0.7 13.7 _+0.5 9.6 +1.0 6.8 _+0.3* 11.5 _+2.0 19.6 _+2.0*
Leydig cells 3.0 +0.2 0.8 +0.3** n.d.** n.d.** 0.t +0.0"* 2.6 +0.7
Macrophages 0.3 +0.1 0.5 ___0.1 0.8 +0.1"* 0.4 • 0.5 _+0.1 0.4 _+0.t
Volume per testis (~1)
Total interstitial tissue 141 +6 188 +12" 109 _+20 71 _+1"* 99 _+23* 145 +28
Leydig cells 37 +2 11 _+5'* n.d.** n.d.** t +0"* 19 +4**
Macrophages 4 +l 7 +1" 9 _+2* 3 _+1 4 +_1 3 -+-0

n.d., not detected


*P < 0.05 or 0.0t, **P < 0.001, EDS treatments vs. vehicle control

cantly smaller ( P < 0.001) than the total volume within con- within the interstitial tissue; ii) the question of the origin
trol testes, but showed a marked restoration towards nor- of new "foetal-like" Leydig cells and the factors stimulating
mal values between weeks 1 and 4 following EDS adminis- their differentiation; and iii) the response of the seminifer-
tration. Total volume of macrophages was significantly ous epithelium to a period of androgen deprivation due
( P < 0.05, 0.01) increased 1 and 3 days after EDS treatment, to temporary depletion of the Leydig cell population.
but was unchanged at other times compared to the normal In earlier studies, it has been suggested that EDS exerts
testis. a deleterious effect upon rat Leydig cells via an interaction
with the plasma membrane receptors for hCG, since a brief
( 4 1 2 h) prior exposure of the testis to EDS essentially abol-
Discussion
ishes the capacity of the Leydig cells to produce cyclic AMP
In a series of studies over many years (Jackson 1964; Jack- in response to in vitro hCG stimulation (Bu'Lock and Jones
son and Jackson 1984) the antispermatogenic and antiferti- 1976). Whether this action involves alkylation of cellular
lity action of EDS in rats was thought to be mediated via components similar to the alkylating actions on the rat
a direct cytotoxic inhibition of the function of the Leydig seminiferous epithelium observed with other diesters of
cells. However, the subsequent recovery of Leydig cell func- methane sulphonic acid is not yet resolved (Morris and
tion accompanying the restoration of spermatogenic activi- McCluckie 1979). The rapid disintegration of Leydig cells
ty and fertility raised the possibility that a proportion of after EDS treatment is consistent with the acute impairment
the Leydig cell population was resistant to EDS action or of steroidogenic metabolism and inhibition of cyclic AMP
alternatively normal androgenic function was restored via and testosterone production (Bu'Lock and Jackson 1971,
proliferation of Leydig cells from other interstitial cells. 1975). Although the precise mode of action of EDS upon
However these views have not been resolved, since the ef- Leydig cells requires additional study, the present observa-
fects of EDS upon the morphology of the Leydig cells and tions indicate that destruction of Leydig cells elicits an im-
other elements of the interstitial tissue have not been stud- mune response involving macrophage-lymphocyte interac-
ied. In the present study the use of semi-thin plastic sections tion. At the time of Leydig cell disintegration, the total
of perfusion-fixed testes together with the application of volume of macrophages within the testis had increased sig-
contemporary morphometric analysis has provided a signif- nificantly compared to the control testis tissue and their
icant step forward in elucidating the morphological re- greatly increased quantities of cytoplasmic granules and va-
sponse of the interstitial tissue following EDS treatment. cuoles suggested a highly activated state of phagocytosis
The results show that within 24 h of EDS treatment, Leydig of Leydig cell debris. During the induction of immunity,
cells have undergone advanced disintegration which results macrophages are believed to facilitate the engagement of
in their complete elimination from the testis on day 3. Ley- antigen-sensitized lymphocytes via secretion of lymphokin-
dig cells are absent from the testis for at least 5 days (days etic factors (Drutz 1976; Roitt 1980). Upon presentation
3-7 after treatment) but remarkably, during weeks 2 4, re- of antigen to the lymphocytes via macrophages, additional
generation of the Leydig cells occurs resulting in a substan- lymphocytes are attracted to the site of antigen-responding
tial restoration of their total volume normally seen in the cells and this response was reflected by the infiltration of
adult rat testis. We believe these findings to be unique for many lymphocytes into focal areas of the interstitial tissue
two reasons. First, the selective destruction of Leydig cells seen 7 days after EDS treatment. Activated lymphocytes
in the mammalian testis has heretofore not been described are known to produce lymphokines which selectively attract
and secondly, their rapid re-appearance following complete monocytes and their derivatives, the macrophages, to the
elimination from the testis has no previous precedent. sites of antigenic challenge (Rocklin 1976), and may also
A number of fundamental aspects of the cell biology stimulate proliferation of fixed macrophages such as those
