regulation of micro environment within the oviduct

70
REGULATION OF MICRO-ENVIRONMENT WITHIN THE OVIDUCT September, 2016 Yoshihiko KOBAYASHI Graduate School of Environmental and Life Science (Doctor’s Course) OKAYAMA UNIVERSITY .

Upload: others

Post on 28-Jul-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

REGULATION OF MICRO-ENVIRONMENT

WITHIN THE OVIDUCT

September, 2016

Yoshihiko KOBAYASHI

Graduate School of Environmental and Life Science

(Doctor’s Course)

OKAYAMA UNIVERSITY

.

Page 2: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

CONTENTS

PREFACE ------------------------------------------------------------------------------------------------- 1

ACKLOWLEDGEMENT ------------------------------------------------------------------------------- 2

ABSTRACT ---------------------------------------------------------------------------------------------- 3

CHAPTER 1 GENERAL INTRODUCTION ----------------------------------------- 5

CHAPTER 2 REGULATION OF BOVINE OVIDUCTAL NO SYNTHESIS

BYFOLLICULAR STEROIDS AND PROSTAGLANDINS

INTRODUCTION -------------------------------------------------------- 7

RESULTS ------------------------------------------------------------------ 8

DISCUSSION ----------------------------------------------------------- 18

SUMMARY -------------------------------------------------------------- 22

CHAPTER 3 ROLES OF EDNS IN REGULATING OVIDUCTAL NO

SYNTHESIS AND SMOOTH MUSCLE MOTILITY IN COWS

INTRODUCTION ------------------------------------------------------ 23

RESULTS ---------------------------------------------------------------- 24

DISCUSSION ----------------------------------------------------------- 31

SUMMARY -------------------------------------------------------------- 35

CHAPTER 4 SUMMER HEAT STRESS AFFECTS PROSTAGLANDIN

SYNTHESIS IN THE BOVINE OVIDUCT

INTRODUCTION ------------------------------------------------------ 36

RESULTS ---------------------------------------------------------------- 37

DISCUSSION ----------------------------------------------------------- 42

SUMMARY -------------------------------------------------------------- 45

CHAPTER 5 CONCLUSIONS -------------------------------------------------------- 46

MATERIALS AND METHODS --------------------------------------------------------------------- 47

REFERENCES ------------------------------------------------------------------------------------------ 58

ABSTRACT IN JAPANESE -------------------------------------------------------------------------- 66

Page 3: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

1

PREFACE

The experiments described in this dissertation were carried out at the Graduate School of

Environmental and Life Science (Doctor’s course), Okayama University, Japan, from April,

2014 to February, 2016, under the supervision of Professor Emeritus Kiyoshi OKUDA (April,

2014 – December, 2015) and Professor Koji KIMURA (January, 2016 – September, 2016).

This dissertation has not been submitted previously in whole or in part to a council,

university or any other professional institution for a degree, diploma or other professional

qualifications.

Yoshihiko KOBAYASHI

September, 2016

Page 4: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

2

ACKNOWLEDGEMENT

I wish to express my deep gratitude to Kiyoshi OKUDA, DVM, Ph.D., Professor emeritus of

Okayama University, Okayama, Japan and President of Obihiro University of Agriculture and

Veterinary Medicine, Obihiro, Japan, for his encouragement, guidance, constructive criticisms

and excellent supervision. I also wish to be grateful to Koji KIMURA, Ph.D., Professor of

Graduate School of Environmental and Life Science, Okayama University, and Yuki

YAMAMOTO, DVM, Ph.D., Assistant Professor of Okayama University, for their supports,

advices, encouragement to conduct the present study. I am grateful to Hiroki HIRAYAMA,

Ph.D., Associate Professor of Tokyo University of Agriculture, Abashiri, Japan for collecting

follicular fluids. I appreciate Toshimitsu HATABU, Ph.D., Associate Professor of Graduate

School of Environmental and Life Science, Okayama University and Yuka YOSHIMOTO,

M.Sc., a graduate of Laboratory of Animal Reproductive Physiology for supporting the

isometric contraction test. Con-focal microscopic images were obtained with the cooperation

of Department of Instrumental Analysis, Advanced Science Research Center, Okayama

University. It is a pleasure to express my thanks to all the members of Laboratory of Animal

Reproductive Physiology for helping me during carrying out this study.

Page 5: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

3

ABSTRACT

Summer heat stress negatively affects the functions of female reproductive tracts in cattle.

Heat stress decreases estradiol-17β (E2) concentrations in follicular fluid of the dominant

follicle. In vitro studies indicate that heat stress stimulates prostaglandin (PG) F2α secretion

from the endometrial tissue, suggesting that it induces early regression of the corpus luteum

and kills the embryo. Hot conditions also induce mortality of early embryos. Early embryos

develop within the oviduct during a few days after fertilization. This development is supported

by molecules secreted from the oviductal epithelium. These molecules also play roles in the

smooth muscle motility for the transport of embryos. Little is known about the effects of

summer heat stress on oviductal functions.

In early pregnancy, mammalian oviducts provide transient micro-environments for

each of several events including capacitation and super-activation of spermatozoa, transport

of gametes, fertilization and early embryonic development and transport. Oviductal functions

are controlled by the steroids E2 and progesterone (P4) that are released from the ovary. These

steroids regulate the secretion of molecules including PGE2, PGF2α and endothelins (EDNs)

that affect smooth muscle motility of the oviduct. Additionally, these molecules which are

secreted by oviductal epithelial cells are considered to regulate oviductal secretion by

autocrine, producing micro-environment within the oviduct. However, detailed mechanisms

of the regulation remain unclear.

In the present study, at first, the regulation of NO synthesis by follicular steroids and

PGs was investigated to clarify the regulatory mechanisms of oviductal secretions by ovaries.

Secondly, roles of EDNs which are secreted by oviductal epithelial cells in NO synthesis

within the oviduct to elucidate the autocrine regulation in the oviductal epithelium. The

differences of mRNA expressions of PG synthases in the bovine oviduct between summer and

winter and the effects of elevated temperature on PG secretions in cultured epithelial cells

were also investigated to clarify the effects of summer heat stress on the secretory functions

of the oviduct.

Quantification of inducible NO synthase (iNOS) mRNA expression in the ampulla

showed that iNOS mRNA expression was highest on the day of ovulation (Day 0) in oviductal

tissue. PGE2 and PGF2α increased iNOS mRNA expression in cultured ampullary oviductal

epithelial cells after 1 h incubation. In contrast, iNOS mRNA expression in the isthmus was

highest at Days 5-6 and lowest at Days 19-21. Expression of estrogen receptor α (ESR1)

Page 6: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

4

mRNA was highest at Days 19-21. Twenty-four-hour exposure to E2 decreased the expression

of iNOS mRNA expression in cultured isthmic epithelial cells. EDN1 stimulated iNOS mRNA

and protein expressions only in the ampullary epithelial cells. Only in the ampullary tissues,

the expression of EDN receptor type B was increased after ovulation than before ovulation.

HSP90 and cytosolic PGE synthase (cPGES) mRNA expressions in oviductal tissue in

summer were higher than in winter. On the other hand, the expression of carbonyl reductase

1 (CBR1), which catalyze the convert from PGE2 to PGF2α, was lower in summer than in

winter. Elevated temperature (40.5°C) stimulated the secretions of PGE2 in cultured ampullary

oviductal epithelial cells.

Our study indicates the presence of different regulatory mechanisms of NO in

different regions of the oviduct. In the ampulla, PGE2 and PGF2α contained in follicular fluid

entering the oviduct immediately after ovulation increase the synthesis of NO which

contributes to oocyte/embryo survival. In the isthmus, NO synthesis is suppressed by E2

released from the dominant follicle to the circulation at before ovulation. In addition to the

regulation by ovaries, oviductal secretion was indicated to be regulated by the oviduct itself.

These multiple ways of regulating oviductal secretions may provide optimal micro-

environment for the events in the first days of pregnancy. Under hot summer conditions,

secretions of PGE2 and NO, which are relaxant factors, upset optimal waves of contraction

and relaxation of the smooth muscle, resulting in low fertility.

Page 7: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

5

CHAPTER 1 GENERAL INTRODUCTION

Effects of summer heat on a fertility in cattle

Conception rates in warm months are lower than in cold months in cattle (Gwazdauskas et al.

1975). Summer heat stress weakens estrous behavior and suppresses folliculogenesis,

inducing low fertility (Cavestany et al. 1985, De Rensis & Scaramuzzi 2003, Sakatani et al.

2004, Collier et al. 2006). Hot conditions negatively affect the functions of female

reproductive tracts in cattle. For example, heat stress decreases estradiol-17β (E2)

concentrations in follicular fluid of the dominant follicle (Wolfenson et al. 1997, Bridges et

al. 2005). In vitro studies indicate that heat stress stimulates prostaglandin (PG) F2α secretion

from the endometrial tissue (Putney et al. 1988, Malayer et al. 1990), suggesting that it induces

early regression of the corpus luteum, leading to death of the embryo. Hot conditions also

induce mortality of early embryos in vitro (Sakatani et al. 2004). Early embryos develop

within the oviduct during a few days after fertilization. This development is supported by

molecules secreted from the oviductal epithelium. These molecules also play roles in smooth

muscle motility, which is needed for transport of embryos. However, little is known about the

effects of summer heat stress on the oviductal secretions.

Oviductal physiology in reproductive events

Mammalian oviducts are the starting site of pregnancy. To achieve pregnancy, the oviduct

provides transient micro-environments for each of several events including capacitation and

super-activation of spermatozoa, transport of gametes, fertilization and early embryonic

development and transport (Hunter 2012). The fimbria, which is the distal entrance of the

oviduct, receives the ovulated cumulus-oocyte complex (COC) (Croxatto 2002). A stream of

oviductal fluid which is produced by ciliary beating of ciliated epithelial cells of the oviduct

transports the COC to the site of fertilization, the ampulla (Croxatto 2002). A Fertilized egg

is transported by ciliary beating and waves of contraction and relaxation which is produced

by the thick smooth muscle of the isthmus. The early embryo grows to around the 16-cell

stage within the oviduct (Ulbrich et al. 2010). This growth is stimulated by molecules that are

secreted by oviductal epithelial cells (Ulbrich et al. 2010). Oviductal functions are controlled

by two steroids, E2 and progesterone, that are released from the ovary, whose morphology

dramatically changes with progression of the estrous cycle (Ulbrich et al. 2003). Receptors of

Page 8: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

6

these hormones are expressed in the oviduct (Ulbrich et al. 2003). These ovarian steroids

regulate the secretion of molecules including PGE2, PGF2α and endothelins (EDNs)

(Wijayagunawardane et al. 1999, Wijayagunawardane et al. 2001a, Priyadarsana et al. 2004,

Jeoung et al. 2010) that affect the smooth muscle motility of the oviduct. Additionally, these

molecules which are secreted by oviductal epithelial cells are considered to regulate oviductal

secretion by autocrine, producing micro-environment within the oviduct. However, detailed

mechanisms of the regulation remain unclear.

Aim of the present study

At first, the regulation of NO synthesis by follicular steroids and PGs was investigated to

clarify the regulatory mechanisms of oviductal secretions by ovaries. Secondly, roles of EDNs

which are secreted by oviductal epithelial cells in NO synthesis within the oviduct to elucidate

the autocrine regulation in the oviductal epithelium. The differences of mRNA expressions of

PG synthases in the bovine oviduct between summer and winter and the effects of elevated

temperature on PG secretions in cultured epithelial cells were also investigated to clarify the

effects of summer heat stress on the secretory functions of the oviduct.

Page 9: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

7

CHAPTER 2 Regulation of bovine oviductal NO synthesis

by follicular steroids and prostaglandins

INTRODUCTION

In mammals, the oviduct is essential organ for transport of gametes and embryos (Menezo &

Guerin 1997, Suarez 2008). Ovulated cumulus-oocyte complexes (COCs) and sperms are

transported to the ampulla of the oviduct where fertilization occurs (Ulbrich et al. 2010). Early

embryos are transported to the uterus within a few days (Freeman et al. 1991). The transport

of gametes and embryos is induced by alternate waves of contraction and relaxation of the

oviductal smooth muscle and the ciliary beating of oviductal epithelial cells (Halbert et al.

1976, Hunter 2012). Various substances including nitric oxide (NO), ovarian steroids and

prostaglandins (PGs) regulate the waves of contraction and relaxation (Helm et al. 1982,

Rosselli et al. 1994, Ekerhovd et al. 1997, Ekerhovd et al. 1999, Al-Alem et al. 2007).

Nitric oxide is a physiological mediator of numerous cellular and organ functions

(Ignarro et al. 2001). It is synthesized from L-arginine by NO synthases (NOSs). Nitric oxide

synthases have three isoforms, neuronal NOS (nNOS), inducible NOS (iNOS) and endothelial

NOS (eNOS) (Ulbrich et al. 2006). Nitric oxide relaxes the oviductal smooth muscle for

gamete/embryo transport (Rosselli et al. 1994, Yilmaz et al. 2012). In addition, NO has crucial

roles in gamete/embryo activity and survivability. NO stimulates the motility of spermatozoa

(Miraglia et al. 2011), and the inhibition of NO synthesis increases the mortality of early

embryos in mouse (Gouge et al. 1998).

Each-oviductal region has various specific functions. The ampulla transports

fertilized oocytes to the site of fertilization, and supports fertilization (Hunter 2012). The

isthmus promotes embryo development by secreting various molecules, and transports

embryos to the uterus by smooth muscle activity (Hunter 2012). To perform these functions,

the oviductal regions are morphologically different. For example, the proportion of two-types

of epithelial cells, ciliated cells and secretory cells, is different among regions of the bovine

oviduct (Kölle et al. 2009). In the murine oviduct, the ampullary region has high ciliary

activity while the isthmus has strong muscular contraction and weak ciliary activity (Noreikat

et al. 2012) The expressions of genes for endothelins, PG synthases and sex steroid receptors

are also different among regions of the oviduct (Ulbrich et al. 2003, Gauvreau et al. 2010,

Page 10: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

8

Jeoung et al. 2010, Saint-Dizier et al. 2012). Similarly, iNOS mRNA expression at the day of

ovulation is higher in the ampulla than in the isthmus of the bovine oviduct (Ulbrich et al.

2006). Thus, NO synthesis may be regulated differently in the ampulla and isthmus.

