You are on page 1of 12

BIOLOGY OF REPRODUCTION 61, 1468–1479 (1999)

Luteal Regression in the Primate: Different Forms of Cell Death During Natural
and Gonadotropin-Releasing Hormone Antagonist or Prostaglandin Analogue-
Induced Luteolysis

H.M. Fraser,1,2 S.F. Lunn,2 D.J. Harrison,3 and J.B. Kerr4


MRC Reproductive Biology Unit,2 Edinburgh, EH3 9ET, United Kingdom
Department of Pathology,3 University of Edinburgh, Edinburgh, EH3 9ET, United Kingdom
Department of Anatomy,4 Monash University, Clayton, Melbourne, Victoria 3168, Australia

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


ABSTRACT are replaced with bundles of collagen fibers, scattered fi-
broblasts, and occasional macrophages [5–9]. Most corpora
Morphological changes in the corpus luteum following nat-
ural and induced luteolysis in the marmoset were investigated
albicantia are resorbed and replaced by ovarian stroma [10].
by light and electron microscopy. Functional corpora lutea were Morphological regression of the corpus luteum necessarily
studied in the mid and late luteal phase, naturally regressed cor- involves cell death, but the mechanisms by which each of
pora lutea in the early and late follicular phase, and corpora the cell types within luteal tissue is destroyed remain un-
lutea induced to regress by administration of GnRH antagonist clear.
or prostaglandin F2a analogue in the midluteal phase. Natural Developing follicles within the ovary are continually un-
luteolysis was associated with lutein cell atrophy, condensation dergoing remodeling, with cell proliferation and death; fol-
of cytoplasmic inclusions and organelles, and accumulation of licular atresia involving degeneration of granulosa cells is
lipid. GnRH antagonist treatment resulted in aggregations of an example of apoptosis [11, 12], a process in which indi-
smooth membranes and myelin-like bodies in the cytoplasm of vidual cells die within healthy tissue. Since the discovery
the lutein cells together with complex aggregations of degen- that apoptosis is regulated by a group of survival and death
erative cells. After prostaglandin treatment, the lutein cells con- factors associated with specific genes, there has been ex-
tained numerous small and large vesicles; as the degenerative tensive investigation into the control of cell death in the
changes advanced, these vesicles coalesced into alveolar-type ovary, and many of these factors have been shown to be
vacuoles, and nuclei involuted. These results show that in the present in the corpus luteum [12–15]. Recently, several re-
marmoset, natural luteolysis and the two luteolytic treatments ports have described significant tissue apoptosis as indicat-
reveal different forms of luteal degeneration and cell death,
ed by DNA electrophoresis or the in situ terminal deoxynu-
none of which fit the ultrastructural criteria for apoptosis. More
emphasis needs to be placed on understanding these predomi-
cleotidyl transferase-mediated dUTP nick end-labeling re-
nant nonapoptotic forms of cell death in order to elucidate the action for demonstrating DNA fragmentation in sheep [16],
process of luteolysis in the primate. cow [17, 18], rabbit [19], rat [20], and hamster [21] corpora
lutea. These results confirm earlier studies showing mor-
INTRODUCTION phological evidence for apoptosis during luteal regression
in some species [22–24].
After ovulation in primates, the resultant corpus luteum Because of the important differences in the mechanisms
remains functional for only about 2 weeks unless rescued of luteal regression between species [25], it is essential for
by chorionic gonadotropin (CG). This process is essential our understanding of the physiology of the human corpus
for the establishment and maintenance of early pregnancy luteum that suitable primate models be investigated. Mor-
since, in the absence of the luteotrophic signal, the corpus phological features of cell death in the regressing human
luteum undergoes spontaneous loss of cell function and [5–7, 26] and primate corpus luteum [27] have been de-
structural regression. The mechanisms involved in luteol- scribed. Although the evidence for apoptosis is less con-
ysis in primates are poorly understood. The hormonal prod- vincing, it has been tempting to assume that these reports
ucts of the corpus luteum are under the control of pituitary describe apoptotic cells, at least in part. Recently, using 39
LH in Old World primates [1, 2], New World primates [3], end-labeling, apoptosis in the corpus luteum has been re-
and women [4], but luteal regression is not caused directly ported in the human [28, 29] and marmoset [30].
by a decline in LH secretion [2]. This suggests that a spe- However, our studies on natural and induced luteolysis
cific luteal phase length is an inherent property of this tis- in the marmoset demonstrated another form of cell death
sue. associated with pronounced cellular vacuolation [14, 30].
The demise of the corpus luteum, resulting in its trans- Ultrastructural descriptions of natural luteolysis have not
formation into the irregular connective tissue that consti- been reported for primate tissue in association with recent
tutes the corpus albicans, involves degeneration of all luteal advances in the knowledge of different forms of cell death,
cells leading to their disappearance. Involution of the tissue and we question whether any of the previous reports have
is accompanied by progressive fibrosis and shrinkage. Thus, demonstrated apoptosis convincingly. In addition, little is
the architecture of the mature corpus luteum is dramatically known about the ultrastructural features of the primate cor-
altered so that the rich vascular supply, the supporting con- pus luteum after induced luteolysis [31]. In the current
nective tissue cells, and the granulosa and theca lutein cells study, histological and ultrastructural changes in luteal cell
death in the marmoset after natural and induced luteolysis
Accepted July 15, 1999. have been explored and compared. The marmoset has the
Received February 2, 1999.
1
Correspondence: H.M. Fraser, MRC Reproductive Biology Unit, Cen- advantage that induction of luteolysis can be reliably
tre for Reproductive Biology, 37 Chalmers Street, Edinburgh EH3 9ET, UK. achieved subsequent to GnRH antagonist treatment (by re-
FAX: 44 131 228 5571; e-mail: h.fraser@ed-rbu.mrc.ac.uk moving LH support), or by a direct inhibitory effect of
1468
LUTEAL CELL DEATH IN THE PRIMATE 1469