of the testis are raised by the present study: i) the mecha- within the rat testis (Miller et al. 1983). These mechanisms
nism by which EDS selectively destroys the Leydig cells probably account for the significant increase in total macro-
and the involvement of macrophages and lymphocytes phage volume during and soon after the destruction of the
154

Leydig cells on days 1-3. Augmentation of increased mac- develop from the peritubular tissues surrounding the semi-
rophage phagocytic activity also occurs via lymphocyte- niferous tubules, an observation also noted in the interstitial
macrophage interaction mediated by other lymphokine sub- tissue 4 weeks after EDS treatment. Serum FSH levels are
stances. This phenomenon raises the question of the prima- significantly elevated above the normal range within 3 days
ry site of action of EDS. EDS perhaps specifically disrupts after EDS treatment, attaining maximal elevation (3 fold)
the Leydig cell and in some way presents an antigenic chal- at 4 weeks and thereafter decline towards normal values
lenge to the nearby macrophages or alternatively, EDS may (Jackson and Morris 1977). Since the target tissue for FSH
be an allergen specific to the macrophages which precipi- is the Sertoli cell (Means et al. 1976), the present observa-
tates their phagocytic activities and interaction with lym- tions together with those mentioned above introduces the
phocytes. Although either or both of these suggested modes possibility that in addition to the trophic action of LH,
of action may conceivably operate, recent in vitro studies the seminiferous tubules also in some way exert a local
(Rommerts, unpublished observations) have shown that the stimulatory effect upon the development and function of
addition of EDS to isolated adult rat Leydig cells severely the interstitial tissue, a concept which is attracting increas-
impairs steroid production within 3 h and induces morpho- ing support (Aoki and Fawcett 1978; deKretser 1982;
logical alterations by 24 h. It therefore seems likely that deKretser and Kerr 1983; Parvinen 1982; Parvinen et al.
the former mechanism of EDS action upon Leydig cells 1984; Sharpe 1983, 1984). Although not studied in detail
may occur in vivo. here, preliminary observations indicate that occasional de-
The regeneration of abundant numbers of new Leydig generating germ cells were noted 1 week after EDS treat-
cells 4 weeks after EDS treatment and within 3 weeks fol- ment although maximum impairment of spermatogenesis
lowing their total depletion raises the question of their ori- occurred approximately 2 weeks after EDS treatment. A
gin from within the interstitial tissue. Since Leydig cells qualitative improvement of the seminiferous epithelium in
were absent from the testis during days 3-7, the new genera- most seminiferous tubules was observed in association with
tion of Leydig cells cannot have arisen by mitosis or hyper- new Leydig cell growth at 4 weeks. A detailed analysis of
trophy of pre-existing Leydig cells although these mecha- impairment and restoration of spermatogenesis is underway
nisms are thought to explain Leydig cell hyperplasia in the in our laboratory. The seminiferous tubules are thought
normal testis following chronic stimulation with hCG to be the site of production of factors modifying both the
(Chemes et al. 1976; Christensen and Peacock 1980). The size (Bergh 1983) and secretory properties of the Leydig
present observations favour an alternative mechanism in cells (Parvinen et al. 1984; Sharpe and Cooper 1984; Sharpe
which a more primitive interstitial stem-cell population dif- and Bartlett 1985). Taken with the histological evidence
ferentiates into histologically recognisable Leydig cells. As that during pubertal maturation, new Leydig cells may dif-
the newly developed Leydig cells exhibited a very similar ferentiate from peritubular interstitial cells (Christensen
morphology to foetal rat Leydig cells (Lording and 1975; deKretser 1967; Fawcett 1973; van Straaten and
deKretser 1972; Tapanainen et al. 1984), we suggest that Wensing 1978), the development of numerous peritubular
their differentiation followed the pattern of development Leydig cells 4 weeks post-EDS treatment suggests a func-
of Leydig cells in the foetal testis in which precursor mesen- tional relationship between spermatogenesis and Leydig cell
chymal cells differentiate into histologically recognisable growth. Interstitial cells suggested to be Leydig cell precur-
Leydig cells (Wartenberg 1981). The interstitial tissue in sors have been noted in association with the peritubular
2- and 4-week EDS-treated rats contained many fusiform tissue of the canine testis (Connell and Christensen 1975),
mesenchymal cells in addition to numerous interstitial cells and fully differentiated Leydig ceils are commonly noted
with a morphology intermediate between the former cell within the peritubular tissue of the human testis in cases
type and the larger Leydig cells. Occcasionally, mitotic fig- of cryptorchidism or Sertoli cell only syndrome (Mori et al.