Ovarian steroids are major molecules controlling oviductal functions

(Wijayagunawardane et al. 1999, Ulbrich et al. 2003, Szóstek et al. 2011). During the estrous

cycle except at ovulation, these steroids participate in oviductal physiological events via

systemic way. The highest levels of estrogen in circulation and oviductal tissue at the follicular

phase (Wijayagunawardane et al. 1998, Acosta et al. 2003) support that idea. At ovulation,

follicular fluid with COC enters the oviduct through fimbria, the entrance of the oviduct. Since

follicular fluid contains high concentrations of PGE2, PGF2α and ovarian steroids (Acosta et

al. 1998, Acosta et al. 2000), these molecules are possible to influence oviductal milieu.

In the present study, we hypothesized that molecules derived from follicles

participate oviductal NO synthesis via two distinct routes: 1) a systemic way via blood vessels

during the estrous cycle, and 2) a direct way through fimbria only at ovulation. To test the

above hypothesis, we determined 1) cyclic changes of NOSs and receptors for estrogen (ESRs),

progesterone (PGR), PGE (PTGERs) and PGF (PTGFR) expressions in bovine ampullary and

isthmic oviductal tissues during the estrous cycle, and 2) the effects of exogenous E2, P4, PGE2

and PGF2α on NO synthesis in cultured ampullary and isthmic oviductal epithelial cells.

RESULTS

Expressions of NOSs in oviductal tissues

Inducible NOS (iNOS) mRNA expression in the ampulla was highest at the day of ovulation

(Day 0) (Fig. 1A, P<0.05). In the isthmus, iNOS mRNA expression was highest at Days 5-6

and gradually decreased toward the follicular phase. iNOS expression in the isthmus was

lowest at Days 19-21 (Fig.1A P<0.05). On the other hand, endothelial NOS (eNOS)

expressions in both segments did not significantly change during the estrous cycle. Proteins

of iNOS and eNOS were immunohistochemically detected in epithelial, stromal and smooth

muscle layers of the ampullary region (Fig. 1B). In the isthmus, eNOS was expressed in the

epithelium, stroma, and smooth muscle. iNOS was clearly detected in epithelium, but only

weakly detected in stroma and smooth muscle. Activity of NO synthesis by iNOS is well

known to be extremely higher than by eNOS (Moncada & Higgs 1993). Since eNOS

expression did not change during the estrous cycle in the present study, only iNOS expression

Page 11: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

9

was investigated in the following experiments.

Expressions of ESRs, PGR, PTGERs and PTGFR

ESR1 mRNA expression was highest at Days 19-21 (follicular phase) in isthmic oviductal

tissues, while the expression of ESR1 mRNA did not change in ampullary tissues during

estrous cycle (Fig. 2A). The mRNAs of other steroid and PG receptors were stably expressed

in both ampullary and isthmic tissues during the estrous cycle (Fig. 2A, 3A). Proteins of ESR1

and PGR were localized in epithelial cells of the ampulla and isthmus of the oviduct (Fig. 2B).

PGR protein was detected in smooth muscle layer of the isthmic region (Fig. 2B). Proteins of

PTGER2, PTGER4 and PTGFR were distributed in epithelium of both regions (Fig. 3B).

PTGER2 and PTGFR were also detected in stromal and smooth muscle layer (Fig. 3B).

Concentrations of ovarian steroids and PGs in follicular fluid immediately before ovulation

Concentrations of E2, P4, PGE2 and PGF2α in follicular fluid obtained from cows which were

treated to induce super-ovulation were determined. Ranges of concentrations of E2, P4, PGE2

and PGF2α were 106.3-409.9 nmol/L, 771.0-2763 nmol/L, 5.68-40.1 nmol/L and 61.9-835

nmol/L, respectively (Table 1).

Effects of estradiol-17β (E2) or progesterone (P4) on iNOS mRNA expression

To determine how NO synthesis in the oviduct is regulated, we investigated the effects of

ovarian steroid hormones (E2 or P4) on iNOS mRNA expression in cultured oviductal epithelial

cells. Estradiol-17β suppressed iNOS mRNA expression and NO production in isthmic

epithelial cells after 24 h incubation, but not in ampullary epithelial cells (Fig. 4A).

Progesterone stimulated iNOS mRNA expression after 24 h incubation but not after 1 or 4 h

incubations. By contrast, P4 did not affect iNOS expression in epithelial cells obtained from

the isthmus (Fig. 4A). An ESR1 antagonist blocked the inhibiting the effect of E2 on iNOS

expression, whereas an ESR2 antagonist did not (Fig. 4B).

Effects of PGs on iNOS mRNA expression in cultured oviductal epithelial cells

Inducible NOS mRNA expression was increased by PGE2 and PGF2α after 1 h incubation,

whereas iNOS mRNA expression was decreased after 24 h incubation in ampullary epithelial

cells (Fig. 5). On the other hand, iNOS mRNA expression was not affected by PGs in isthmic

epithelial cells. A PTGER2 antagonist decreased iNOS mRNA expression stimulated by PGE2

Page 12: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

10

after 1 h incubation, while a PTGER4 antagonist did not (Fig. 5B).

Differential expression of iNOS mRNA between the ampulla and ithsmus

In both oviductal tissues and cultured epithelial cells, iNOS mRNA expression was higher in

the ampulla than in the isthmus (Fig, 6).

Page 13: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

11

Figure 1. A) Cyclic changes of inducible nitric oxide synthase (iNOS) and endothelial NOS

(eNOS) mRNA expressions in tissues collected from the ampulla and isthmus of the bovine

oviduct (mean ± SEM, n=5 oviducts). Significance of differences was assessed by analysis of

variance (ANOVA) followed by Tukey-Kramer test for multiple comparisons. Different

superscript letters indicate significant differences (P<0.05). B) Immunohistochemical analysis

of NOS protein distributions in the ampulla and isthmus of the oviduct obtained from cows at

day of ovulation (L: lumen, E: epithelium, ST: stroma, SM: smooth muscle). Green (Alexa

488) indicates each target protein (iNOS or eNOS), and Blue (DAPI) indicates nuclei of the

cells. Scale is the same in all the photomicrographs.

Page 14: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

12

Figure 2. A) Cyclic changes of estrogen receptor α (ESR1), ESR2 and progesterone receptor

(PGR) mRNA expressions in tissues collected from the ampulla and isthmus of the bovine

oviduct (mean ± SEM, n=5 oviducts). Significance of differences was assessed by analysis of

variance (ANOVA) followed by Tukey-Kramer test for multiple comparisons. Different

superscript letters indicate significant differences (P<0.05). B) Immunohistochemical analysis

of ESR1 and PGR protein distributions in the ampulla and isthmus of the oviduct obtained

from cows at follicular stage (L: lumen, E: epithelium, ST: stroma, SM: smooth muscle).

Green (Alexa 488) indicates each target protein (ESR1 or PGR), and Blue (DAPI) indicates

nuclei of the cells. Scale is the same in all the photomicrographs.

Page 15: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

13

Figure 3. A) Cyclic changes of E-prostanoid receptor 2 (PTGER2), PTGER4 and F-

prostanoid receptor (PTGFR) mRNA expressions in tissues collected from the ampulla and

isthmus of the bovine oviduct (mean ± SEM, n=5 oviducts). Significance of differences was

assessed by analysis of variance (ANOVA) followed by Tukey-Kramer test for multiple

comparisons. There were no significant differences (P>0.05). B) Immunohistochemical

analysis of PTGER and PTGFR protein distributions in the ampulla and isthmus of the oviduct

obtained from cows at day of ovulation (L: lumen, E: epithelium, ST: stroma, SM: smooth

muscle). Green (Alexa 488) indicates each target protein (PTGER2, PTGER4 or PTGFR), and

Blue (DAPI) indicates nuclei of the cells. Scale is the same in all the photomicrographs.

Page 16: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

14

Table 1 Concentrations of hormones in follicular fluid immediately before ovulation

Hormone Concentration (nmol/L)

E2 106.3-409.9

P4 771-2763

PGE2 5.68-40.1

PGF2α 61.9-835

Table 1. Concentrations of estradiol-17β (E2), progesterone (P4), prostaglandin E2 (PGE2) and

PGF2α in follicular fluid obtained from cows which were treated with super-ovulation.

Follicular fluid was collected 26-27 h after administering a GnRH analogue.

Page 17: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

15

Figure 4. A) Effects of estradiol-17β (E2) or progesterone (P4) on iNOS mRNA expression in

the oviductal epithelial cells isolated from the ampulla (black bar) and isthmus (gray bar) of

the oviduct (mean ± SEM, n=8 oviducts, n=12 only in the incubation with P4 for 24 h).

Significance of differences was assessed by analysis of variance (ANOVA) followed by

Tukey-Kramer test for multiple comparisons. Different superscript letters indicate significant

differences (P<0.05). B) Effects of selective antagonists of each ESR on iNOS mRNA

expression in isthmic oviductal epithelial cells (mean ± SEM, n=8 oviducts). MPP

dihydrochloride (ESR1 antagonist) or PHTPP (ESR2 antagonist) was co-incubated with E2

(10 nmol/L) for 24 h. Significance of differences was assessed by analysis of variance

(ANOVA) followed by Tukey-Kramer test for multiple comparisons. Different superscript

letters indicate significant differences (P<0.05).

Page 18: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

16

Figure 5. A) Effects of prostaglandin (PG) E2 and PGF2α on iNOS mRNA expression in

ampullary and isthmic oviductal epithelial cells (mean ± SEM, n=5 oviducts). Significance of

differences was assessed by analysis of variance (ANOVA) followed by Tukey-Kramer test

for multiple comparisons. Different superscript letters indicate significant differences

(P<0.05). B) Effects of selective antagonists of each PTGER on iNOS mRNA expression in

isthmic oviductal epithelial cells (mean ± SEM, n=7 oviducts). AH6809 (PTGER2 antagonist)

or AH23848 (PTGER4 antagonist) was co-incubated with PGE2 (1 μmol/L) for 1 h.

Significance of differences was assessed by analysis of variance (ANOVA) followed by

Tukey-Kramer test for multiple comparisons. Different superscript letters indicate significant

differences (P<0.05).

Page 19: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

17

Figure 6. Differential expression of iNOS mRNA between the ampulla and isthmus in

oviductal tissues (A, n=6, mean ± SEM) and cultured epithelial cells (B, n=5, mean ± SEM).

Significance of differences was assessed by analysis of variance (ANOVA) followed by

Student’s t-test. Asterisks indicate significant differences (P<0.05).

Page 20: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

18

DISCUSSION

Nitric oxide (NO), a molecule which has been identified as an endothelium-derived relaxing

factor, participates in various physiological events such as the immune response,

neurotransmission and cell-proliferation (Bredt & Snyder 1994, Ignarro et al. 2001). In the

oviduct, NO is also known to relax smooth muscle (Rosselli et al. 1994), and its synthesis is

regulated by ovarian hormones during the estrous cycle (Lapointe et al. 2006, Ulbrich et al.

2006). The present results suggest that prostaglandins (PGs) in follicular fluid as well as

estradiol-17β in blood stream regulate NO synthesis in oviductal epithelial cells. A model of

this regulatory system is shown in Fig. 7. The oviduct is not only the site where an ovulated

oocyte and sperm meet, but it also provides a micro-environment for early embryonic

development in the first days of pregnancy (Ulbrich et al. 2010, Ezzati et al. 2014). The

present finding that NO synthesis in the ampulla and isthmus was regulated by specific

molecules (PGs and E2, respectively) may reflect the idea that the oviduct has region-specific

functions. For example, the ampulla serves to transport the oocyte and to provide an

environment for fertilization, while the isthmus serves to transport spermatozoa and the

embryo and to provide an environment for embryo development.

Estradiol-17β concentrations in the oviduct are highest immediately before ovulation

(Wijayagunawardane et al. 1998), indicating that a large amount of E2 is transported to the

oviduct at the pre-ovulatory stage. In addition, our result showed ESR1 was highly expressed

at the follicular stage in the isthmus (Fig. 2A). These results, together with our finding that E2

decreased NO synthesis in cultured isthmic epithelial cells (Fig. 4A) suggest that the E2

transported to the isthmus of the oviduct suppresses NO synthesis via ESR1 at the follicular

stage. Spermatozoa entering the oviduct bind to cilia of the oviductal epithelium, which

temporarily stops their migration toward the oocyte (Kölle et al. 2009). Because NO is

reported to stimulate the motility of spermatozoa (Miraglia et al. 2011), suppression of NO

synthesis by E2 in the isthmus may decrease the motility of spermatozoa, contributing to their

binding to epithelial cilia until they become hyperactivated. We showed that effects of E2 on

NO synthesis were different between ampullary and isthmic epithelial cells. The suppressing

effect of E2 on iNOS mRNA expression was observed only in isthmic epithelial cells despite

less expression of ESR1 protein in isthmic tissues than in ampullary tissues (Ulbrich et al.

2003). These different effects of E2 may be involved in the specific characters of epithelial

cells in each region of the oviduct. Although oviductal epithelium is composed by two

Page 21: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

19

epithelial cells, ciliated and secretory, the isthmic secretory cells interestingly have short

kinocilia on the cell surface, but not ampullary secretory cells (Kölle et al. 2009). These

differences imply that the secretory cells play specific roles in each region of the oviduct. In

fact, iNOS mRNA expressions in the ampullary tissues and epithelial cells were higher than

in the isthmic tissues and epithelial cells, respectively (Fig. 6). In addition, partial differences

of NO synthases in cyclic epithelium of the oviduct (Ulbrich et al. 2006) and whole layers

containing stroma and smooth muscle of the oviduct (Fig. 1) suggest layer-specific control of

NO synthesis in the oviductal milieu. Further studies are needed to clarify the region-specific

roles of epithelial cells in the oviduct.

The high concentrations of PGE2 and PGF2α in the follicular fluids by injection of

gonadotropin-releasing hormone are consistent with the previous finding that PGE2 and PGF2α

concentrations increase immediately before ovulation, and with the idea that PGs induce

follicular rupture (Acosta et al. 1998). After the rupture, the oocyte and follicular fluid

containing high concentrations of PGs enters the oviduct. There is evidence that PGs affect

NO synthesis in other tissues. For example, PGE2 appears to activate NO synthesis in mouse

aortic epithelium (Hristovska et al. 2007), and PGF2α increases NO production in bovine luteal

endothelial cells (Lee et al. 2009). PGE2 and PGF2α bind to G protein-coupled receptors

(GPCRs), named E-prostanoid receptors or F-prostanoid receptor (Gabler et al. 2008, Zhang

et al. 2010), inducing rapid signal transductions (Hristovska et al. 2007, Siemieniuch et al.