prostaglandin treatment on the hormone-producing lutein plasma progesterone concentrations were 186 and 124 ng/
cells. It was postulated that differences in response to the ml; they were 20 and 12 ng/ml in the late luteal phase
treatments would reveal divergent pathways of luteal cell marmosets. Values obtained during the early and midfolli-
death. Finally, by compressing the time scale of luteal re- cular phase marmosets were , 4 ng/ml. Both GnRH and
gression with such treatments, it was anticipated that the prostaglandin treatment resulted in a dramatic fall in pro-
model might show changes in the corpus luteum more read- gesterone concentrations, such that even after 12 h, pro-
ily than by examination of the more protracted process of gesterone was , 4 ng/ml, i.e., at follicular phase levels.
natural luteolysis.
Light Microscopic Observations
MATERIALS AND METHODS
Treatments and Collection of Tissue Normal corpus luteum. In contrast to observations in
most other primate species studied, distinct enfoldings of
Adult female common marmosets (Callithrix jacchus) lutein cells derived from the theca layer and separate from

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


were housed as described previously [14]. Blood samples regions of granulosa-derived lutein cells are not present in
were collected 3 times per week by femoral venepuncture the marmoset corpus luteum [34]. While a small number
without anesthesia to confirm normal ovulatory cycles. of hormone-producing cells had morphological features and
Plasma was stored at 2208C until required for assay. Cri- locations consistent with theca lutein cells, i.e., were dis-
teria for the occurrence of ovulation (Day 0) and normal tributed in occasional aggregations peripheral to the exten-
luteal phase length (18–22 days) were based on determi- sive areas of large granulosa lutein cells, for the purposes
nation of plasma progesterone concentrations as described of this report differences in the origin of the lutein cells
previously [32]. The experiments were carried out in ac- were not taken into consideration. The most prominent fea-
cordance with the Animals (Scientific Procedures) Act, tures in the normal corpora lutea collected during the mid-
1986. Ovaries were collected from animals during the mid- luteal phase were the large polyhedral lutein cells, charac-
luteal (Day 10), late luteal (Days 20–22, but prior to func- terized by circular nuclei in cross section with a nucleolus
tional luteolysis), early follicular (1–3 days postfunctional and large cytoplasmic volume (Fig. 1A). Dense bodies
luteolysis), and late follicular (5–7 days postfunctional lu- within the cytoplasm included mitochondria and lysosomes
teolysis) phases of the cycle (n 5 2 animals per group). To together with less basophilic inclusions typical of lipid
examine the effect of induced luteal regression, on Day 8/ droplets. In some tissue sections the lutein cells showed
9 after the estimated time of ovulation, the animals were varying degrees of basophilia, although it was not possible
treated with either 1 mg prostaglandin F2a analogue [33] to determine whether cytoplasmic density was correlated
(Planate, Coopers Animal Health Ltd., Crewe, Cheshire, with distinct differences in organelle or inclusion content.
UK) i.m. or the GnRH antagonist [14] Antarelix ([N-Ac-D- The lutein cells were supported by connective tissue dis-
Nal1,D-pCl-Phe2,D-Pal3,D-(Hic)6,-Lys(iPr)8,D-Ala10] GnRH, playing fibroblasts, and there was an extensive blood supply
500 mg/kg s.c; Europeptides, Argenteuil, France). Ovaries characterized by the occurrence of endothelial cell nuclei
were obtained 12 h or 24 h after treatment (n 5 3 animals and numerous lumina often containing erythrocytes.
per group). GnRH antagonist treatment. After GnRH antagonist
treatment, most of the hormone-producing (lutein) cells
Fixation were dramatically altered (Fig. 1B). Within each corpus lu-
The animals were sedated using 100 ml ketamine hydro- teum, varying degrees of structural change indicative of
chloride (Parke-Davis Veterinary, Pontypool, Gwent, UK) degeneration were observed, so that for descriptive purpos-
i.m. and killed with an i.v. injection of 400 ml Euthetal es, data for the 12- and 24-h posttreatment groups were
(sodium pentobarbitone; Rhone Merieux, Dublin, Ireland). combined. The lutein cells were characterized by the pres-
Ovaries were removed immediately and the corpora lutea ence of numerous small and large aggregations of granular
of the cycle identified macroscopically. After bisection, half material in the cytoplasm; basophilia in some cells was as-
of each corpus luteum (up to 3 per animal) was cut into 1- sociated with a range of intracellular inclusions, from het-
to 2-mm cubes using a razor blade and was immersion fixed erogeneous aggregations of granular materials to very
for 2 h in 3% glutaraldehyde in 0.1 M cacodylate buffer, dense, condensed material suggestive of degeneration.
pH 7.3. Specimens were then rinsed overnight at 48C in the When present in the plane of section, some lutein cell nu-
same buffer, postfixed in buffered 2% osmium tetroxide for clei appeared normal whereas others showed evidence of
2 h, and embedded in Araldite (Ladd Research, Burlington, extraction or dissolution of nuclear content, i.e., karyolysis.
VT) after dehydration in ethanol and propylene oxalate. In the more advanced state, the cells exhibited condensed
Semithin (1 mm) sections were stained with toluidine granular materials with only occasional nuclei present. In
blue for light microscope analysis. At least three blocks capillaries of the supporting tissue, erythrocytes were ob-
from each corpus luteum were studied in detail. Thin sec- served, and the connective tissue contained pale-staining
tions were stained with uranyl acetate and lead citrate and elliptical or fusiform nuclei characteristic of fibroblasts and
examined in a Philips EM 301 (The Netherlands) electron endothelial cells. Areas of advanced luteolysis comprised
microscope. The remainder of the ovary was fixed in 4% complex aggregations of degenerative cells and cellular de-
paraformaldehyde for 24 h and used in studies to determine bris, and it was not possible to distinguish accurately ele-
changes in other factors associated with control of luteal ments of the vascular system within the supporting tissue.
cell function described elsewhere. Prostaglandin analogue treatment. After prostaglandin
analogue treatment, luteal tissue from both 12- and 24-h
treatment groups displayed a heterogeneous range of de-
RESULTS generative change, so again, results for the two time points
Plasma progesterone concentrations after ovulation in were combined. Typically, some lutein cells exhibited de-
the marmoset are some 30 times higher than in women and generative alterations similar to those seen in the GnRH
macaques [32]. In two control midluteal phase animals, antagonist-treated group. However, most of the lutein cells
Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020
FRASER ET AL.
1470
LUTEAL CELL DEATH IN THE PRIMATE 1471