ures were seen amongst the interstitial cells. These observa- 1978; Schulze and Holstein 1978). These observations sug-
tions suggest that the regeneration of Leydig cells occurs gest that newly differentiated Leydig cells may arise from
initially via mesenchymal cell maturation and possibly to peritubular ceils and subsequent leave the lamina propria
a lesser extent, by Leydig cell division, in agreement with to gain access to the surrounding interstitial tissue.
the pattern of Leydig cell development observed during foe- At present it is not known why extensive degeneration
tal and postnatal life (Christensen 1975; Christensen and of spermatogenesis does not occur even in the chronic ab-
Gillim 1969; Gondos et al. 1976, 1977; Prince 1984). Addi- sence of the Leydig cells, a condition in which intratesticular
tional ultrastructural and autoradiographic studies using levels of testosterone would presumably be undetectable.
tritiated thymidine will be necessary to determine the loca- Since quantitative spermatogenesis is dependent upon high
tion and identity of Leydig cell precursors and their pattern intratesticular testosterone concentrations (Stevens and
of maturation into functional Leydig cells. It is widely be- Steinberger 1983) failure to observe any significant germ
lieved that LH (or CG) is the hormone which during post- cell degeneration before 2 weeks post-EDS treatment sug-
natal or foetal life exerts a major stimulatory influence upon gests that the seminiferous tubules conserve their endoge-
the development of Leydig cells (Faiman et al. 1981; nous supply of testosterone possibly via the intratubular
Swerdloff and Heber 1981). Since serum LH levels are ele- presence of androgen-binding protein (Hansson et al. 1976)
vated up to five-fold above the normal range for 4 weeks and a recently reported high capacity androgen-binding fac-
after EDS treatment (Jackson and Morris 1977), LH is per- tor (Parvinen 1984). Alternatively, disruption of Sertoli cell
haps responsible for the regeneration of Leydig cells. How- - Leydig cell interaction by EDS may activate local control
ever, we have recently shown (Kerr and Sharpe 1985a, b) mechanisms within the seminiferous tubule in an attempt
that, in the immature rat testis, FSH provokes rapid hyper- to preserve intratubular testosterone at levels sufficient to
plasia and hypertrophy of Leydig cells and an accompany- support qualitative spermatogenesis. We are investigating
ing reduction in the numbers of mesenchymal cells. These the acute and chronic effects of EDS upon the distribution
studies showed that many of the Leydig cells appeared to of intratesticular testosterone levels to define more clearly
155

the d y n a m i c relationships b e t w e e n the two c o m p a r t m e n t s ation of Leydig cells in the rabbit testis. Am J Anat
o f the testis w h i c h are o f f u n d a m e n t a l i m p o r t a n c e in the 145:167-182
r e g u l a t i o n o f fertility. Hansson V, Weddington SC, French FS, McLean W, Smith A,
Nayfeh SN, Ritzen EM, Hagenas L (1976) Secretion and role
of androgen-binding proteins in the testis and epididymis. J
Acknowledgements. We thank Professor Sir Alistair Currie (Dept. Reprod Fert Suppl 24:17-33
of Pathology, University of Edinburgh) for access to ultramicro- Jackson H (1964) The effects of alkylating agents on fertility. Br
tome equipment. This work was performed while J.B.K. was sup- Med Bull 20:10%114
ported by a C.J. Martin Research Fellowship from the National Jackson H, Jackson CM, Jones P (1973) Hormonal antagonism
Health and Medical Research Council of Australia. to the antispermatogenic effect of ethylene dimethanesutphon-
ate in rats. J Reprod Fert 34:133-135
Jackson CM, Jackson H (1984) Comparative protective actions
of gonadotrophins and testosterone against the antispermato-
References genic action of ethane dimethanesulphonate. J Reprod Fert