2009). Therefore, our finding that iNOS mRNA expression in ampullary epithelial cells

increased after 1 h incubation with PGs at concentrations similar to that in follicular fluid (Fig.

5A) suggests that PGs in follicular fluid directly stimulate NO synthesis in the ampulla, thus

controlling the oviductal micro-environment immediately after ovulation. Inhibition of NO

synthesis suppresses development of pre-implantation murine embryos in vitro (Manser et al.

2004). NO also delays oocyte aging, suggesting that it has a role in maintaining the quality of

oocytes (Goud et al. 2005). Therefore, immediately after ovulation, follicular fluid might have

a role in maintaining the optimal concentrations of NO in the oviductal cavity for survival of

oocytes and development of embryos. The findings that an NO donor activates ciliary beat

frequency in rat oviductal ciliated cells (Chiu et al. 2010) implies that PGs in follicular fluid

promote oocyte transport to the site of fertilization via stimulating NO production immediately

after ovulation. In contrast to exposing ampullary oviductal epithelial cells to PGs for 1 h,

exposing them to PGs for 24 h decreased iNOS mRNA expression (Fig. 5A). This down-

regulation of iNOS may be caused by other molecules that are secreted by PG stimulation. In

Page 22: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

20

our unpublished data, a NO donor (NONOate) inhibited iNOS mRNA expression in ampullary

oviductal epithelial cells. Thus, iNOS expression might be decreased by NO stimulated by

PGs. In contrast to PGs, P4 increased iNOS expression after 24 h stimulation in cultured

ampullary cells (Fig. 4). Because NO can be involved in both cellular survival and death

(Manser et al. 2004, Gotoh & Mori 2006), NO concentration in the oviductal cavity is possible

to be strictly controlled by various molecules derived from inside and outside of the oviduct.

The isthmus is an crucial region of the oviduct for transport and development of

embryos (Ulbrich et al. 2010). A thick smooth muscle layer surrounding isthmic mucosa

contributes to the waves of contraction and relaxation (Menezo & Guerin 1997, Hunter 2012),

while effective transport driven by ciliary beating is not observed in this region (Noreikat et

al. 2012). Various molecules regulate the motility of oviductal smooth muscle. Contraction is

induced by PGF2α, endothelins, angiotensin II and E2 (Helm et al. 1982, Wijayagunawardane

et al. 2001a, Wijayagunawardane et al. 2001b, Al-Alem et al. 2007, Siemieniuch et al. 2009),

while relaxation is induced by NO, PGE2, luteinizing hormone and P4 (Helm et al. 1982,

Rosselli et al. 1994, Gawronska et al. 1999, Siemieniuch et al. 2009). Some of these molecules

are secreted by the oviductal epithelium (Rosselli et al. 1994, Wijayagunawardane et al. 2009,

Szóstek et al. 2011), contributing to activate motility of the oviductal smooth muscle around

the time of ovulation. In cattle, the embryo reaches the uterus around 4 days after fertilization

(Freeman et al. 1991). Utero-tubal junction is known to strictly contract immediately before

the embryo passes through (Croxatto 2002). Thus, the finding in the present study that iNOS

expression was highest at Days 5-6 after ovulation, implies that a large amount of NO

produced by isthmic epithelial cells relaxes the isthmus and utero-tubal junction and allows

the embryo to enter the uterus. This is supported by the findings that iNOS mRNA expression

in the isthmic oviduct were highest at Day 3.5 of the estrous cycle (Ulbrich et al. 2006).

Inhibition of NO synthesis kills morula embryos in vitro (Gouge et al. 1998). Since the embryo

is considered to reach the uterus at the morula or young blastocyst stage (Hunter 2012), a large

amount of NO produced by isthmic epithelial cells may contribute to embryonic development

several days after fertilization.

In conclusion, our results indicate that the regulatory mechanisms of NO synthesis

are different between the ampulla and isthmus of the oviduct. Our results also suggest that

PGs in follicular fluid regulate NO synthesis in the ampullary oviductal epithelium. The

different regulations of NO synthesis in the ampulla and isthmus might reflect the different

roles of these regions during the first days of pregnancy.

Page 23: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

21

Figure 7. Proposed model to illustrate region-specific regulations of nitric oxide (NO)

synthesis in the ampulla and isthmus of the oviduct. Before ovulation, high level of estradiol-

17β (E2) released from follicles systemically suppressed NO production by isthmic oviductal

epithelial cells via estrogen receptor α (ESR1). Because NO stimulates the motility of

spermatozoa (Miraglia et al. 2011), suppressing NO by E2 in the isthmus may contribute to

decreasing the motility of spermatozoa and promoting their binding to epithelial cilia until

they become hyperactivated. On the other hand, prostaglandin (PG) E2 and PGF2α derived

from follicular fluid directly stimulate NO production by ampullary oviductal epithelial cells

immediately after ovulation via E-prostanoid receptor type 2 (PTGER2) and F-prostanoid

receptor (PTGFR), respectively.. Because NO contributes to survival of oocyte and embryos

(Manser et al. 2004, Goud et al. 2005), PGs may play roles in maintaining the optimal

concentrations of NO in the ampulla for survival of oocyte and embryos.

Page 24: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

22

SUMMARY Nitric oxide (NO) is a regulator of sperm motility, oocyte/embryo survival and waves of

contraction/relaxation in mammalian oviducts. Since follicles control oviductal functions by

two routes at least, 1) a systemic way via blood vessels before ovulation, 2) a direct way by

entering of follicular fluid through fimbria at ovulation, we hypothesized that NO synthesis in

the bovine oviduct is regulated by follicular steroids and prostaglandins (PGs). Quantification

of mRNA expressions in the ampullary tissues showed that inducible NO synthase (iNOS)

mRNA expression was highest at the day of ovulation (Day 0). In contrast, iNOS mRNA

expression in the isthmus was highest at Days 5-6 and lowest at Days 19-21. Endothelial NOS

(eNOS) mRNA expressions in either the ampulla or the isthmus did not change during the

estrous cycle. PGE2 and PGF2α increased iNOS mRNA expressions in cultured ampullary

oviductal epithelial cells after 1 h incubation. These increases were suppressed by an

antagonist of E-prostanoid receptor type 2, one of the PGE2 receptor. Estradiol-17β decreased

the expression of iNOS mRNA expression in cultured isthmic epithelial cells 24 h after

treatment. This effect was suppressed by an antagonist of estrogen receptor α (ESR1).

Expression of ESR1 was highest at Days 19-21 in the isthmic tissues. The overall findings

indicate region-specific difference of NO synthesis in the oviduct: PGs, which are contained

in follicular fluid, may up-regulate NO synthesis in the oviductal epithelium, while circulating

E2 seems to inhibit NO synthesis via ESR1 in the isthmus at the follicular stage.

Page 25: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

23

CAHPTER 3 Roles of EDNs in regulating oviductal NO synthesis

and smooth muscle motility in cows

INTRODUCTION

Mammalian oviducts play crucial roles in the first days of pregnancy (Ulbrich et al. 2010).

Fertilized eggs are transported by two key oviductal actions, ciliary beating and smooth

muscle motility (Hunter 2012). Distal parts of the oviduct including the infundibulum and

ampulla have well developed mucosal folds surrounded by abundant ciliated cells (Noreikat

et al. 2012). Ciliary activity is more important for the transport of eggs in the distal part of the

oviduct which has a thin layer of smooth muscle. Indeed, inhibition of smooth muscle activity

did not disturb the transport of eggs through the ampullary region (Halbert et al. 1976). By

contrast, the isthmus has a few ciliated cells and thick smooth muscle layer, producing waves

of contraction and relaxation that contribute to embryo transport. The amplitude, frequency

and tonus of the waves are regulated by various hormones and factors including nitric oxide

(NO) and endothelins (EDNs) (Rosselli et al. 1994, Priyadarsana et al. 2004).

Endothelins are peptide hormones composed of 21 amino acids. Endothelins have

three isoforms, EDN1, EDN2 and EDN3 (Jeoung et al. 2010). Endothelin-1 was first

identified as a vasoactive factor in porcine aortic smooth muscle (Yanagisawa et al. 1988).

Receptors of EDNs have two types, type A (EDNRA) and type B (EDNRB). Endothelin-1 and

EDN2 bind to both EDNRA and EDNRB, while EDN3 only binds to EDNRB (Bridges et al.

2011). In various tissues, EDNRA is involved in signaling toward contraction of smooth

muscle, so that only EDN1 and EDN2 promote smooth muscle motility (Rosselli et al. 1994,

Sakamoto et al. 1999, Al-Alem et al. 2007). On the other hand, EDNRB signaling induces the

synthesis of nitric oxide (NO), a vasodilation factor (Hirata et al. 1993), and so all the EDN

isoforms induce the relaxation of smooth muscle via EDNRB. Nitric oxide has also other

functions in physiological events in the oviduct including embryonic development (Manser et

Page 26: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

24

al. 2004), epithelial secretion (Siemieniuch et al. 2009, Yilmaz et al. 2012) and ciliary beating

which are essential for pregnancy (Chiu et al. 2010).

Recently, we demonstrated that all the EDN isoforms are expressed in the epithelia

of both the ampulla and the isthmus of the bovine oviduct (Yamamoto et al. 2014). Thus,

EDNs may regulate oviductal smooth muscle motility in a paracrine way, and may affect

synthesis of NO in both autocrine and paracrine manners. In the present study, we

hypothesized that EDNs produced by epithelial cells play several roles in the regulation of

smooth muscle motility and/or NO synthesis in the oviduct. To test this hypothesis, we

investigated 1) the localizations of EDNRA and EDNRB in the ampulla and isthmus, 2) the

effects of EDNs on NO synthesis in cultured oviductal epithelial cells isolated from the two

regions, and 3) the effects of EDNs on smooth muscle motility in oviductal tissues.

RESULTS

Expressions of EDN receptors

In the ampulla, EDNRB mRNA expression was higher after ovulation than before ovulation (P<0.05,

Fig. 8A), while EDNRA mRNA expression did not significantly change around ovulation (P>0.05, Fig.

8A). In the isthmus, mRNA expressions of EDNRA was higher after ovulation than before ovulation

(P<0.05), while that of EDNRB did not change around ovulation (P>0.05, Fig. 8A). Protein of EDNRA

was distributed in most of the epithelial cells and some of the smooth muscle cells, while EDNRB

protein was localized only in the epithelial cells in both the ampulla and isthmus (Fig. 8B).

Effects of EDNs on iNOS expressions

Incubating ampullary epithelial cells with EDN1 increased inducible NOS (iNOS) mRNA expression

after 1 h (P<0.05, Fig. 9A) but not after 4 h (P>0.05, Fig. 9A) and increased iNOS protein expression

after 1 h (P<0.05, Fig. 9B). Neither EDN2 nor EDN3 affected iNOS expression in the ampullary cells

(P>0.05, Table 2). iNOS expression in isthmic epithelial cells was not affected by EDN1, EDN2 or

EDN3 (P>0.05, Fig. 9 and table 2).

Page 27: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

25

Effects of EDNs on oviductal smooth muscle motility

Weak spontaneous waves of contraction and relaxation were observed in the ampullary region (Fig.

10A). In that region, EDN1 increased tonus (average values of top and bottom of the waves, P<0.05),

amplitude (P<0.05) and frequency (P<0.01) of smooth muscle motility (Fig. 10C-E). By contrast, the

isthmic region showed strong spontaneous waves (Fig. 10B). Endothelin 1 increased the tonus of

spontaneous waves in the isthmus (P<0.01, Fig. 10B and C). Neither ampullary nor isthmic smooth

muscle motility was significantly affected by EDN2 and EDN3 (Fig. 10C-E, P>0.05).

Page 28: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

26

Figure 8 A) Changes of endothelin receptor A (EDNRA) and EDNRB mRNA expressions in tissues

collected from the ampulla (black bar) and the isthmus (white bar) of the oviduct around ovulation

(mean ± SEM, n=6, before ovulation; n=12, after ovulation). Significant differences were indicated by

* (P<0.05) and ** (P<0.01). B) Distributions of EDNRA and EDNRB proteins in ampullary and

isthmic tissues obtained from cows after ovulation (L: lumen, EP: epithelium, SM: smooth muscle).

Red (Alexa 568) indicates each target protein (EDNRA or EDNRB), and Blue (DAPI) indicates nuclei

of the cells. All the scale bars indicate 50 μm.

Page 29: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

27

Figure 9 Effects of endothelin-1 on inducible nitric oxide synthase (iNOS) mRNA (A, n=5 oviducts)

and protein (B, n=4 oviducts) expressions in cultured oviductal epithelial cells isolated from the

ampulla and isthmus of the oviduct (mean ± SEM). Asterisks indicate significant differences (P<0.05,

compared to the groups without EDN1).

Page 30: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

28

Figure 10 Effects of endothelins (EDNs) on spontaneous waves of contraction and relaxation in the

ampulla (A) and isthmus (B) obtained from cows after ovulation. Horizontal long lines indicate tonus

of the waves at the start of the test. A horizontal and vertical scale bars indicates 2 min and 10 mN,

respectively. Effects of EDN1 on tonus (C), amplitude (D) and frequency (E) of contractile motility in

the ampulla (solid lines, n=5) and isthmus (dashed lines, n=6). Significant differences were indicated

by * (P<0.05) and ** (P<0.01) as compared to the groups without EDN1.

Page 31: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

29

Incubation period

1 h 4 h

Region Treatment nmol/L Fold changes

Ampulla EDN2 0 1.00 ± 0.42 1.00 ± 0.39

0.01 1.18 ± 0.56 0.82 ± 0.14

0.1 1.43 ± 0.67 0.56 ± 0.18

1 0.83 ± 0.22 0.73 ± 0.29

EDN3 0 1.00 ± 0.68 1.00 ± 0.31

0.01 0.84 ± 0.61 0.67 ± 0.21

0.1 0.79 ± 0.47 0.76 ± 0.25

1 1.33 ± 0.88 0.74 ± 0.31

Isthmus EDN2 0 1.00 ± 0.47 1.00 ± 0.20

0.01 0.86 ± 0.48 1.18 ± 0.32

0.1 0.75 ± 0.45 0.92 ± 0.33

1 1.17 ± 0.83 0.97 ± 0.24

EDN3 0 1.00 ± 0.15 1.00 ± 0.22

0.01 1.22 ± 0.19 0.99 ± 0.16

0.1 0.96 ± 0.06 0.95 ± 0.30

1 1.02 ± 0.48 1.03 ± 0.21

Table 2 Effects of endothelin (EDN) 2 and EDN3 on inducible nitric oxide synthase (iNOS) mRNA

expression in cultured epithelial cells obtained from the ampulla and isthmus of bovine oviducts (n=5,

mean ± SEM). There were no significant differences (P>0.05).