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


FIG. 2. Naturally regressing corpora lutea. a) Early phase of luteal regression showing condensed, intensely stained structures, probably lutein cells
(curved arrow), and smaller bodies (arrowhead) of uncertain identity. 3530. b) Another region of the same corpus luteum showing pyknotic-type bodies
(curved arrows) and fragmented structures (arrowheads). 3530. Both areas also exhibited lutein cells without apparent degenerative change. c) Corpus
luteum in advanced regression. The lutein cells were shrunken, often condensed, and surrounded by much connective tissue. 3530. d) Apoptotic
granulosa cells of an atretic follicle showing nuclear condensation, rounding-up of the degenerating cells, and fragmentation into apoptotic bodies.
3630.

showed combinations of dense granules and minute clear no significant changes in histology compared to the mid-
vacuoles in the cytoplasm (Fig. 1C). The nuclei, where luteal control tissue (not shown). In the two animals from
present, were at times considerably smaller than in com- which tissue was collected during the early follicular phase,
parable cells in control tissues. Some lutein cells showed and whose corpora lutea had ceased to function, numerous
intact nuclei within heterogeneous clumps of granular ma- condensed, intensely stained structures that had probably
terials in the cytoplasm. In the more advanced state, nu- been lutein cells were observed together with smaller bod-
merous lutein cells showed larger vacuoles that often gave ies and fragmented structures of unknown identity (Fig. 2,
the impression of coalescence into extensive clear areas of a and b). Often these dense bodies were associated with
cytoplasm with a minor proportion of granular material at spaces or vacuoles that appeared to be empty (Fig. 2b), but
times surrounding a small central nucleus or condensed ma- whether these were intracellular or extracellular could not
terial. Nuclei of the supporting tissue were seen, but often be determined with light microscopy. At this stage, the cor-
no clear distinction could be determined between the cap- pora lutea also exhibited lutein cells without apparent de-
illary endothelial cells and the fibroblasts of the connective generative change.
tissue. The corpora lutea from ovaries of the late follicular
Naturally regressed corpus luteum. Corpora lutea from phase were markedly decreased in volume compared to
the late luteal phase, prior to functional luteolysis, showed those within the midluteal control tissues, and they were
surrounded by an abundance of connective tissue. Numer-
b ous lipid inclusions, partly extracted, were present in most
of these cells. Scattered at random throughout the luteal
FIG. 1. A) Normal midluteal phase corpus luteum showing lutein cells tissue were pyknotic elements, at times showing morpho-
with central nucleus, and abundant cytoplasm containing mitochondria
and lipid. Capillaries containing erythrocytes are evident. B) Luteal tissue logical features suggesting cell degeneration and/or cell
after GnRH antagonist treatment illustrating a complex mixture of rec- death (Fig. 2c). Because of their small cross-sectional area,
ognizable lutein cells with small and large cytoplasmic inclusions (curved it was not possible to determine the nature of the degen-
arrows) and fragments of cells, vacuoles, and debris within the connective erating cells. Small arterioles and venules were structurally
tissue. When present, most lutein cell nuclei appeared morphologically normal, but the close aggregation of atrophic lutein cells
normal (arrowheads). C) Luteal tissue after prostaglandin treatment show-
ing two types of structural alterations, with most lutein cells with or with-
and the supporting cells and matrix of the connective tissue
out a nucleus displaying many cytoplasmic vesicles while others con- made it difficult to determine the morphological status of
tained mostly dense inclusions. Scattered cellular debris and necrotic-type the capillaries.
structures were noted in the connective tissue. 3920. Since apoptosis had been expected in luteal tissue, an
1472 FRASER ET AL.

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020

FIG. 3. A) A typical normal lutein cell with central nucleus (N), abundant mitochondria (M), and smooth endoplasmic reticulum (SER). 33500. B)
Lutein cell 24 h after GnRH antagonist treatment showing multiple myelin bodies (Mn) of various densities, forming membranous whorls at times in
association with areas of compact membranes. 33300. C) Lutein cells after prostaglandin treatment showing many vesicles of variable size. Nuclei (N)
were intact. 32800. D) Higher magnification of part of C showing small and large vesicles often closely associated or coalesced (arrows). The nucleus
(N) showed peripheral aggregations of heterochromatin. 38050.
LUTEAL CELL DEATH IN THE PRIMATE 1473

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


FIG. 4. A) Lutein cell, after prostaglandin administration, showing clumping of nuclear chromatin (C). The nucleolus (N), mitochondria (M) with
particulate morphology, and vesicles (V) are indicated. 37900. B) Lutein cell after prostaglandin, showing vesicle coalescence into giant intracellular
spaces. The nucleus (N) was involuted, and misshapen and mitochondria (M) were still observed. 33100.