71 : 393-401
Aoki A, Fawcett DW (1978) Is there a local feedback from the Jackson CM, Morris ID (1977) Gonadotrophin levels in male rats
seminiferous tubules affecting activity of the Leydig cells? Biol following impairment of Leydig cell function by ethylene di-
Reprod 19:144-158 methanesulphonate. Andrologia 9: 29-35
Bergh A (1983) Paracrine regulation of Leydig cells by the semini- Kerr JB, Sharpe RM (1985 a) Stimulatory effect of follicle-stimulat-
ferous tubules. Int J Androl 6:57-65 ing hormone on rat Leydig ceils. A morphometric and ultra-
Bu'Lock DE, Jackson CM (1971) Suppression of testicular andro- structural study. Cell Tissue Res 239:405-415
gen synthesis in the rat by ethylene dimethane-sulphonate. Gyn- Kerr JB, Sharpe RM (1985b) Follicle-stimulating hormone induc-
ecol Invest 2:305-308 tion of Leydig cell maturation. Endocrinology (in press, June)
Bu'Lock DE, Jackson CM (1975) Suppression of testicular andro- Kerr JB, Mayberry RA, Irby DC (1984) Morphometric studies
gen synthesis in the rat by ethane 1,2, dimethanesulphonate. on lipid inclusions in Sertoli cells during the spermatogenic
J Steroid Biochem 6:1181-1185 cycle in the rat. Cell Tissue Res 236:699-709
Bu'Lock DE, Jones P (1976) Effect of ethane 1,2-dimethanesul- Lording DW, deKretser DM (1972) Comparative ultrastructural
phonate on the cyclic AMP response in rat testis to stimulation and histochemical studies of the interstitial cells of the rat testis
by HCG. J Reprod Fert 47:151-153 during fetal and postnatal development. J Reprod Fert
Chemes HE, Rivarola MA, Bergada C (1976) Effect of H C G on 29 : 261-269
the interstitial cells and androgen production in the immature Means AR, Fakunding JL, Huckins C, Tindall D J, Vitale R (1976)
rat testis. J Reprod Fert 46:279-282 Follicle-stimulating hormone, the Sertoli cell and spermatogen-
Christensen AK (1975) Leydig cells. In: Hamilton DW, Greep esis. Recent Prog Horm Res 32:477-527
RO (eds) Handbook of physiology, sect 7, vol 5. Williams and Miller SC, Bowman BM, Roland H G (1983) Structure, cytochem-
Wilkins, Baltimore, pp 57-94 istry, endocytic activity, and immunoglobulin (Fc) receptors
Christensen AK, Gillim W (1969) The correlation of the fine struc- of rat testicular interstitial-tissue macrophages. Am J Anat
ture and function in steroid-secreting cells, with emphasis on 168:1 13
those of the gonads. In : McKern KW (ed) The gonads. Apple- Mori H, Shiraishi T, Matsumoto K (1978) Ectopic Leydig ceils
ton Century Crofts, New York, pp 415-488 in seminiferous tubules of an infertile human male with a chro-
Christensen AK, Peacock KC (1980) Increase in Leydig cell mosomal aberration. Andrologia 10:434-443
number in testes of adult rats treated chronically with an excess Morris ID, McCluckie JA (1979) Temporal changes in serum an-
of human chorionic gonadotrophin. Biol Reprod 22:383-391 drogen after temporary impairment of Leydig cell function by
Connell C J, Christensen AK (1975) The ultrastructure of the canine ethane-l,2-dimethane sulphonate. J Steroid Biochem
testicular interstitial tissue. Biol Reprod 12:368-382 10 : 467-469
Cooper ERA, Jackson H (1970) Comparative effects of methylene, Parvinen M (1982) Regulation of the seminiferous epithelium. En-
ethylene and propylene dimethanesulphonates on the male rat docr Rev 3:404-417
reproductive system. J Reprod Fert 23:103-108 Parvinen M (1984) Interactions between germinal cells and Sertoli
deKretser DM (1967) The fine structure of the testicular interstitial cells. In : Saez J, Forest MG (eds) Molecular and cellular endo-
cells in men of normal androgen status. Z Zellforsch crinology of the testis. I N S E R M Symposia, (in press)
80: 594-609 Parvinen M, Nikula H, Huhtaniemi I (1984) Influence of rat semi-
deKretser DM (1982) Sertoli cell - Leydig cell interaction in the niferous tubules on Leydig cell testosterone production in vitro.