Page 32: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

30

Region Treatment (nmol/L) ΔTonus (μN) Amplitude (%) Frequency

(mHz)

Ampulla EDN2 0 0.00 ± 0.00 100.0 ± 0.00 66.8 ± 6.25

10-10 84.1 ± 149.08 95.6 ± 6.65 64.1 ± 5.03

10-9 74.7 ± 130.05 116.3 ± 13.57 66.4 ± 7.19

10-8 126.7 ± 162.90 131.6 ± 11.72 71.3 ± 7.05

EDN3 0 0.00 ± 0.00 100.0 ± 0.00 55.3 ± 3.26

10-10 -151.0 ± 66.16 104.2 ± 7.17 62.6 ± 2.33

10-9 -57.6 ± 89.24 118.9 ± 7.43 60.9 ± 4.32

10-8 -143.7 ± 108.18 117.9 ± 11.05 59.8 ± 5.00

Isthmus EDN2 0 0.00 ± 0.00 100.0 ± 0.00 91.0 ± 6.10

10-10 -20.8 ± 63.42 98.2 ± 2.43 94.4 ± 5.54

10-9 35.0 ± 79.81 102.7 ± 2.26 93.7 ± 5.88

10-8 248.22 ± 150.22 103.6 ± 3.42 94.4 ± 6.60

EDN3 0 0.00 ± 0.00 100.0 ± 0.00 95.2 ± 7.60

10-10 -28.0 ± 87.50 100.4 ± 1.67 94.0 ± 7.66

10-9 171.97 ± 88.52 100.0 ± 1.61 93.3 ± 5.27

10-8 140.47 ± 77.54 102.7 ± 1.52 93.7 ± 5.14

Table 3 Effects of endothelin (EDN) 2 and EDN3 on tonus, amplitude and frequency of contractile

motility (mean ± SEM) in the ampulla (n=5) and isthmus (n=6). There were no significant differences

(P>0.05).

Page 33: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

31

DISCUSSION

The preceding results show that EDN1 affect at least two oviductal functions: NO synthesis

(Fig. 9) and spontaneous waves of contraction and relaxation of oviductal smooth muscle (Fig.

10). These effects are shown schematically in Fig. 11. Spontaneous waves of smooth muscle

are responsible for the transport of the embryo through the isthmus (Hunter 2012). Nitric oxide

promotes ciliary beating (Chiu et al. 2010) and embryo development (Manser et al. 2004).

The structures of the oviductal regions are dramatically different. Numerous motile cilia

surround the luminal epithelium in the distal parts of the oviduct including the infundibulum

and ampulla. Beating of these cilia produces a stream of oviductal fluid, transporting the

ovulated oocyte and the embryo toward the site of fertilization and uterus, respectively (Kölle

et al. 2009). By contrast, in the isthmus, a few mucosal folds are surrounded by thick smooth

muscle layer which produces waves of contraction and relaxation. These spontaneous waves

of smooth muscle allow the early embryo to pass through the narrow isthmic cavity. The

present results suggest that the specific functions of the ampulla and isthmus are regulated by

EDNs.

Two-types of receptors for EDNs showed different distribution in the present study,

i.e., smooth muscle cells expressed EDNRA but not EDNRB (Fig. 8). Binding of EDNs to

EDNRA has been demonstrated to immediately activate phospholipase C, generate inositol

triphosphate, mobilize extracellular Ca2+ and induce contraction in vascular smooth muscle

cells (Pollock et al. 1995, Vignon-Zellweger et al. 2012). Since EDNRA has a low affinity for

EDN3 (Yanagisawa 1994), only EDN1 and EDN2 seems to bind to EDNRA on smooth muscle

cells of the oviduct. This is supported by the finding which EDN1 but not EDN3, stimulated

the activity of smooth muscle (Fig. 10 and Table 2). Since EDN1 is synthesized in epithelial

cells of the oviduct (Yamamoto et al. 2014), these EDNs are suggested to be released from

epithelial cells, to bind EDNRA expressed on smooth muscle cells, to activate motility of

smooth muscle and to contribute to the transport of the embryo toward the uterus.

Since the smooth muscle layer of the ampullary region is not well developed, ciliary

Page 34: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

32

activity in the ampullary epithelium is required for transport of the oocyte/embryo. In fact,

inhibition of ciliary beating suppresses egg transport (Halbert et al. 1976). The ampullary

lumen is surrounded by numerous cilia whose motility produces the stream toward the uterus

(Kölle et al. 2009). Ciliary beating is regulated by several factors including NO (Chiu et al.

2010). Since EDN1 stimulated iNOS mRNA/protein expression in ampullary epithelial cells

in the present study (Fig. 9), EDN1 may participate in regulation of ciliary activity.

Immediately after ovulation, the ovulated oocyte with follicular fluid containing EDN1 is

suggested to enter the oviduct and to be transported to the site of fertilization within a few

hours (Acosta et al. 1998). EDN1 derived from not only oviductal epithelium but ruptured

follicle seems to facilitate ciliary motility for oocyte transport by stimulating NO synthesis.

The binding of EDN1 to EDNRB increases endothelial NOS (eNOS) derived NO in

vascular endothelial cells (Hirata et al. 1993, Tsukahara et al. 1994). This eNOS derived NO

relax vascular smooth muscle. On the other hand, a specific antagonist of EDNRB (BQ-788)

suppresses iNOS expression/NO production which are stimulated by EDN1 via activation of

nuclear factor κB (NF-κB) in rat brain astrocytes (Wang et al. 2011). Activation of NF-κB

signaling induces iNOS expressions in some cell types in the brain (Harris et al. 2009, Pérez-

Rodríguez et al. 2009). In the oviduct, several factors are known to activate NF-κB signaling

(Gabler et al. 2008, Shaw et al. 2011). Our previous findings that iNOS mRNA expression is

highest on the day of ovulation in the ampullary region during the estrous cycle (Kobayashi

et al. 2016) could be evidence that EDNRB mediates iNOS expression/NO production via

NF-κB in the oviduct. Other isoforms of NOS, eNOS and neuronal NOS (nNOS), are also

expressed in the bovine oviduct (Lapointe et al. 2006, Yilmaz et al. 2012). Yilmaz et al.

describes that eNOS and nNOS are possible to participate in secretions from epithelial cells

because eNOS and nNOS are also distributed in epithelail layer of the oviduct. Therefore,

these isoforms regulated by EDNs might affect oviductal functions. Further studies are

required to determine the regulatory mechanism of NO synthesis by EDN system in the

oviduct.

An antagonist of both types of EDNs (tezosentan) decreases early embryonic

Page 35: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

33

development in mice (Jeoung et al. 2010). Transforming growth factor β (TGFβ) family

regulated by EDNs may be involved in embryonic development according to a microarray

analysis of tezosentan-treated mice (Jeoung et al. 2010). In fact, TGFβ-SMAD signaling is

required for early embryonic development in cattle (Zhang et al. 2015), and TGFβ type I, II

and III are expressed in human oviductal epithelium (Zhao et al. 1994). We also propose that

ET-1-EDNRB signaling via iNOS-derived NO affects embryonic development because NO

promotes early embryonic development (Manser et al. 2004). In the oviductal cavity, various

factors derived from oviductal cells, follicular fluid and the circulation participate in

fertilization and embryonic development. Mechanisms of formation of oviductal milieu to

make sure successful pregnancy are expected to be clarified by further studies.

In conclusion, the present results indicate the EDN1 have specific effects on 1) NO

synthesis in the ampulla and 2) smooth muscle motility in the isthmus. These roles of EDN1

could be involved in region-specific physiological events, ciliary beating for oocyte transport

and fertilization in the ampulla, and waves of contraction and relaxation of oviductal smooth

muscle that are needed to transport the early embryo to the uterus.

Page 36: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

34

Figure 11. Region-specific roles of endothelins (EDNs) in the ampulla and isthmus of the oviduct. In

the ampulla, EDN1 stimulates iNOS expression and NO synthesis via EDNRB which is highly

expressed in the epithelial cells at the day of ovulation. Since NO contributes to survival of oocyte and

embryos (Manser et al. 2004) and ciliary beating (Chiu et al. 2010), EDN1 may promote early

embryonic development and ciliary beating in the ampullary region. By contrast, EDN1 promote the

contractility of isthmic smooth muscle via EDNRA expressed on smooth muscle cell surface,

participating in the complicated regulatory mechanism of smooth muscle motility regulated by various

factors and contributing to successful embryo transport toward the uterus.

Page 37: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

35

SUMMARY

Endothelins (EDNs) participate in various physiological events including smooth muscle

contraction, nitric oxide (NO) synthesis and embryonic development. Here, we investigated

regional roles of EDNs produced by bovine oviductal epithelial cells in NO synthesis and

smooth muscle motility. Quantification of mRNA expressions indicated that expression of

EDN receptor B (EDNRB) was higher after ovulation than before ovulation in the ampullary

region, while EDNRA expression in the isthmic region was higher after ovulation than before

ovulation. Immunohistochemistry revealed that EDN receptors (EDNRA and EDNRB) were

expressed in the epithelium, whereas smooth muscle showed positive staining only for

EDNRA. The expressions of iNOS mRNA and protein in cultured epithelial cells isolated

from the ampulla were stimulated by EDN1, but not by EDN2 or EDN3 after 1 h incubation.

In isthmic epithelial cells, none of the EDNs affected iNOS expression. Isometric contraction

tests indicated that spontaneous waves were strong in the isthmus but weak in the ampulla.

EDN1 modulated smooth muscle motility in both regions. The overall findings suggest that

EDN1 play region-specific roles in smooth muscle motility and epithelial NO synthesis,

providing an optimal oviductal micro-environment for transports of gametes, fertilization and

development/transport of the early embryo.

Page 38: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

36

CHAPTER 4 Summer heat stress affects prostaglandin synthesis

in the bovine oviduct

INTRODUCTION

Summer heat stress (HS) weakens estrous behavior, suppresses follicular development and

causes early embryo mortality, all of which decrease the fertility rate in cows (Gwazdauskas

et al. 1975, Cavestany et al. 1985, De Rensis & Scaramuzzi 2003, Sakatani et al. 2004, Hansen

2009). HS also increases the production of PGF2α in the endometrium, which might result in

the early regression of CL or the death of embryos (Putney et al. 1988, Malayer et al. 1990).

HS is of special concern to the livestock industry because of recent global warming.

Heat shock proteins (HSPs) are highly expressed under hot conditions, and protect

cells from various stresses including heat and reactive oxidants (Kregel 2002). Especially,

HSP90 is known to activate cPGES which increases the production of PGE2 in rat fibroblasts

(Tanioka et al. 2003). Thus, HS may stimulate PGE2 productions from the oviductal

epithelium, which would reduce gamete/embryo transport.

In mammals, the oviduct plays roles in the transport of gametes and fertilized oocytes

as well as the development of embryos (Menezo & Guerin 1997, Suarez 2008, Ulbrich et al.

2010). After fertilization occurs in the ampulla of the oviduct, the embryo is transported to the

uterus within several days. The transport to the uterus is caused by waves of contraction and

relaxation of the smooth muscle and the ciliation of oviductal epithelial cells (Halbert et al.

1976, Hunter 2012). Prostaglandin (PG) E2 and PGF2α concentrations in the ipsilateral oviduct

to the corpus luteum (CL) or the dominant follicle are highest in the periovulatory phase

(Wijayagunawardane et al. 1998). These PGs seem to control gamete/embryo transport by

stimulating smooth muscle motility (Wijayagunawardane et al. 2001a).

PG biosynthesis starts from the liberation of arachidonic acid from phospholipids

(Okuda et al. 2002). Cyclooxygenases convert arachidonic acid to PGH2 which is the

precursor for various PGs including PGE2 and PGF2α (Garavito et al. 2002, Gabler et al. 2008).

The conversions of PGH2 to PGE2 or PGF2α are catalyzed by the specific downstream enzymes,

PGE synthase (PGES) and PGF synthase (PGFS), respectively (Arosh et al. 2002). The bovine

oviduct expresses three PGES isozymes, microsomal PGES1 (mPGES1), mPGES2 and

cytosolic PGES (cPGES) (Gauvreau et al. 2010).

Page 39: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

37

We hypothesized that HS negatively affects prostaglandin synthesis in the bovine

oviduct. In the present study, to prove the above hypothesis, we investigated the effects of

elevated temperatures on the synthesis of PGE2 and PGF2α in cultured bovine oviductal

epithelial cells. We also studied the expressions of PGESs and HSP90 in bovine oviducts

obtained in winter and summer.

RESULTS

Comparison of mRNA expression of PG synthases and HSP90 between summer and winter

in bovine oviducal tissues

The expression of HSP90 mRNA in the bovine ampullary oviduct was higher in summer than

in winter (Fig. 12A). On the other hand, HSP90 mRNA expression in the isthmic tissue was

not significantly different in summer and winter (Fig. 12B).

The expression of cPGES mRNA in the bovine ampullary oviduct was higher in

summer than in winter (Fig. 13A). In contrast, the mRNA expression of carbonyl reductase 1

(CBR1) was lower in summer than in winter (Fig. 13E) the mRNA expressions of mPGES1,

mPGES2 and PGFS in the ampulla were not significantly different between in summer and

winter (Fig. 13B-D).

Effect of elevated temperature on HSP90 mRNA and protein expressions in cultured bovine

oviductal epithelial cells.

The expression of HSP90 mRNA was higher at 40.5°C than at 38.5°C in the cultured

ampullary oviductal epithelial cells (Fig. 14A). HSP90 protein expression was also higher at

40.5°C than at 38.5°C in the cultured ampullary oviductal epithelium (Fig. 14B).

Effects of elevated temperatures on prostaglandin productions in cultured bovine oviductal

epithelial cells

The level of PGE2 was significantly higher at 40.5°C than that at 38.5°C (Fig. 15A). The

elevated temperature had no effect on PGF2α production in the ampullary cells (Fig. 15B).