established source of such cells, the granulosa layer of an or fragments of lutein cells, or of connective tissue origin,
atretic follicle, was examined to demonstrate nuclear con- was not determined.
densation (Fig. 2d). Degeneration of granulosa cells con- Prostaglandin analogue treatment. After prostaglandin
sistent with apoptosis was shown by the rounded cell con- analogue treatment, the smooth ER of lutein cells had a
tours, single or multiple dense aggregations of nuclear chro- vesicular appearance, and large dilated membrane-bound
matin or condensed cytoplasmic inclusions/organelles, and vesicles occupied much of the cytoplasmic volume (Fig.
numerous dense cell fragments 2–4 mm in diameter, com- 3C). When apposed, the larger vesicles were often conflu-
monly referred to as ‘‘apoptotic bodies.’’ ent, and in many lutein cells the coalescence of vesicles
gave rise to extensive interconnected structures resembling
Ultrastructural Observations an alveolar-type morphology (Fig. 3D). In contrast, the nu-
clei remained intact, but often the heterochromatin was con-
Normal corpus luteum. In control and mid and late lu- densed into many small irregular clumps subjacent to the
teal phase corpora lutea, the lutein cells were characterized nuclear membrane (Fig. 3, C and D), a feature not observed
by a central circular nucleus in cross section and a single in lutein cells from the normal corpus luteum. As the lutein
nucleolus. In the cytoplasm, the dominant components were cells became shrunken, the nuclear heterochromatin in
vesicles or short anastomosing tubules of smooth endo- some cells aggregated into several clumps (Fig. 4A), the
plasmic reticulum (ER), variable numbers of small lipid cytoplasm showing many vesicles together with abnormal
droplets, and many mitochondria (Fig. 3A). The morphol- mitochondria in which the cristae showed disruption into
ogy of the mitochondria was variable between the hor- particulate matter. In the more degenerate cells, multiple
mone-producing cells, some cells having lamellar-type cris- vesicles occupied much of the cell volume, while in others,
tae and others displaying tubular cristae that filled the mi- fewer vesicles were noted but their large size again made
tochondrial matrix. them the dominant feature (Fig. 4B). In these cells, the
GnRH antagonist treatment. The striking accumulation nuclei were involuted and often misshapen.
of a variety of dense bodies within lutein cells, as noted Within the connective tissue, the morphology of the cells
with light microscopy, was confirmed by ultrastructural and the extracellular matrix was suggestive of tissue dis-
analysis. Where present, the nuclei and mitochondria of lu- ruption with ruptured cell membranes and transformation
tein cells appeared normal, but the cytoplasm contained nu- of cellular organelles into fragments, particulate matter, and
merous large structures of variable form and density sug- residual bodies.
gestive of phases of degeneration and condensation of Naturally regressed corpus luteum. In corpora lutea of
membranes during formation of myelin-type inclusions ovaries collected from the early follicular phase of the cy-
(Fig. 3B). Similar myelin bodies, disrupted cytoplasm, and cle, the tissue contained morphologically normal lutein
cellular debris occurred external to the identifiable lutein cells in addition to degenerating lutein cells identified by
cells; but whether these structures were slender extensions their increased electron density, irregular contours, and
1474 FRASER ET AL.

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020

FIG. 5. A) Early natural regression of the corpus luteum showing shrinkage of lutein cells into condensed, irregular fragments (fr) and small, dense
circular bodies (arrows) scattered within the extracellular space or within macrophages (Ma). Intact lutein cells (N), a neutrophil (Ne), and fibroblasts
(F) are indicated. 34000. B) Early regressing corpus luteum showing fragmentation of lutein cells into smooth membrane structures (S), mitochondria-
rich components (M), and condensed cellular material (arrow). Note densely stained clustered mitochondria in adjacent lutein cells. 34000.
LUTEAL CELL DEATH IN THE PRIMATE 1475

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


FIG. 6. A) Cytoplasm of a naturally regressing lutein cell with several condensed, particulate bodies (arrows). 37800. B) A highly shrunken lutein cell
fragment engulfed by a macrophage in the regressing corpus luteum showing a single large dense body, and mitochondria (M), lipid (L), and lysosomes
(Ls). 35800. C) Higher magnification showing the particulate nature of dense bodies and a membrane surrounding the degenerating cell (arrow), with
the macrophage membrane (m) surrounding the ingested cell fragment. 313 700.

fragmentation (Fig. 5A). Small, dense circular elements 7D). Within the tissue examined, not every lutein cell was
representing clumps of cellular debris were numerous, and either fragmented or engulfed by phagocytes. Some were
most of these had been engulfed by macrophages located atrophied, often aggregated together, and their markedly
in the connective tissue between normal or degenerating shrunken cytoplasm contained secondary lysosomes, large
lutein cells. The presence of neutrophils suggested an in- lipid inclusions, and crystalline bodies (Fig. 7E). The nuclei
flammatory-type response associated with the destruction of these cells retained a morphologically normal appear-
and removal of dead and dying cells. The ultrastructural ance.
response of lutein cells was markedly variable; it included
clustering and increased density of the mitochondria, or
changes in the smooth ER to form innumerable small ves- DISCUSSION
icles, or giant whorls of tubular smooth ER (Fig. 5B). Con-
densation of cytoplasm was a universal feature either within To our knowledge, this is the first report of ultrastructural
focal regions of lutein cells, or as distinct, often extracel- changes in the corpus luteum following GnRH antagonist-
lular clumps of cellular debris. induced luteolysis. In addition, while extensive information
At higher magnification of recognizable lutein cells, or is available on the luteolytic effects of prostaglandin F2a
fragments derived from them, numerous particulate bodies analogue in the nonprimate, this is the first description of
were seen in the cytoplasm (Fig. 6A). With further con- such effects on luteal morphology in the primate corpus
densation, portions of degenerating lutein cells were phago- luteum.
cytosed by macrophages and became fully or partly sur- Recently, there has been an emphasis on apoptosis as
rounded by the macrophage plasma membrane (Fig. 6, B representing the principal form of luteal cell death, primar-
and C), indicating enclosure by a vacuole or phagosome. ily stemming from the localization of fragmented DNA
By the late follicular phase, the corpora lutea were in an [16–21]; and, in the case of nonprimates, there is also un-
advanced stage of regression characterized by general cell equivocal evidence for apoptosis based upon ultrastructural
atrophy, fragmentation, and increased proportion of extra- studies. However, it is emphasized that our observations do
cellular matrix and collagen. In some of the atrophied lutein not indicate rapid or widespread cell degeneration via ap-
cells, lipid inclusions had accumulated together with ex- optosis, as defined by accepted histological or ultrastruc-
tracted, oblong cytoplasmic structures resembling choles- tural criteria [35–37]. With the exception of a very small
terol-rich crystalline inclusions known to occur in other ste- number of degenerating lutein cells, including some en-
roidogenic cells, particularly in association with a decline gulfed by macrophages (e.g., Figs. 4A and 7D), classic ap-
in metabolic or synthetic activity (Fig. 7A). Fragments of optotic-type nuclei of lutein cell identity were rare. Our
condensed lutein cell cytoplasm were occasionally noted findings indicate that, during luteolysis in the marmoset,
within the lumina of capillaries (Fig. 7B) and often within death of the vast majority of lutein cells proceeds via non-
macrophages (Fig. 7C). In the latter, whole condensed nu- apoptotic mechanisms. During natural luteolysis, this in-
clei had been ingested by the macrophages, and the asso- volves cell atrophy with phagocytosis of cytoplasmic de-
ciated presence of lipid inclusions and crystalline-type bod- bris. This contrasts with both GnRH antagonist and pros-
ies suggested their identity as degenerating lutein cells (Fig. taglandin-induced luteolysis, which are associated with au-
Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020
FRASER ET AL.
1476
LUTEAL CELL DEATH IN THE PRIMATE 1477