regulation of testicular function. Int J Androl Suppl 5 : 11 17 Mol Cell Endocrinol 37:331-336
deKretser DM, Kerr JB (1983) The effect of testicular damage Prince FP (1984) Ultrastructure of immature Leydig cells in the
on Sertoli and Leydig cell function. In: deKretser DM, Burger human prepubertal testis. Anat Rec 209:165-176
HG, Hudson B (eds) The pituitary and the testis. Clinical and Rocklin RE (1976) Mediators of cellular immunity. In: Fudenberg
experimental studies. Springer, Berlin, pp 133-154 HH, Stites DP, Caldwell JL, Wells JV (eds) Basic and clinical
Drutz DJ (1976) Immunity and infection. In: Fudenberg HH, Stites immunology. Lange, Los Altos, pp 10~113
DP, Caldwell JL, Wells JV (eds) Basic and clinical immunology. Roitt IM (1980) Essential immunology. Blackwell, Oxford
Lange, Los Altos, pp 182-194 Schulze C, Holstein A F (1978) Leydig cells within the lamina pro-
Elias H, Hyde DM (1980) An elementary introduction to stereo- pria of seminiferous tubules in four patients with azoospermia.
logy (quantitative microscopy). Am J Anat 159:412-446 Andrologia 10: 44~452
Faiman C, Winter JSD, Reyes FI (1981) Endocrinology of the Sharpe RM (1983) Local control of testicular function. Quart J
fetal testis. In: Burger H, deKretser D (eds) The testis. Raven, Exp Physiol 68 : 265-287
New York, pp 81-105 Sharpe RM (1984) Intratesticular factors controlling testicular
Fawcett DW (1973) Observations on the organisation of the inter- function. Biol Reprod 30:29-49
stitial tissue of the testis and on the occluding cell junctions Sharpe RM, Bartlett JMS (1985) Intratesticular distribution of tes-
in the seminiferous epithelium. In: Raspe G (ed) Advances in tosterone in rats and the relationship to the levels of a peptide
the biosciences 10. Pergamon Press, Oxford, pp 83-99 that stimulates testosterone secretion. J Reprod Fert (in press)
Gondos B, Morrison KP, Renston R H (1977) Leydig cell differen- Sharpe RM, Cooper I (1984) Intratesticular secretion of a factor(s)
tiation in the prepubertal rat testis. Biol Reprod 17:745-748 with major stimulatory effects on Leydig cell testosterone secre-
Gondos B, Renston RH, Goldstein DA (1976) Postnatal differenti- tion in vitro. Molec Cell Endocrinol 37:159-163
156

Stevens RW, Steinberger E (1983) Effect of testosterone on quanti- testicular endogenous steroids and number of Leydig cells be-
tative maintenance of spermatogenesis in hypophysectomized tween the fetal period and sexual maturity. Biol Reprod
rats. Endocrinology 112 : Suppl 175, abst 377 31 : 1027-1035
Straaten HWM van, Wensing CJG (1978) Leydig cell development Wartenberg H (1981) Differentiation and development of the
in the testis of the pig. Biol Reprod 18:86-93 testes. In: Burger H, deKretser D (eds) The testis. Raven Press,
Swerdloff RS, Heber D (1981) Endocrine control of testicular func- New York, pp 39-80
tion from birth to puberty. In: Burger H, deKretser D (eds).
The testis. Raven, New York, pp 107 126
Tapanainen J, Kuopio T, Pelliniemi LJ, Huhtaniemi l (1984) Rat Accepted April 22, 1985

You might also like