Page 40: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

38

Figure 12 Expression of HSP90 mRNA in the bovine ampullary (A) and isthmic (B) oviduct

(mean ± SEM) in post-ovulatory phase collected in winter (November 21-March 1, 2011-2012,

n=14) and summer (July 23-September 6, 2012, n=14). The asterisk indicates a significant

difference (P<0.05).

Page 41: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

39

Figure 13 Expression of mPGES1, mPGES2, cPGES, PGFS, and CBR1 mRNA in the bovine

ampullary oviduct (mean ± SEM) in post-ovulatory phase collected in winter (November 21-

March 1, 2011-2012, n=14) and summer (July 23-September 6, 2012, n=14). Asterisks

indicate significant difference (P<0.05).

Page 42: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

40

Figure 14 Effects of elevated temperature (40.5°C) on the expression of heat shock protein

(HSP) 90 mRNA (A, n=7 oviduct) and protein (B, n=4 oviduct) in the epithelial cells collected

from the ampulla of the bovine oviduct (mean ± SEM). Asterisks indicate significant

difference (P<0.05).

Page 43: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

41

Figure 15 Effects of temperature (38.5°C and 40.5°C) on the productions of prostaglandin

(PG) E2 (A) and PGF2α (B) in epithelial cells collected from the ampulla of the bovine oviduct

(mean ± SEM, n=4 oviducts). Asterisks indicate significant difference (P<0.05).

Page 44: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

42

DISCUSSION

In the present study, elevated temperature promoted PGE2 production but did not affect PGF2α

production in cultured ampullary oviductal epithelial cells (Fig. 15). The body temperature of

cows is approximately 38.5°C, but it increases under hot conditions. For example, the vaginal

temperature of cows was found to increase to approximately 40.5°C under high temperature

conditions (Nabenishi et al. 2011). Oviductal smooth muscle is relaxed by PGE2 and induced

to contract by PGF2α (Al-Alem et al. 2007, Siemieniuch et al. 2009). Since gametes and

embryos are transported by waves of contraction and relaxation, upsetting the balance between

PGE2 and PGF2α by HS may negatively affect the transport.

Elevated temperature increased PGE2 production in the ampullary epithelial cells (Fig.

15). The increased PGE2 production could be due to either increases in the expressions of PGE

synthases (PGESs) or increases in the PGES activity. Mammals have three PGES isozymes,

mPGES1, mPGES2 and cPGES (Murakami et al. 2002). In the present study, the expressions

of mPGES1 and cPGES mRNA were higher at 40.5°C than at 38.5°C in the ampullary

epithelial cells. However, only cPGES mRNA was higher in summer than in winter in the

ampullary oviductal tissue (Fig. 13). Thus, cPGES may play an important role to stimulate

PGE2 synthesis in the ampullary oviduct under HS. Since PGE2 is known to relax the oviductal

smooth muscle (Al-Alem et al. 2007, Siemieniuch et al. 2009), HS may be a cause of summer

infertility by decreasing the oviductal motility via promoting PGE2 production. Since the

mPGES1 mRNA expressions in both oviductal sections did not show any difference between

in summer and winter in contrast to the in vitro experiment. This discrepancy may be due to

the different conditions between in vivo and in vitro. However, since cPGES expression was

increased by heat stress both in vivo and in vitro, we believe that the summer heat stress

increases PGE2 production in bovine oviducts.

HSP90 expressions in the oviduct were higher in summer than in winter (Fig. 12) and

were increased by elevated culture temperature (Fig. 14). HSPs protect cells from various

stresses including heat and reactive oxidants (Kregel 2002). HSPs are highly expressed at high

temperature, and help the folding of proteins to prevent protein denaturation (Kregel 2002).

Especially, HSP90 plays crucial roles as a molecular chaperone for the activation of various

client proteins including cPGES (Pearl & Prodromou 2006). In rat fibroblast cells, HSP90

activates cPGES and consequently promotes PGE2 production (Tanioka et al. 2003). Therefore,

it is possible that HS increases HSP90 expression and the activity of cPGES, resulting in

Page 45: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

43

increasing PGE2 production in bovine oviductal epithelial cells. Furthermore, the HSP90

expression in summer could be assumed to increase not only in the oviduct but also in other

organs. Since HSP90 is an important chaperone for the actions of various client proteins (Pearl

& Prodromou 2006), HS may affect not only oviductal functions but also other body

conditions. HSP90 and cPGES mRNA expressions in the ampullary oviduct was higher in

summer than in winter, whereas there was no difference in the isthmic oviduct between in

summer and winter. It has been reported that expressions of HSPs were higher in porcine

oocytes in summer than in winter, although there was no seasonal changes in porcine cumulus

cell (Pennarossa et al. 2012). Therefore, it is possible that HSP90 and cPGES expressions in

the isthmic oviduct are not affected by seasonal different conditions in contrast to the

ampullary oviduct.

The overall findings suggest that heat stress upsets the balance between PGE2 and

PGF2α secretions, which would be expected to decrease the motility of the oviductal smooth

muscle, which is important for gamete/embryo transport. This could lead to the low fertility

in cattle in summer (Fig. 16).

Page 46: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

44

Figure 16 Effects of summer heat stress (HS) on the oviductal motility. Only relaxant factors,

i.e., prostaglandin E2 and nitric oxide in the oviductal epithelium, are stimulated by HS,

upsetting the waves of contraction and relaxation of oviductal smooth muscle, negatively

affect the transport of gametes and embryos and inducing low fertility in cattle.

Page 47: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

45

SUMMARY

Summer heat stress (HS) negatively affects reproductive functions including prostaglandin

(PG) F2α secretion in the endometrium, and decreases fertility in cattle. Here, we examined

the difference of expressions of heat shock protein 90 (HSP90), PG synthases and inducible

nitric oxide (NO) synthase (iNOS) in bovine oviductal tissues obtained in summer and winter,

and effects of the elevated temperature on PG and NO syntheses in oviductal epithelial cells.

The expression of HSP90, cytosolic PGES (cPGES) and iNOS mRNA was higher in the

ampullary tissues in summer than in winter. Carbonyl reductase 1 (CBR1) mRNA expression

was lower in summer than in winter. The epithelial cells obtained from the ampulla of the

oviduct were incubated at 38.5°C and 40.5°C for 24h. The concentrations of PGE2 and NO

metabolites and HSP90 mRNA and protein expressions were higher at 40.5°C than at 38.5°C,

while PGF2α production was not affected the elevated temperature in cultured ampullary

epithelial cells. In summary, hot conditions stimulated the productions of PGE2 and NO which

relaxes oviductal smooth muscle, suggesting that HS upsets the PG and NO secretions and

reduces oviductal smooth muscle motility, which in turn could suppress gamete/embryo

transport through the oviduct and could induce the low fertility in summer.

Page 48: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

46

CHAPTER 5 CONCLUSIONS

Nitric oxide is a multi-role molecule in the oviduct, e.g., NO stimulates activity of

spermatozoa (Miraglia et al. 2011), contributes to oocyte/embryo survival (Manser et al. 2004,

Goud et al. 2005) and relaxes the smooth muscle of the oviduct (Rosselli et al. 1994). The

present study indicates differences in the regulatory mechanisms of NO synthesis between the

ampulla and isthmus of the bovine oviduct. Molecules derived from follicular fluid entering

the oviduct seem to regulate NO synthesis in the ampulla immediately after ovulation. In

contrast, E2 derived from the dominant follicle could suppress NO synthesis at the pre-

ovulatory stage. These differences are suggested to contribute to the transient roles of the

oviduct in various physiological events including transports of gametes and transport and

embryonic development. In addition to the regulation by ovaries, oviductal secretions were

indicated to be regulated by molecules which are secreted by the oviduct itself. These multiple

ways of regulating oviductal secretions may provide optimal micro-environment for the events

in the first days of pregnancy.

During the past several decades, many scientists tried to clarify the cause of summer

low fertility in cattle. In female reproductive tracts, hot conditions suppress E2 concentration

in the dominant follicle (Wolfenson et al. 1997, Bridges et al. 2005) and increase the secretion

of PGF2α from the endometrium (Putney et al. 1988, Malayer et al. 1990). The present study

indicates that summer heat stress affects the mRNA expressions of PG synthases and NO

synthase in the oviduct. In vitro study also suggests that the elevated temperature stimulates

the secretions of PGE2 and NO which are relaxant factor of oviductal smooth muscle. Since

the waves of contraction and relaxation are intricately regulated various molecules, the

stimulus of the relaxant factors may upset the waves produced by oviductal smooth muscle,

resulting in failure of oocyte/embryo transport and the low fertility in summer.

Page 49: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

47

MATERIALS AND METHODS

Collection of bovine oviducts

Oviducts of Holstein cows were collected at a local abattoir within 10-20 min after the

exsanguination. The stages of the estrous cycle were determined based on a macroscopic

observation of the ovary and the uterus (Okuda et al. 1988, Miyamoto et al. 2000). After

trimming of the oviducts being ipsilateral to the corpus luteum, the ampullary and the isthmic

sections were immediately frozen and stored at -80°C until mRNA and protein extraction.

Oviductal tissues for immunohistochemistry were fixed in PBS with 10% (v/v) neutral

formaldehyde for 24 h, then embedded in paraffin. For cell culture, the oviducts were

submerged in ice-cold saline and transported to the laboratory.

Oviductal tissues (Days 0-6 after ovulation) used for the determination of PG

synthases and HSP90 mRNA expression were collected from 14 cows in winter (November-

March, 2011-2012) and 14 cows in summer (July-September, 2012). The cows were

slaughtered at a local abattoir in Okayama, Japan. Average temperatures in Okayama in

sampling periods are 5.3°C in winter, and 27.5°C in summer. The range of temperatures during

the summer sampling period was 22.1°C -36.8°C. Although its average temperature was 27°C

as written in the original MS, there were 33 Days (of 46 Days) that the daily maximum

temperature was higher than 33°C. This climate seems to be under an apparent heat stress

condition for dairy cows. The temperature data were obtained from Japan Meteorological

Agency.

Determination of PG concentrations in collected follicular fluid

A Japanese black cow and an Angus cow were utilized for superovulation treatment and

collecting follicular fluids. A total of 20 Arrmour units (AU) of follicle-stimulating hormone

(FSH; Antorin R-10, Kyoritsu Seiyaku Corporation, Kanagawa, Japan) was administered

twice daily in decreasing doses during 3 days (5, 5, 3, 3, 2 and 2 AU, respectively). A PGF2α

analogue (cloprostenol, Resipron-C; ASKA Pharmaceutical Co., Tokyo, Japan, 0.5 mg) was

administered on the third day of FSH administration. Cows had a controlled internal drug

release device (CIDR) (CIDR1900; Pfizer Japan Inc., Tokyo, Japan) inserted for 8 to 9 days

until the injection of the PGF2α analogue and were received estradiol benzoate (Kyoritsu

Seiyaku Corporation, 1 mg) 4 days before the first FSH administration. These cows were

injected with a GnRH analogue (fertirelin acetate; Conceral, Nagase Pharmaceutical, Tokyo,

Page 50: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

48

Japan, 100 µg) 24 h after CIDR removal. Transvaginal collection of follicular fluids was

carried out at 26 to 27 h after GnRH administration. Follicular aspiration was conducted using

an ultrasound scanner (SSD-900, ALOKA, Tokyo, Japan) and 7.5 MHz convex array

transducer (UST-9106P-7.5, ALOKA) attached with a 17-gauge stainless steel needle guide.

Follicles over 8 mm in diameter were aspirated by an aspirator (FV4; FHK, Tokyo, Japan)

equipped with a disposable aspiration needle (Misawa Medical Industry Co., Ltd. Ibaraki,

Japan). Collected follicular fluids were centrifuged (1,800 × g, 10 min, 4°C) and supernatants

were stored at -20°C until measuring concentrations of PGs. All procedures with animal

subjects had been reviewed and approved by the Animal Care Committee of the Animal

Research Center, Hokkaido Research Organization.

The concentrations of PGE2 in the collected follicular fluids were determined by

enzyme immunoassay as described previously (Tanikawa et al. 2005). The PGE2 standard

curve ranged from 0.039 to 10 ng/mL, and the ED50 of the assay was 0.625 ng/mL. The intra-

assay coefficients of variation were on average 2.6%. The range of the concentrations of PGE2

in follicular fluids was 0.006-0.04 μmol/L.

The concentrations of PGF2α in the collected follicular fluids were determined by

enzyme immunoassay as described previously (Uenoyama et al. 1997) with our modification

using peroxidase-labeled PGF2α as a tracer (1:25000 final dilution) and anti-PGF2α serum

(1:100000 final dilution). The anti-PGF2α serum (OK-PGF) was produced by Sigma-Aldrich

(St. Louis, MO, USA). Cross-reactivities of the anti-PGF2α serum were validated by

comparing the inhibition of binding of peroxidase-labeled PGF2α to antiserum, were as

follows: PGF2α, 100%; PGA2, 0.38%; PGD2, 2.51%; PGE1, 0.53%; PGE2, 0.08%; PGF1α,

5.62%; PGF2αβ, 0.49%; PGF2β, 0.58%. The PGF2α standard curve ranged from 0.016 to 4

ng/mL, and the ED50 of the assay was 0.25 ng/mL. The intra-assay coefficients of variation

were on average 3.9%. The range of the concentrations of PGF2α in follicular fluids was 0.06-

0.83 μmol/L. This assay system of PGF2α has no cross-reaction with cloprostenol, a PGF2α

analogue. In the present study, cloprostenol was utilized for luteolysis 50-51 h before

collection of follicular fluid. The half-life of cloprostenol was approximately 3 h and 14C-

labeled cloprostenol was not detected in ovary 48 h after dose of the PGF2α analogue (Reeves

1978). Thus, we considered the concentration of PGF2α which we detected in follicular fluid

did not contain exogenous PGF2α.