tophagocytosis and nonlysosomal cell disintegration, smooth ER becoming markedly swollen and forming many
respectively. vesicles such that in the advanced state, these vesicles co-
Ultrastructural changes during natural luteolysis in the alesced into large vacuoles. This observation may be sim-
marmoset are consistent with earlier reports on luteal re- ilar to the heavily vacuolated cells described in paraffin
gression in the human [5–7, 26, 38] in that the atrophied sections in the naturally regressing human corpus luteum
lutein cells show persistence of an intact nucleus, with a [6], and implies the production of a fluid that is not released
cytoplasm that contains autophagocytic vacuoles and both from the cell—an event indicating disrupted cell metabo-
lipid and crystalloid inclusions. Cellular debris is present lism, synthesis, or secretion.
in the involuting lutein cells and in adjacent macrophages. The primary action of GnRH antagonist treatment is to
Furthermore, heterochromatin does not condense or mar- suppress LH secretion [3], direct effects on the corpus lu-
ginate into aggregates or crescentic caps; the nucleus does teum being unlikely in the marmoset [39]. The fall in pro-
not convolute into separate protuberances or ‘‘blebs’’; and gesterone secretion after GnRH antagonist administration is
many of the cytoplasmic organelles do not remain intact dependent upon the prior decline in LH [3]. In contrast, the
but exhibit spontaneous autolysis and are degraded by ly- luteolytic action of prostaglandin F2a in the marmoset is

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


sosomes to form autophagosomes and structurally complex directly upon the corpus luteum; in vivo treatment with
debris. As regression proceeds, with phagocytic ingestion prostaglandin analogue is followed by a more rapid sup-
of cell debris, the remaining lutein cells are considerably pression of plasma progesterone secretion without change
atrophied and accumulate lipid and crystalline-type inclu- in LH levels [3]; and in vitro, LH-stimulated progesterone
sions indicating cessation of steroidogenesis. It is likely that production is inhibited at pre- and post-cAMP sites [3, 40].
these shrunken lutein cells are destroyed by macrophages This would agree with in vivo studies in which prostaglan-
as the previously highly cellular corpus luteum is converted din analogue treatment at or after the midluteal phase led
into a corpus albicans. Taken together, these results suggest to irreversible suppression of progesterone secretion in the
that natural regression of lutein cells in the human and mar- marmoset [33]. While the suppressive effects of GnRH an-
moset is a form of nonapoptotic degeneration. tagonist treatment can be overcome by concomitant admin-
The absence of detectable degenerative changes in the istration of hCG [1, 3], the marmoset corpus luteum cannot
late luteal phase, prior to functional luteolysis in the normal be ‘‘rescued’’ when prostaglandin analogue and hCG are
cycle of the marmoset, was somewhat surprising, since given together [3]. Since both treatments result in failure
shrinkage of lutein cells at this period has been described of LH stimulation of steroidogenesis, the ultrastructural dif-
in the human corpus luteum [6]. The luteal phase of the ferences indicate an additional effect of prostaglandin on
marmoset is slightly longer and of more variable length the lutein cell as a result of its direct action. Further studies
than in Old World primates and women, and perhaps this may help provide an explanation at the cellular level as to
is associated with an extended phase of maintenance of why these treatments are associated with differing out-
structural integrity. This may be followed by a more rapid comes in vivo.
degenerative phase when functional luteolysis finally oc- It is surprising that neither GnRH antagonist nor pros-
curs. taglandin treatment resulted in changes identical to those
Both GnRH antagonist and prostaglandin analogue treat- occurring during natural luteolysis. In the marmoset, the
ments were associated with dramatic changes in the mor- mechanism of natural luteolysis is not known; and if there
phology of the lutein cells within 12 h. Degenerative chang- is an endogenous luteolysin, there is no direct evidence that
es in the smooth ER were more advanced than any ob- it acts rapidly. However, it is not unreasonable to expect
served in the mitochondria, suggesting that the early failure that both treatments may mimic and accelerate naturally
to secrete progesterone was the result of an acute sensitivity occurring phenomena. For example, it is known that the
of the smooth ER. Interestingly, the changes to smooth ER responsiveness of the LH receptor declines toward the end
in the lutein cells were markedly different according to the of the luteal phase [41], so removal of interference with the
luteolytic treatment. In the GnRH antagonist-treated ani- normal LH stimulus by luteolytic treatments may be ex-
mals, the membranes of the smooth ER, and possibly seg- pected to induce a rapid precipitation of events associated
ments of the redundant plasma membrane, became con- with loss of LH receptor activation. Although the demise
densed into islands of concentric membranes that resulted of the luteal cells during natural luteolysis does not follow
in the formation of myelin bodies. This response suggests the same morphological pattern as for induced luteolysis in
cell degeneration via autophagocytic mechanisms followed the marmoset, GnRH antagonist and prostaglandin treat-
by heterophagocytosis. While this process occurs on a small ments may still provide a valuable approach for further
scale during normal cellular reorganization of lutein cells studies of the mechanisms responsible for the functional
[38], the appearance of the cells from the GnRH antagonist- and morphological demise of the primate corpus luteum.
treated animals indicated a process that would lead to cell In contrast to our findings in the marmoset, prostaglan-
death. After prostaglandin administration, a similar re- din F2a analogue treatment in nonprimate species, e.g., the
sponse was observed in a minority of lutein cells; but the rat [42], guinea pig [23], and sheep [24, 43, 44], results in
remainder showed a completely different appearance, with changes apparently similar to those observed in natural re-
gression. This species difference may originate from the
b fact that natural luteolysis in sheep and cattle is brought
FIG. 7. Advanced naturally regressing corpus luteum. A) Lutein cells about primarily by prostaglandin F2a of uterine origin and
were shrunken and contained lipid, lysosomes, and (extracted) cholester- that this process is advanced by the exogenous administra-
ol-rich crystalline inclusions (arrows). 32900. B) Fragments of a degen- tion of the analogue. Lutein cells of the ovine corpus lu-
erated lutein cell (arrows) within a capillary; note endothelial cell nucleus teum were reported to be highly vacuolated with contracted
(EN) and cytoplasm (c). 310 360. C) Degenerating body within a mac-
rophage (Ma). 34000. D) Higher magnification showing a degenerating
cytoplasm, accumulation of lipid droplets, a desegregation
nucleus (N) and organelles (arrows) engulfed by macrophage cytoplasm of smooth ER into myelin bodies, and an increase in the
(Ma). 38400. E) Shrunken lutein cells containing lipid (L), lysosomes (Ly), number of autophagosomes [44]. Sawyer et al. [24] pro-
crystals (arrows), and morphologically normal nuclei (N). 34900. posed that the initial site of action of prostaglandin is the
1478 FRASER ET AL.

lutein cell, where it causes an inhibition of steroidogenesis, structures within one cycle, while in the rat, the luteal tissue
together with an increase in oxytocin release that in turn of previous cycles remains in the ovary.
stimulates prostaglandin secretion from the uterus. At the There is evidence that death of the endothelial cells by
same time the lumina of small blood vessels within regress- apoptosis, with attendant reduction in blood supply, could
ing corpora lutea become filled with cellular debris, most play an important part in the luteolytic process, at least in
of which represents fragments of capillary endothelial cells some species [23, 24]. There was little evidence of endo-
[22, 24]. This cascade would result in ischemia and hypoxia thelial cell death by apoptosis in the present study, but this
leading to apoptosis of endothelial cells, followed later by is likely to be underestimated because of the small size of
the apoptosis of lutein cells. the blood vessels and the attenuated nature of the endothe-
In previous reports we described the occurrence of ap- lial lining. Dead and dying cells would exfoliate into the
optosis following natural and induced luteolysis in the mar- lumina of affected vessels, with the remaining viable en-
moset using light microscopy and 39 end-labeling to detect dothelium and basal lamina normally forming a barrier be-
apoptotic cells [14, 30, 45]. The same protocols and study tween dead cells and other cell types [23]. The engulfment
of the apoptotic cells would be limited to the adjacent (en-

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


periods were employed in the current experiments, and
some of the ovaries were employed in both approaches. dothelial) cells or perhaps the pericytes, which have a low
Indeed, in the semithin sections examined, structures re- phagocytic capability. The dying cells may undergo further
sembling apoptotic bodies were observed, although this degeneration in the lumen, but most if not all of these frag-
classification could not be established with certainty, par- ments might be expected to be lost via the flushing effects
ticularly in comparison with degenerating granulosa cells. of continued blood flow. Indeed, a study of the regressing
However, under the electron microscope, these structures in bovine corpus luteum [47] indicated that most of the en-
the corpus luteum were considered to be degenerating cells dothelial cells detach from the basement membrane before
showing either autophagocytosis or nonlysosomal disinte- undergoing apoptosis so that apoptotic endothelial cells
gration according to the classifications of these types of cell were not observed in tissue sections. In the current study,
death, and that of apoptosis, reviewed by Clarke [35]. In there is no evidence either for or against the concept that
the case of autophagocytosis, the pyknosis or heterochro- endothelial cell death leads to the degenerative changes ob-
matin clumping phenomenon is not prevalent. The nucleus served in the lutein cells.
Finally, a review of the literature shows that degenera-
ultimately disintegrates and is digested by autolysosomes,
tive changes in the cells of other endocrine target tissues
which are the abundant and characteristic feature seen in or hormone-producing tissues, such as the rodent uterine
this type of lutein cell degeneration. Ultimately the lutein epithelium [48], marmoset Leydig cells [49], rat, rabbit, and
cell debris or fragments are destroyed by macrophages, i.e., primate ovarian theca cells [11], and the prostate and pi-
by heterophagocytosis. In the case of nonlysosomal disin- tuitary gland [50], may also show nonapoptotic death. The
tegration, organelles swell, and empty spaces arise by fu- current results suggest that as far as the marmoset is con-
sion, with cell destruction achieved by fragmentation. The cerned, and probably other primates, more emphasis should
destruction of the nucleus is delayed (in contrast to what be on understanding the nonapoptotic forms of cell death
occurs in apoptosis), and it disintegrates in a manner similar that we have shown to be predominant during luteal re-
to that for the cytoplasm. Cytoplasmic lysosomes do not gression.
contribute to autophagolysosomes, and there is no signifi-
cant engulfment of cellular debris by macrophages. ACKNOWLEDGMENTS
In situ 39 end-labeling of DNA fragments in cells of the
regressing corpus luteum, and demonstration of laddering We thank Prof. A.H. Wyllie for helpful discussions; K. Morris and
staff for animal management; S. McKenzie, M. Millar, and S. MacPherson
patterns on gels of DNA isolated from regressing corpora for preparation of the sections; R. McDougal, Department of Anatomy,
lutea, have been shown in all species reported—adding University of Edinburgh, for assistance with electron microscopy; and Dr.
weight to the argument that luteolysis involves apoptosis R. Deghengi (Europeptides) for the gift of Antarelix.
[16–21, 28, 30, 45, 46]. However, as pointed out in a num-
ber of such reports, the DNA laddering is accompanied by REFERENCES
smear patterns indicating nonapoptotic degradation of nu-
1. Fraser HM, Nestor JJ Jr, Vickery BH. Suppression of luteal function
clear chromatin [28, 45, 46]. This has led these workers to by a luteinizing hormone-releasing hormone antagonist during the ear-
suggest that luteolytic cell death might be a combination of ly luteal phase in the stumptailed macaque monkey and the effects of
apoptosis-type and necrotic-type degeneration, a concept subsequent administration of human chorionic gonadotropin. Endo-
supported by the present study. In the studies on human crinology 1987; 121:612–618.
and monkey corpora lutea that combined 39 end-labeling 2. Zeleznik AJ, Benyo DF. Control of follicular development, corpus
luteum function, and the recognition of pregnancy in higher primates.
with morphological analysis, study of luteal tissue was lim- In: Knobil E, Neill JD (eds.), The Physiology of Reproduction. New
ited to light microscopic examination [28–30]. The most York: Raven Press; 1994: 751–782.
likely explanation for these findings is that the studies under 3. Webley GE, Hodges JK, Given A, Hearn JP. Comparison of the lu-
the light microscope (using larger cross-sectional-area his- teolytic action of gonadotrophin-releasing hormone antagonist and
tological sections compared with epoxy resin sections) de- cloprostenol, and the ability of human chorionic gonadotrophin and
tected both true apoptotic cells together with structures that melatonin to override their luteolytic effects in the marmoset monkey.
J Endocrinol 1991; 128:121–129.
appeared both morphologically and by 39 end-labeling to 4. Dubourdieu S, Charbonnel B, Massai M, Marraoul J, Spitz I, Bou-
be apoptotic cells or bodies but that do not, in fact, fit the chard P. Suppression of corpus luteum function by the gonadotropin
ultrastructural criteria for apoptosis. Although an important releasing hormone antagonist Nal Glu: effect of the dose and timing
difference between primates and nonprimates appears to be of human chorionic gonadotropin administration. Fertil Steril 1991;
related to the extent to which apoptosis occurs, such dif- 56:440–445.
5. Green JA, Maquedo M. Ultrastructure of the human ovary. Am J
ferences are also apparent between nonprimates. For ex- Obstet Gynecol 1965; 92:946–957.
ample, the dramatic apoptosis observed in the hamster cor- 6. Corner GW Jr. The histological dating of the human corpus luteum
pus luteum [21] accounts for the disappearance of these of menstruation. Am J Anat 1956; 98:377–401.
LUTEAL CELL DEATH IN THE PRIMATE 1479