Isolation of oviductal cells

Page 51: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

49

Oviductal tissues collected at Days 0-3 after ovulation were utilized for cell culture. The

oviduct was separated into infundibulum, ampulla, ampullary-ithmic junction, isthmus and

utero-tubal junction (Fig. 17). The epithelial cells were isolated from the ampullary and the

isthmic sections of the oviduct. A Teflon catheter (internal diameter was 0.5 mm) was inserted

into the ampullary and the isthmic sections of the oviduct, and the luminal wall was washed

five times with 1 mL of sterile Hank’s balanced salt solution (HBSS) containing 0.1% (wt/vol)

BSA (Roche, Manheim, Germany), 100 IU/mL penicillin (Meiji Seika Pharma, Tokyo, Japan)

and 100 μg/mL streptomycin (Meiji Seika Pharma). The oviduct was connected to a peristaltic

pomp (Gilson, Middleton, WI, USA), and perfused with 20 mL of sterile HBSS containing

0.25% (wt/vol) bovine trypsin (>7500 BAEE units/mg solid; Sigma-Aldrich, St. Louis, MO,

USA), 0.02% (wt/vol) EDTA2Na (Sigma-Aldrich), 0.1% (wt/vol) BSA, 100 IU/mL penicillin

and 100 μg/mL streptomycin (at flow rate of 10 mL/min, 38°C, for 30 min). After the perfusion,

the dissociated epithelial cells were filtered through metal meshes (150 μm and 77 μm) to

remove undissociated tissue fragments. The filtrates were washed by centrifugation (180 × g

for 10 min at 4°C) with Tris-buffered ammonium chloride (pH 7.5) to remove hemocytes, and

Dulbecco’s Modified Eagle’s Medium (DMEM; Sigma-Aldrich) supplemented with 0.1%

(wt/vol) BSA, 100 IU/mL penicillin and 100 μg/mL streptomycin, respectively. After the

washes, the cells were counted with a hemocytometer. Cell viability was higher than 95% as

assessed by 0.5% (wt/vol) trypan blue dye exclusion. The final pellets of the epithelial cells

were resuspended in DF (DMEM/Ham’s F-12; 1:1 (vol/vol) (Invitrogen, Carlsbad, CA, USA)

supplemented with 10% (vol/vol) bovine serum (Invitrogen), 20 mg/mL gentamicin

(Invitrogen) and 2 mg/mL amphotericin B (Sigma-Aldrich)). These cells were seeded at a

density of 1.0 × 105 viable cells/mL to the 25 cm2 culture flasks (Greiner Bio-One,

Frickenhausen, Germany) and cultured at 38.5°C in a humidified atmosphere of 5% CO2 in

air. Because the epithelial cells attached 24-48 h after plating, the medium in the epithelial

cell culture was replaced 48 h after plating. The medium was changed every 2 days until

confluency was reached.

To purify the epithelial cells, when the epithelial cells reached sub-confluence, the

cells were trypsinized. Briefly, the cells in the culture flask were washed with PBS (-) twice.

After the washing, 0.02% (wt/vol) porcine trypsin (1000-2000 BAEE units/mg solid; Sigma-

Aldrich) with 0.008% (wt/vol) EDTA (Sigma-Aldrich) in PBS was added to the flask and the

cells were incubated for 5 min at 38.5°C to detach stromal cells. Then, the solution containing

the stromal cells was removed, and the remaining cells were incubated with 2 mmol/L EDTA

Page 52: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

50

in PBS for 2 min at 38.5°C. After the incubation, the solution was removed and the cells were

washed with PBS (-), and the cells were incubated for 10 min at 38.5°C with 0.02% (wt/vol)

bovine trypsin in PBS. After the incubation, the cells were washed by centrifugation (180 × g

for 10 min at 4°C) with culture medium. Then, the cells were placed in fresh DF to adjust

them to a density of 1.0 × 105 viable cells/mL. These cells were seeded to 4-well plates

(Thermo Fisher Scientific, Waltham, MA, USA), 48-well plates (Greiner Bio-One) and 75

cm2 culture flasks (Greiner Bio-One) and cultured at 38.5°C in a humidified atmosphere of

5% CO2 in air. The medium was exchanged every 48 h until confluency was reached. When

the cells reached confluence (10-11 days after starting the culture), they were used for

experiments.

The homogeneity of the epithelial cells was evaluated using immunofluorescent

staining for specific marker of epithelial (cytokeratin) and stromal (vimentin) cells as

described previously (Malayer & Woods 1998, Tanikawa et al. 2008) with our modification.

Briefly, oviductal epithelial and endometrial stromal cells (control) were seeded on collagen

coated sterile cover glasses in 6-well plates at a density of 5.0 × 104 cells/mL. The endometrial

stromal cells were isolated as described previously (Tanikawa et al. 2008). 48 h after the

seeding, the cells were fixed by 4% (wt/vol) paraformaldehyde (Nacalai Tesque, Inc., Kyoto,

Japan) for 10 min. Then, the cells were incubated with 5% (wt/vol) skim milk (Nacalai Tesque,

Inc.) in PBS containing 0.1% (vol/vol) Tween 20 (PBS-T) for 60 min at room temperature.

After the blocking, the cells were incubated with specific primary antibodies to cytokeratin

(anti-cytokeratin-IgG-mouse; Sigma-Aldrich, 1:500 dilution) or vimentin (anti-vimentin-IgG-

mouse; Sigma-Aldrich, 1:5000 dilution) in 5% (wt/vol) skim milk overnight at 4°C. After the

incubation, the cells were incubated again with secondary antibody (anti-mouse-IgG Alexa

594 conjugate-donkey; Sigma-Aldrich; 1:500 dilution for cytokeratin 18, anti-mouse-IgG

FITC conjugate-donkey; Sigma-Aldrich; 1:500 dilution for vimentin, in PBS-T) for 60 min at

room temperature, and enclosed with ProLong Gold Antifade Reagent with DAPI (Invitrogen)

on slide glasses. Fluorescent was observed by fluorescence microscope (Olympus, Tokyo,

Japan). In the epithelial cells culture, contamination of the oviductal stromal cells was <1%

(Fig. 18).

Cell treatment for CHAPTER2

Oviductal epithelial cells reached confluence were incubated with estradiol-17β (E2, E8875,

Sigma-Aldrich, St. Louis, MO, USA; 0.1, 1, 10 nmol/L), progesterone (P4, P8783, Sigma-

Page 53: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

51

Aldrich; 1, 10, 100 nmol/L), prostaglandin E2 (PGE2, 14010, Cayman Chemical, Ann Arbor,

MI, USA; 0.01, 0.1, 1 μmol/L) and PGF2α (16010, Cayman Chemical; 0.01, 0.1, 1 μmol/L)

in phenol-red free DMEM/F-12 Ham (D2906, Sigma-Aldrich) supplemented with 500 μmol/L

ascorbic acid (013-12061, Wako Pure Chemical Industries, Osaka, Japan), 5 μg/mL holo-

transferrin (T4132, Sigma-Aldrich), 5 ng/mL sodium selenite (S5261, Sigma-Aldrich), 2

μg/mL insulin (I4011, Sigma-Aldrich), 0.1% (w/v) bovine serum albumin (A7888, Sigma-

Aldrich) and 20 mg/mL gentamicin (G1397, Sigma-Aldrich) for 1, 4 and 24 h at 38.5°C.

Selective antagonists of estrogen receptors (ESRs) were co-incubated with E2 (10 nmol/L),

respectively. MPP dihydrochloride (100 nmol/L; M7068, Sigma-Aldrich) for ESR1 and

PHTPP (100 nmol/L; 2662, R&D Systems, Minneapolis, MN, USA) for ESR2 were used as

selective antagonists. MPP dihydrochloride have been demonstrated selective antagonist for

ESR1 (Sun et al. 2002) and PHTPP is known to display 36-fold selectivity for ESR2 than

ESR1 (Compton et al. 2004). Antagonists of PTGER2 or PTGER4 were incubated in

combination with PGE2 (1 μmol/L) in cultured ampullary epithelial cells. AH6809 and

AH23848 are selective antagonists for PTGER2 and PTGER4, respectively (Coleman et al.

1994, Woodward et al. 1995). Both antagonists were pre-incubated with the cells for 1 h before

incubation with each ligand. After incubations, supernatants were collected for measuring NO

concentration and the cells were collected to measure DNA content for standardize the NO

concentration. Total RNA of the cells was extracted from another culture plate for

determination of mRNA expressions.

Cell treatment for CHAPTER3

Epithelial cells reached confluence on 4-well plates were used for this experiment. The cells

were incubated with serum-free DF (DMEM/Ham’s F-12 1:1 (vol/vol) supplemented with

0.1% BSA, 5 μg/mL holo-transferrin (Sigma-Aldrich) and 2 μg/mL insulin (Sigma-Aldrich))

at 38.5°C and 40.5°C for 24 h. After the incubation, the medium were collected in 1.5-mL

tube containing 1% stabilizer solution (0.3 mol/L EDTA, 1% (w/v) acid acetyl salicylic pH

7.3) and the concentrations of PGE2 and PGF2α in the medium were measured by enzyme

immunoassay. The cells were collected for measurement of the DNA amount by DNA assay.

Enzyme immunoassay

The concentrations of PGE2 and PGF2α in the culture medium were determined by enzyme

immunoassay as described previously (Uenoyama et al. 1997, Tanikawa et al. 2005). The

Page 54: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

52

PGE2 standard curve ranged from 0.039 to 10 ng/mL, and the ED50 of the assay was 0.625

ng/mL. The intra- and inter-assay coefficients of variation were on average 2.6% and 8.4%,

respectively. The PGF2α standard curve ranged from 0.016 to 4 ng/mL, and the ED50 of the

assay was 0.25 ng/mL. The intra- and inter-assay coefficients of variation were on average

3.9% and 17.7%, respectively. DNA content was measured by the spectrophotometric method

(Labarca & Paigen 1980), and used to standardize the results.

Total RNA extraction and quantitative RT-PCR

Total RNA was extracted from oviductal tissues and cells using TRIsure according to the

manufacturer’s directions. Using iScript RT Supermix for RT-qPCR (170-8841, Bio-Rad

Laboratories, Hercules, CA, USA), 1 µg of each total RNA was reverse transcribed.

Quantifications of mRNA expressions were determined by Quantitative RT-PCR using MyiQ

(Bio-Rad Laboratories) and SooAdvanced SYBR Green Supermix (1725261B10, Bio-Rad

Laboratories) starting with 4 ng of reverse-transcribed total RNA as described previously

(Sakumoto et al. 2006). All primers were designed to amplify specific products for iNOS

(forward: 5'-TAC CCT CAG TTC TGC GCT TT-3'; reverse: 5'-GGG ATC TCA ATG TGG

TGC TT-3'), eNOS (forward: 5'-AGG CTC TCA CCT TCT TCC TG-3'; reverse: 5'-AAC CAC

TTC CAC TCC TCG TA-3'), ESR1 (forward: 5'-CAG GCA CAT GAG CAA CAA AG-3';

reverse: 5'-TCC AGC AGC AGG TCG TAG AG-3'), ESR2 (forward: 5'-CTG AAG CAT

GAA CTC CAG CAC-3'; reverse: 5'-CAG GAA GGA CCA CAT AGC AGA-3'), PGR

(forward: 5’-AAC GAA AGC CAA GCC CTA AG-3’; reverse: 5'-GCT GGA GGT ATC

AGG TTT GC-3'), PTGER2 (forward: 5'-TAA GCG TCC TTG TTG AAT GA-3'; reverse: 5'-

GCC CAA ATC CAC TAC CTA AA-3’), PTGER4 (forward: 5'-TCC CCC TTG GAA ATG

TGT ATA G-3'; reverse: 5'- ACA TGC CCA AGT TTT GTA CCT C-3'), PTGFR (forward:

5'-CAG ATT TCT TTG GGC ACC TC-3'; reverse: 5'-AAA GTG GGC ACA GAC CAG AG-

3'), EDNRA (forward: 5’-GCA TCC AGT GGA AGA ACC AT -3’; reverse: 5’- AAC CAG TCA ACC

CTT CAA CG -3’), EDNRB (forward: 5’-GCT CCA TCC CAC TCA GAA AA -3’; reverse: 5’-GCT

CCA TCC CAC TCA GAA AA -3’), HSP90 (forward: 5'-GTA TGG ACA ATG ACT CCA ATC

AAG T-3'; reverse: 5'-CCG TTT GTT GTA AGG TGT GTA TGT A-3'), mPGES-1 (forward:

5'-AGG ACG CTC AGA GAC ATG GA-3'; reverse: 5'-TTC GGT CCG AGG AAA GAG TA-

3'), mPGES-2 (forward: 5'-CCT ACA GAA AGG TGC CCA TC-3'), cPGES (forward: 5'–

AAG GAG AAT CTG GCC AGT CA-3'; reverse: 5'-TCG GAA TCA TCT TCC CAG TC-3'),

PGFS (forward: 5'-TGG CCT CAA CAG CAA TAT GA-3'; reverse: 5'-TGC ACG TGT ACA

Page 55: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

53

GCT CAC AG-3') and CBR1 (forward: 5'-AAA ACC GCA AGG CAG AGT GGT G-3';

reverse: 5'-CTC CAT ATG CGG TAT CGG GCC A-3'). The specificity of each primer set

was confirmed by running the PCR products on a 2.0% agarose gel. Protocol conditions were

consisted of denaturation at 95°C for 30 sec, followed by 45 cycles at 95°C for 6 sec, 60°C

for 6 sec and 72°C for 6 sec with a final dissociation (melting) curve analysis. To standardize

the relative level of expression of each mRNA, three potential housekeeping genes, β-actin

(ACTB; forward: 5'-CAG CAA GCA GGA GTA CGA TG-3'; reverse: 5'-AGC CAT GCC AAT

CTC ATC TC-3'), 18S ribosomal RNA (18S rRNA; forward: 5'-TCG CGG AAG GAT TTA

AAG TG-3'; reverse: 5'-AAA CGG CTA CCA CAT CCA AG-3') and glyceraldehyde-3-

phosphate dehydrogenase (GAPDH forward: 5'-CAC CCT CAA GAT TGT CAG CA-3';

reverse: 5'-GGT CAT AAG TCC CTC CAC GA-3') were initially tested. GAPDH was found

to be the most stable of the three genes by Normfinder software (http://moma.dk/normfinder-

software), and so GAPDH transcripts were selected as the internal control in our experiments.

Amplification efficiencies of all the primers were checked by determination of Ct values for

a dilution series of the target template. Efficiencies of all the primers were 95-100%. The

relative level of expression of each mRNA was measured with the 2-∆∆CT method was used

(Livak & Schmittgen 2001).

Immunohistochemistry

Six-μm sections were deparaffined and rehydrated in a graded series of ethanol and washed

in tap-water. Antigens were retrieved by using microwave in Tris-EDTA buffer (pH 9.0, for

iNOS, eNOS and PGR) or in 0.01 mo/L citrate buffer (pH 6.0, the other antigens) for 15 min

at 600W. Nonspecific binding was blocked in 2.5% horse serum (S-2012, Vector Laboratories

Inc, Burlingame, CA, USA) for 20 min at room temperature. The sections were incubated with

specific primary antibodies for iNOS (160862, Cayman Chemical, Ann Arbor, MI, USA),

eNOS (160880, Cayman Chemical), ESR1 (SAB2100712, Sigma-Aldrich), PGR

(SAB4502185, Sigma-Aldrich), PTGER2 (ab167171, Abcam, Cambridge, UK), PTGER4

(ab133170, Abcam) or PTGFR (101802, Cayman Chemical) overnight at 4°C, washed with

PBS 3 times and incubated with secondary antibody for rabbit-IgG conjugated with Alexa 488

(21206, Life Technologies) for 1 h at room temperature, washed with PBS 3 times, covered

with ProLong Gold Antifade Reagent with DAPI (36935, Life Technologies) and observed

using a fluorescence microscope (FSX100, Olympus, Tokyo, Japan).

Page 56: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

54

Western blotting

Each protein in the cultured bovine oviductal cells was detected by Western blotting analysis

as described previously (Nishimura et al. 2008). Briefly, the cultured cells were lysed in 200

μl lysis buffer, and the protein concentrations were determined by BCA method (Osnes et al.

1993). The proteins were heated with SDS gel-loading buffer containing 1% (v/v) β-

mercaptoethanol (Wako Pure Chemical Industries Ltd., Osaka, Japan) at 95°C for 10 min. The

samples (50 μg protein) were loaded on 10% (v/v) SDS-PAGE (200 V, 80 min), and

transblotted to a 0.2-μm nitrocellulose membrane (GE Healthcare, Milwaukee, WI, USA; 250

mA, 180 min). The membrane was then incubated in PVDF Blocking Reagent for Can Get

Signal (TOYOBO, Osaka, Japan) for 60 min at room temperature. After the blocking, the

membrane was separated into two pieces, and each membrane was incubated separately with

specific primary antibodies to HSP90 (anti-HSP90-IgG-mouse; Abcam, Cambridge, UK,

1:1000 dilution), iNOS (anti-iNOS-IgG-rabbit; sc-651; Santa Cruz Biotechnology, Dallas, TX, USA;

1:1000) and ACTB (anti-b-actin-IgG-mouse; Sigma-Aldrich, 1:20000 dilution) in Can Get

Signal Immunoreaction Enhancer Solution 1 (TOYOBO) overnight at 4°C. After the

incubation, the membranes were incubated again with secondary antibody (anti-mouse, HRP-

linked whole antibody produced in sheep; GE Healthcare, 1:5000 dilution for HSP90 and 1:

40000 dilution for ACTB) in Can Get Signal Immunoreaction Enhancer Solution 2 for 60 min

at room temperature. The signal was detected by ECL Western Blotting Detection System

(GE Healthcare), and the intensity of the immunological reaction was estimated by measuring

the optical density in the defined area by computerized densitometry using Image J (National

Institutes of Health, USA).

Isometric contraction test

Isometric contraction test of oviductal smooth muscle was performed as described previously

(Sogawa et al. 2010, Ning et al. 2014) with some modifications. Briefly, the ampullary and

isthmic tissues of bovine oviducts obtained from cows after ovulation were cut open, and 3

mm-length strips were prepared. Each strip was incubated in a Magnus tube filled with 10 mL

of Krebs-Ringer solution (136.9 mM NaCl, 5.4 mM KCl, 1.5 mM CaCl2, 1.0 mM MgCl2,

23.8 mM NaHCO3, 5.6 mM glucose). The Krebs-Ringer solution was kept at 38.5°C, and

aerated with 95% O2 and 5% CO2 during the experiment. The strips were equilibrated for 1

h before the experiment. The tension of each strip was measured under a resting tension of 1

g. Oxytocin (0.1 μM) and noradrenalin (1 μM) were utilized as a constrictor and a relaxant,

Page 57: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

55

respectively. Then, the strips responded to the constrictor and relaxant utilized for the

incubation with EDN1, EDN2 or EDN3 (0.1 nM, 1 nM or 10 nM). The isometric tension of

each strip was recorded using a force-displacement transducer (Minebea Co. Ltd., Nagano,

Japan) connected to a polygraph (Yokogawa Electric Corp., Tokyo, Japan) with the chart

running at 1000 mm/h.

Statistical analysis

All experimental data are shown as the mean ± SEM. The statistical significance of

differences was assessed by analysis of variance (ANOVA) followed by Tukey-Kramer test

for multiple comparisons using GraphPad Prism (GraphPad Software, La Jolla, CA, USA). P

values less than 0.05 were considered to be statistically significant.

Page 58: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

56

Figure 17 Oviducts were separated into five sections (1, infundibulum; 2, ampulla; 3,

ampullary-isthmic junction; 4, isthmus; 5, utero-tubal junction). The ampulla and the isthmus

sections were utilized for mRNA determination and epithelial cell culture.

Page 59: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

57

Figure 18 Representative photomicrographs of immunostaining by anti-cytokeratin and anti-

vimentin antibody in ampullary oviductal epithelial cells (A, B) and endometrial stromal cells

(C, D). Alexa 594 (red) was used for staining cytokeratin and FITC (green) was also used for

staining vimentin as the secondary antibody. DAPI (blue) was used to visualize nuclei. Scale

is the same in all photomicrographs. Staining in the isthmus was virtually the same as that in

the ampulla.

Page 60: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

58

REFERENCES

Acosta T, Hayashi K, Ohtani M & Miyamoto A 2003 Local changes in blood flow within the

preovulatory follicle wall and early corpus luteum in cows. Reproduction 125 759-767.

Acosta T, Miyamoto A, Ozawa T, Wijayagunawardane M & Sato K 1998 Local release of steroid

hormones, prostaglandin E2, and endothelin-1 from bovine mature follicles In vitro: effects of

luteinizing hormone, endothelin-1, and cytokines. Biology of Reproduction 59 437-443.

Acosta T, Ozawa T, Kobayashi S, Hayashi K, Ohtani M, Kraetzl W, Sato K, Schams D &

Miyamoto A 2000 Periovulatory changes in the local release of vasoactive peptides, prostaglandin f2α,

and steroid hormones from bovine mature follicles in vivo. Biology of Reproduction 63 1253-1261.

Al-Alem L, Bridges P, Su W, Gong M, Iglarz M & Ko C 2007 Endothelin-2 induces oviductal

contraction via endothelin receptor subtype A in rats. Journal of Endocrinology 193 383-391.

Arosh J, Parent J, Chapdelaine P, Sirois J & Fortier M 2002 Expression of cyclooxygenases 1

and 2 and prostaglandin E synthase in bovine endometrial tissue during the estrous cycle. Biology of

Reproduction 67 161-169.

Bredt DS & Snyder SH 1994 Nitric oxide: a physiologic messenger molecule. Annual Review of

Biochemistry.

Bridges P, Brusie M & Fortune J 2005 Elevated temperature (heat stress) in vitro reduces

androstenedione and estradiol and increases progesterone secretion by follicular cells from bovine

dominant follicles. Domestic Animal Endocrinology 29 508-522.

Bridges PJ, Cho J & Ko CM 2011 Endothelins in regulating ovarian and oviductal function.

Frontiers in Bioscience 3 145-155.

Cavestany D, el-Wishy A & Foote R 1985 Effect of season and high environmental temperature

on fertility of Holstein cattle. Journal of Dairy Science 68 1471-1478.

Chiu PC, Liao S, Lam KK, Tang F, Ho JC, Ho PC, O WS, Yao YQ & Yeung WS 2010

Adrenomedullin regulates sperm motility and oviductal ciliary beat via cyclic adenosine 5'-

monophosphate/protein kinase A and nitric oxide. Endocrinology 151 3336-3347.

Coleman RA, Smith WL & Narumiya S 1994 International Union of Pharmacology classification

of prostanoid receptors: properties, distribution, and structure of the receptors and their subtypes.

Pharmacological Reviews 46 205-229.

Collier R, Dahl G & VanBaale M 2006 Major advances associated with environmental effects on

dairy cattle. Journal of Dairy Science 89 1244-1253.

Compton DR, Sheng S, Carlson KE, Rebacz NA, Lee IY, Katzenellenbogen BS &

Page 61: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

59

Katzenellenbogen JA 2004 Pyrazolo[1,5-a]pyrimidines: estrogen receptor ligands possessing

estrogen receptor β antagonist activity. Journal of Medicinal Chemistry 47 5872-5893.

Croxatto H 2002 Physiology of gamete and embryo transport through the fallopian tube.

Reproductive BioMedicine Online 4 160-169.

De Rensis F & Scaramuzzi R 2003 Heat stress and seasonal effects on reproduction in the dairy

cow--a review. Theriogenology 60 1139-1151.

Ekerhovd E, Brännström M, Alexandersson M & Norström A 1997 Evidence for nitric oxide

mediation of contractile activity in isolated strips of the human Fallopian tube. Human Reproduction

12 301-305.

Ekerhovd E, Brännström M, Weijdegård B & Norström A 1999 Localization of nitric oxide

synthase and effects of nitric oxide donors on the human Fallopian tube. Molecular Human

Reproduction 5 1040-1047.

Ezzati M, Djahanbakhch O, Arian S & Carr BR 2014 Tubal transport of gametes and embryos:

a review of physiology and pathophysiology. Journal of Assisted Reproduction and Genetics 31 1337-

1347.

Freeman D, Weber J, Geary R & Woods G 1991 Time of embryo transport through the mare

oviduct. Theriogenology 36 823-830.

Gabler C, Odau S, Muller K, Schon J, Bondzio A & Einspanier R 2008 Exploring cumulus-

oocyte-complex-oviductal cell interactions: gene profiling in the bovine oviduct. Journal of

Physiology and Pharmacology 59 Suppl 9 29-42.

Garavito R, Malkowski M & DeWitt D 2002 The structures of prostaglandin endoperoxide H

synthases-1 and -2. Prostaglandins and Other Lipid Mediators 68-69 129-152.

Gauvreau D, Moisan V, Roy M, Fortier M & Bilodeau JF 2010 Expression of prostaglandin E

synthases in the bovine oviduct. Theriogenology 73 103-111.

Gawronska B, Paukku T, Huhtaniemi I, Wasowicz G & Ziecik A 1999 Oestrogen-dependent

expression of LH/hCG receptors in pig Fallopian tube and their role in relaxation of the oviduct.

Journal of Reproduction and Fertility 115 293-301.

Gotoh T & Mori M 2006 Nitric oxide and endoplasmic reticulum stress. Arteriosclerosis,

Thrombosis, and Vascular Biology 26 1439-1446.

Goud AP, Goud PT, Diamond MP & Abu-Soud HM 2005 Nitric oxide delays oocyte aging.

Biochemistry 44 11361-11368.

Gouge R, Marshburn P, Gordon B, Nunley W & Huet-Hudson Y 1998 Nitric oxide as a regulator

of embryonic development. Biology of Reproduction 58 875-879.

Page 62: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

60

Gwazdauskas F, Wilcox C & Thatcher W 1975 Environmental and managemental factors

affecting conception rate in a subtropical climate. Journal of Dairy Science 58 88-92.

Halbert S, Tam P & Blandau R 1976 Egg transport in the rabbit oviduct: the roles of cilia and

muscle. Science 191 1052-1053.

Hansen P 2009 Effects of heat stress on mammalian reproduction. Philosophical Transactions of

the Royal Society of London. Series B, Biological Sciences 364 3341-3350.

Harris JA, West AK & Chuah MI 2009 Olfactory ensheathing cells: nitric oxide production and

innate immunity. Glia 57 1848-1857.

Helm G, Owman C, Sjöberg N & Walles B 1982 Motor activity of the human Fallopian tube in

vitro in relation to plasma concentration of oestradiol and progesterone, and the influence of

noradrenaline. Journal of Reproduction and Fertility 64 233-242.

Hirata Y, Emori T, Eguchi S & Kanno K 1993 Endothelin receptor subtype B mediates synthesis

of nitric oxide by cultured bovine endothelial cells. Journal of Clinical Investigation 91 1367-1373.

Hristovska AM, Rasmussen LE, Hansen PBL, Nielsen SS, Nusing RM, Narumiya S, Vanhoutte

P, Skott O & Jensen BL 2007 Prostaglandin E2 Induces Vascular Relaxation by E-Prostanoid 4

Receptor-Mediated Activation of Endothelial Nitric Oxide Synthase. Hypertension 50.

Hunter R 2012 Components of oviduct physiology in eutherian mammals. Biological Reviews of

the Cambridge Philosophical Society 87 244-255.

Ignarro L, Buga G, Wei L, Bauer P, Wu G & del Soldato P 2001 Role of the arginine-nitric oxide

pathway in the regulation of vascular smooth muscle cell proliferation. Proceedings of the National

Academy of Sciences of the United States of America 98 4202-4208.

Jeoung M, Lee S, Hawng H-K, Cheon Y-P, Jeong Y, Gye M, Iglarz M, Ko C & Bridges P 2010

Identification of a novel role for endothelins within the oviduct. Endocrinology 151 2858-2867.

Kölle S, Dubielzig S, Reese S, Wehrend A, König P & Kummer W 2009 Ciliary transport, gamete

interaction, and effects of the early embryo in the oviduct: ex vivo analyses using a new digital

videomicroscopic system in the cow. Biology of Reproduction 81 267-274.

Kobayashi Y, Yamamoto Y, Kageyama S, Hirayama H, Kimura K & Okuda K 2016 Regulation

of bovine oviductal NO synthesis by follicular steroids and prostaglandins. Reproduction. 151 577-

587.

Kregel K 2002 Heat shock proteins: modifying factors in physiological stress responses and

acquired thermotolerance. Journal of Applied Physiology 92 2177-2186.

Labarca C & Paigen K 1980 A simple, rapid, and sensitive DNA assay procedure. Analytical

Biochemistry 102 344-352.

Page 63: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

61

Lapointe J, Roy M, St-Pierre I, Kimmins S, Gauvreau D, MacLaren L & Bilodeau J-F 2006

Hormonal and spatial regulation of nitric oxide synthases (NOS) (neuronal NOS, inducible NOS, and

endothelial NOS) in the oviducts. Endocrinology 147 5600-5610.

Lee S-H, Acosta T, Yoshioka S & Okuda K 2009 Prostaglandin F2α regulates the nitric oxide

generating system in bovine luteal endothelial cells. Journal of Reproduction and Development 55

418-424.