7. Adams EC, Hertig AT. Studies on the human corpus luteum. I. Ob- function of follicle and corpus luteum status. J Clin Endocrinol Metab
servations on the ultrastructure of development and regression of the 1997; 82:3148–3155.
luteal cells during the menstrual cycle. J Cell Biol 1969; 41:696–715. 30. Young FM, Illingworth PJ, Lunn SF, Harrison DJ, Fraser HM. Cell
8. Clement PB. Histology of the ovary. In: Sternberg SS (ed.), Histology death during luteal regression in the marmoset monkey (Callithrix
for Pathologists. Philadelphia: Lippincott-Raven Press; 1997: 929– jacchus). J Reprod Fertil 1997; 111:109–119.
959. 31. Kirton JT, Koering MJ. Prostaglandin F2a and primate corpus luteum:
9. Lei ZM, Chegini N, Rao CV. Quantitative cell composition of human a correlation of structure and function. Fertil Steril 1973; 24:926–934.
and bovine corpora lutea from various reproductive states. Biol Re- 32. Smith KB, Lunn SF, Fraser HM. Inhibin secretion during the ovula-
prod 1991; 44:1148–1156. tory cycle and pregnancy in the common marmoset monkey. J En-
10. Joel RV, Foraker AG. Fate of the corpus albicans: a morphologic docrinol 1990; 126:489–495.
approach. Am J Obstet Gynecol 1960; 80:314–316. 33. Summers PM, Wennink J, Hodges JK. Cloprostenol-induced luteolysis
11. Hsueh AJW, Billig H, Tsafriri A. Ovarian follicle atresia: a hormonally in the marmoset monkey (Callithrix jacchus). J Reprod Fertil 1985;
controlled apoptotic process. Endocr Rev 1994; 15:707–724. 73:133–138.
12. Tilly JL. Apoptosis and ovarian function. Rev Reprod 1996; 1:162– 34. Webley GE, Richardson MC, Smith CA, Masson GM, Hearn JP. Size
172. distribution of luteal cells from pregnant and non-pregnant marmoset
13. Rodger FE, Fraser HM, Krajewski S, Illingworth PJ. Production of monkeys and a comparison of the morphology of marmoset luteal

Downloaded from https://academic.oup.com/biolreprod/article/61/6/1468/2734586 by guest on 08 December 2020


the proto-oncogene BAX does not vary with changing in luteal func- cells with those from the human corpus luteum. J Reprod Fertil 1990;
tion in women. Mol Hum Reprod 1998; 4:27–32. 90:427–437.
14. Fraser HM, Lunn SF, Cowen GM, Illingworth PJ. Induced luteal re- 35. Clarke PGH. Developmental cell death: morphological diversity and
gression in the primate: evidence for apoptosis and changes in c-myc multiple mechanisms. Anat Embryol 1990; 181:195–213.
protein. J Endocrinol 1995; 147:131–137. 36. Wyllie AH, Kerr JFR, Currie AR. Cell death: the significance of ap-
15. Rueda BR, Tilly KI, Botros IW, Jolly PD, Hansen TR, Hoyer PB, optosis. Int Rev Cytol 1980; 68:251–306.
Tilly JL. Increased bax and interleukin-1b-converting enzyme mes- 37. Kressel M, Groscurth P. Distinction of apoptotic and necrotic cell
senger ribonucleic acid levels coincide with apoptosis in the bovine death by in situ labelling of fragmented DNA. Cell Tissue Res 1994;
corpus luteum during structural regression. Biol Reprod 1997; 56: 278:549–556.
186–193. 38. Christensen AK, Gillim SW. The correlation of fine structure and
16. Kenny N, Williams RE, Kelm LB. Spontaneous apoptosis of cells function in steroid-secreting cells, with emphasis on those of the go-
prepared from the non-regressing corpus luteum. Biochem Cell Biol nads. In: McKerns WW (ed.), The Gonads. New York: Appleton-
1994; 72:531–536. Century-Crofts; 1972: 415–488.
17. Juengel JL, Garverick A, Johnson AL, Youngquist RS, Smith MF. 39. Fraser HM, Sellar RE, Illingworth PJ, Eidne KA. GnRH receptor
Apoptosis during luteal regression in cattle. Endocrinology 1993; 132: mRNA expression by in-situ hybridization in the primate pituitary and
249–254. ovary. Mol Hum Reprod 1996; 2:117–121.
18. Zheng J, Fricke PM, Reynolds LP, Redmer DA. Evaluation of growth, 40. Michael AE, Abayasekara DRE, Webley GE. Cellular mechanisms of
cell proliferation, and cell death in bovine corpora lutea throughout luteolysis. Mol Cell Endocrinol 1994; 99:R1-R9.
the estrous cycle. Biol Reprod 1994; 51:623–632. 41. Zeleznik AJ. In vivo responses of the primate corpus luteum to lu-
19. Dharmarajan AM, Goodman SB, Tilly KI, Tilly JL. Apoptosis during teinizing hormone and chorionic gonadotropin. Proc Natl Acad Sci
functional corpus luteum regression: evidence of a role for chorionic USA 1998; 95:11002–11007.
gonadotropin in promoting luteal cell survival. Endocr J 1994; 2:295– 42. Salazar H, Furr BJA, Smith GK, Bentley M, Gonzalez-Angulo A.
303. Luteolytic effects of a prostaglandin analogue, cloprostenol (ICI
20. Gaytán F, Bellido C, Morales C, Sánchez-Criado JE. Both prolactin 80,996) in rats: ultrastructural and biochemical observations. Biol Re-
and progesterone in proestrus are necessary for the induction of ap- prod 1976; 14:458–472.
optosis in the regressing corpus luteum of the rat. Biol Reprod 1998; 43. Stacy BD, Gemmell RT, Thorburn GD. Morphology of the corpus
luteum in the sheep during regression induced by prostaglandin F2a.
59:1200–1206.
Biol Reprod 1976; 14:280–291.
21. McCormack JT, Friedrichs MG, Limback SD, Greenwald GS. Apo-
44. McLellan MC, Abel JHJ, Niswender GD. Function of lysosomes dur-
ptosis during spontaneous luteolysis in the cyclic golden hamster: bio-
ing luteal regression in normally cycling and PGF2a-treated ewes. Biol
chemical and morphological evidence. Biol Reprod 1998; 58:255–
Reprod 1977; 16:499–512.
260. 45. Young FM, Illingworth PJ, Fraser HM. Ubiquitin and apoptosis in the
22. O’Shea JD, Nightingale MG, Chamley WA. Changes in small blood corpus luteum of the marmoset monkey (Callithrix jacchus). J Reprod
vessels during cyclical luteal regression in sheep. Biol Reprod 1977; Fertil 1998; 114:163–168.
17:162–177. 46. Rueda BR, Wegner JA, Marion SL, Wahlen DD, Hoyer PB. Internu-
23. Azmi TI, O’Shea JD. Mechanism of deletion of endothelial cells dur- cleosomal DNA fragmentation in ovine luteal tissue associated with
ing regression of the corpus luteum. Lab Invest 1984; 51:206–217. luteolysis: In vivo and in vitro analyses. Biol Reprod 1995; 52:305–
24. Sawyer HR, Niswender KD, Braden TD, Niswender GD. Nuclear 312.
changes in ovine luteal cells in response to PGF2a. Domest Anim 47. Modlich U, Kaup FJ, Augustin HG. Cyclic angiogenesis and blood
Endocrinol 1990; 72:229–238. vessel regression in the ovary: blood vessel regression during luteol-
25. Behrman HR, Endo T, Aten RF, Musicki B. Corpus luteum function ysis involves endothelial cell detachment and vessel occlusion. Lab
and regression. Reprod Med Rev 1993; 2:153–180. Invest 1996; 74:771–780.
26. Quatacker JR. Formation of autophagic vacuoles during human corpus 48. Welsh AO, Enders AC. Chorioallantoic placenta formation in the rat.
luteum involution. Z Zellforsch 1971; 122:479–487. III. Granulated cells invade the uterine luminal epithelium at the time
27. Koering MJ, Wolf RC, Meyer RK. Morphological changes in the cor- of epithelial cell death. Biol Reprod 1993; 49:38–57.
pus luteum correlated with progestin levels in the rhesus monkey dur- 49. Prince F, Mann DR, Fraser HM. Blockade of the pituitary testicular
ing early pregnancy. Biol Reprod 1973; 9:254–271. axis by a gonadotrophin releasing hormone antagonist in the neonatal
28. Shikone T, Yamoto M, Kokawa K, Yamashita K, Nishimori K, Nak- marmoset: changes in Leydig cell ultrastructure. Cell Tissue Res 1998;
ano R. Apoptosis of human corpora lutea during cyclic luteal regres- 30:651–661.
sion and early pregnancy. J Clin Endocrinol Metab 1997; 81:2376– 50. Aoki MP, Maldonado CA, Aoki A. Apoptotic and non-apoptotic cell
2380. death in hormone-dependent glands. Cell Tissue Res 1998; 291:571–
29. Yuan W, Giudice LC. Programmed cell death in human ovary is a 574.

You might also like