Livak K & Schmittgen T 2001 Analysis of relative gene expression data using real-time

quantitative PCR and the 2-ΔΔ CT Method. Methods 25 402-408.

Malayer J, Hansen P, Gross T & Thatcher W 1990 Regulation of heat shock-induced alterations

in the release of prostaglandins by the uterine endometrium of cows. Theriogenology 34 219-230.

Malayer J & Woods V 1998 Expression of estrogen receptor and maintenance of hormone-

responsive phenotype in bovine fetal uterine cells. Domestic Animal Endocrinology 15 141-154.

Manser R, Leese H & Houghton F 2004 Effect of inhibiting nitric oxide production on mouse

preimplantation embryo development and metabolism. Biology of Reproduction 71 528-533.

Menezo Y & Guerin P 1997 The mammalian oviduct: biochemistry and physiology. European

Journal of Obstetrics, Gynecology, and Reproductive Biology 73 99-104.

Miraglia E, De Angelis F, Gazzano E, Hassanpour H, Bertagna A, Aldieri E, Revelli A & Ghigo

D 2011 Nitric oxide stimulates human sperm motility via activation of the cyclic GMP/protein kinase

G signaling pathway. Reproduction 141 47-54.

Miyamoto Y, Skarzynski D & Okuda K 2000 Is tumor necrosis factor α a trigger for the initiation

of endometrial prostaglandin F2α release at luteolysis in cattle? Biology of Reproduction 62 1109-1115.

Murakami M, Nakatani Y, Tanioka T & Kudo I 2002 Prostaglandin E synthase. Prostaglandins

and Other Lipid Mediators 68-69 383-399.

Nabenishi H, Ohta H, Nishimoto T, Morita T, Ashizawa K & Tsuzuki Y 2011 Effect of the

temperature-humidity index on body temperature and conception rate of lactating dairy cows in

southwestern Japan. Journal of Reproduction and Development 57 450-456.

Ning N, Zhu J, Du Y, Gao X, Liu C & Li J 2014 Dysregulation of hydrogen sulphide metabolism

impairs oviductal transport of embryos. Nature Communications 5 4107.

Nishimura R, Komiyama J, Tasaki Y, Acosta T & Okuda K 2008 Hypoxia promotes luteal cell

death in bovine corpus luteum. Biology of Reproduction 78 529-536.

Noreikat K, Wolff M, Kummer W & Kölle S 2012 Ciliary activity in the oviduct of cycling,

pregnant, and muscarinic receptor knockout mice. Biology of Reproduction 86 120.

Okuda K, Kito S, Sumi N & Sato K 1988 A study of the central cavity in the bovine corpus luteum.

Page 64: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

62

Veterinary Record 123 180-183.

Okuda K, Miyamoto Y & Skarzynski D 2002 Regulation of endometrial prostaglandin F2α

synthesis during luteolysis and early pregnancy in cattle. Domestic Animal Endocrinology 23 255-264.

Osnes T, Sandstad O, Skar V, Osnes M & Kierulf P 1993 Total protein in common duct bile

measured by acetonitrile precipitation and a micro bicinchoninic acid (BCA) method. Scandinavian

Journal of Clinical and Laboratory Investigation 53 757-763.

Pérez-Rodríguez R, Roncero C, Oliván AM, González MPP & Oset-Gasque MJJ 2009

Signaling mechanisms of interferon γ induced apoptosis in chromaffin cells: involvement of nNOS,

iNOS, and NFκB. Journal of Neurochemistry 108 1083-1096.

Pearl L & Prodromou C 2006 Structure and mechanism of the Hsp90 molecular chaperone

machinery. Annual Review of Biochemistry 75 271-294.

Pennarossa G, Maffei S, Rahman M, Berruti G, Brevini T & Gandolfi F 2012 Characterization

of the constitutive pig ovary heat shock chaperone machinery and its response to acute thermal stress

or to seasonal variations. Biology of Reproduction 87 119.

Pollock DM, Keith TL & Highsmith RF 1995 Endothelin receptors and calcium signaling. FASEB

Journal 9 1196-1204.

Priyadarsana M, Wijayagunawardane B & Miyamoto A 2004 Endothelin-1 system in the bovine

oviduct: a regulator of local contraction and gamete transport. Journal of Cardiovascular

Pharmacology 44 Suppl 1 51.

Putney D, Malayer J, Gross T, Thatcher W, Hansen P & Drost M 1988 Heat stress-induced

alterations in the synthesis and secretion of proteins and prostaglandins by cultured bovine conceptuses

and uterine endometrium. Biology of Reproduction 39 717-728.

Reeves PR 1978 Distribution, elimination, and residue studies in the cow with the synthetic

prostaglandin estrumate. Journal of Agricultural and Food Chemistry 26 152-155.

Rosselli M, Imthurn B, Macas E, Keller P & Dubey R 1994 Endogenous nitric oxide modulates

endothelin-1 induced contraction of bovine oviduct. Biochemical and Biophysical Research

Communications 201 143-148.

Saint-Dizier M, Sandra O, Ployart S, Chebrout M & Constant F 2012 Expression of nuclear

progesterone receptor and progesterone receptor membrane components 1 and 2 in the oviduct of

cyclic and pregnant cows during the post-ovulation period. Reproductive Biology and Endocrinology

10 76.

Sakamoto S, Aso T, Masuda H & Goto M 1999 Gestational changes in endothelin-1-induced

receptors and myometrial contractions in rat. Molecular Human Reproduction 5 270-276.

Page 65: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

63

Sakatani M, Kobayashi S-I & Takahashi M 2004 Effects of heat shock on in vitro development

and intracellular oxidative state of bovine preimplantation embryos. Molecular Reproduction and

Development 67 77-82.

Sakumoto R, Komatsu T, Kasuya E, Saito T & Okuda K 2006 Expression of mRNAs for

interleukin-4, interleukin-6 and their receptors in porcine corpus luteum during the estrous cycle.

Domestic Animal Endocrinology 31 246-257.

Shaw JL, Wills GS, Lee K-FF, Horner PJ, McClure MO, Abrahams VM, Wheelhouse N,

Jabbour HN, Critchley HO, Entrican G & Horne AW 2011 Chlamydia trachomatis infection

increases fallopian tube PROKR2 via TLR2 and NFκB activation resulting in a microenvironment

predisposed to ectopic pregnancy. American Journal of Pathology 178 253-260.

Siemieniuch M, Woclawek-Potocka I, Deptula K, Okuda K & Skarzynski D 2009 Effects of

tumor necrosis factor-α and nitric oxide on prostaglandins secretion by the bovine oviduct differ in the

isthmus and ampulla and depend on the phase of the estrous cycle. Experimental Biology and Medicine

234 1056-1066.

Sogawa C, Abe A, Tsuji T, Koizumi M, Saga T & Kunieda T 2010 Gastrointestinal tract disorder

in natriuretic peptide receptor B gene mutant mice. American Journal of Pathology 177 822-828.

Suarez S 2008 Regulation of sperm storage and movement in the mammalian oviduct. International

Journal of Developmental Biology 52 455-462.

Sun J, Huang YR, Harrington WR, Sheng S, Katzenellenbogen JA & Katzenellenbogen BS

2002 Antagonists selective for estrogen receptor α. Endocrinology 143 941-947.

Szóstek A, Siemieniuch M, Deptula K, Woclawek-Potocka I, Majewska M, Okuda K &

Skarzynski D 2011 Ovarian steroids modulate tumor necrosis factor-α and nitric oxide-regulated

prostaglandin secretion by cultured bovine oviductal epithelial cells. Domestic Animal Endocrinology

41 14-23.

Tanikawa M, Acosta T, Fukui T, Murakami S, Korzekwa A, Skarzynski D, Piotrowska K, Park

C & Okuda K 2005 Regulation of prostaglandin synthesis by interleukin-1α in bovine endometrium

during the estrous cycle. Prostaglandins and Other Lipid Mediators 78 279-290.

Tanikawa M, Lee H-Y, Watanabe K, Majewska M, Skarzynski D, Park S-B, Lee D-S, Park C-

K, Acosta T & Okuda K 2008 Regulation of prostaglandin biosynthesis by interleukin-1 in cultured

bovine endometrial cells. Journal of Endocrinology 199 425-434.

Tanioka T, Nakatani Y, Kobayashi T, Tsujimoto M, Oh-ishi S, Murakami M & Kudo I 2003

Regulation of cytosolic prostaglandin E2 synthase by 90-kDa heat shock protein. Biochemical and

Biophysical Research Communications 303 1018-1023.

Page 66: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

64

Tsukahara H, Ende H, Magazine HI, Bahou WF & Goligorsky MS 1994 Molecular and

functional characterization of the non-isopeptide-selective ETB receptor in endothelial cells. Receptor

coupling to nitric oxide synthase. Journal of Biological Chemistry 269 21778-21785.

Uenoyama Y, Hattori S, Miyake M & Okuda K 1997 Up-regulation of oxytocin receptors in

porcine endometrium by adenosine 3',5'-monophosphate. Biology of Reproduction 57 723-728.

Ulbrich S, Kettler A & Einspanier R 2003 Expression and localization of estrogen receptor α,

estrogen receptor β and progesterone receptor in the bovine oviduct in vivo and in vitro. Journal of

Steroid Biochemistry and Molecular Biology 84 279-289.

Ulbrich S, Rehfeld S, Bauersachs S, Wolf E, Rottmayer R, Hiendleder S, Vermehren M,

Sinowatz F, Meyer H & Einspanier R 2006 Region-specific expression of nitric oxide synthases in

the bovine oviduct during the oestrous cycle and in vitro. Journal of Endocrinology 188 205-213.

Ulbrich S, Zitta K, Hiendleder S & Wolf E 2010 In vitro systems for intercepting early embryo-

maternal cross-talk in the bovine oviduct. Theriogenology 73 802-816.

Vignon-Zellweger N, Heiden S, Miyauchi T & Emoto N 2012 Endothelin and endothelin

receptors in the renal and cardiovascular systems. Life Sciences 91 490-500.

Wang H-HH, Hsieh H-LL & Yang C-MM 2011 Nitric oxide production by endothelin-1 enhances

astrocytic migration via the tyrosine nitration of matrix metalloproteinase-9. Journal of Cellular

Physiology 226 2244-2256.

Wijayagunawardane M, Choi Y, Miyamoto A, Kamishita H, Fujimoto S, Takagi M & Sato K

1999 Effect of ovarian steroids and oxytocin on the production of prostaglandin E2, prostaglandin F2α

and endothelin-1 from cow oviductal epithelial cell monolayers in vitro. Animal Reproduction Science

56 11-17.

Wijayagunawardane M, Kodithuwakku S, De Silva N & Miyamoto A 2009 Angiotensin II

secretion by the bovine oviduct is stimulated by luteinizing hormone and ovarian steroids. Journal of

Reproduction and Development 55 570-575.

Wijayagunawardane M, Miyamoto A, Cerbito W, Acosta T, Takagi M & Sato K 1998 Local

distributions of oviductal estradiol, progesterone, prostaglandins, oxytocin and endothelin-1 in the

cyclic cow. Theriogenology 49 607-618.

Wijayagunawardane M, Miyamoto A, Taquahashi Y, Gabler C, Acosta T, Nishimura M,

Killian G & Sato K 2001a In vitro regulation of local secretion and contraction of the bovine oviduct:

stimulation by luteinizing hormone, endothelin-1 and prostaglandins, and inhibition by oxytocin.

Journal of Endocrinology 168 117-130.

Wijayagunawardane MP, Miyamoto A, Taquahashi Y, Acosta TJ, Nishimura M & Sato K

Page 67: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

65

2001b Angiotensin II and atrial natriuretic peptide in the cow oviductal contraction in vitro: direct

effect and local secretion of prostaglandins, endothelin-1, and angiotensin II. Biology of Reproduction

65 799-804.

Wolfenson D, Lew B, Thatcher W, Graber Y & Meidan R 1997 Seasonal and acute heat stress

effects on steroid production by dominant follicles in cows. Animal Reproduction Science 47 9-19.

Woodward DF, Pepperl DJ, Burkey TH & Regan JW 1995 6-Isopropoxy-9-oxoxanthene-2-

carboxylic acid (AH 6809), a human EP2 receptor antagonist. Biochemical Pharmacology 50 1731-

1733.

Yamamoto Y, Kohka M & Kobayashi Y 2014 Endothelin as a local regulating factor in the bovine

oviduct. Reproduction, Fertility, and Development.

Yanagisawa M 1994 The endothelin system. A new target for therapeutic intervention. Circulation

89 1320-1322.

Yanagisawa M, Kurihara H, Kimura S & Tomobe Y 1988 A novel potent vasoconstrictor peptide

produced by vascular endothelial cells. Nature 332 411-415.

Yilmaz O, Całka J, Bukowski R, Zalecki M, Wasowicz K, Jaroszewski J, Markiewicz W,

Bulbul A & Ucar M 2012 Nitric oxide in the bovine oviduct: influence on contractile activity and

nitric oxide synthase isoforms localization. Theriogenology 77 1312-1327.

Zhang J, Gong Y & Yu Y 2010 PG F2α Receptor: A Promising Therapeutic Target for

Cardiovascular Disease. Frontiers in Pharmacology 1 116.

Zhang K, Rajput SK, Lee K-BB, Wang D, Huang J, Folger JK, Knott JG, Zhang J & Smith

GW 2015 Evidence supporting a role for SMAD2/3 in bovine early embryonic development: potential

implications for embryotropic actions of follistatin. Biology of Reproduction 93 86.

Zhao Y, Chegini N & Flanders KC 1994 Human fallopian tube expresses transforming growth

factor (TGF β) isoforms, TGF β type I-III receptor messenger ribonucleic acid and protein, and

contains [125I]TGF β-binding sites. Journal of Clinical Endocrinology and Metabolism 79 1177-1184.

Page 68: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

66

ABSTRACT in JAPANESE

卵管内の微細環境調節メカニズム

小林 芳彦

【背景】哺乳動物の卵管は精子と卵母細胞が出会い受精する場であり、新たな生命

が生まれる場所である。ウシやヒトの場合、卵管内で卵母細胞と精子が受精してから

子宮へ運ばれるまでの期間は 1 週間に満たないが、この短期間に精子の成熟 (超活

性化および先体反応)、卵母細胞の輸送、受精、初期胚の発育および子宮への輸送とい

った多様な生理現象が起こる。それぞれの現象に適した微細環境を整えるのが卵管上

皮の分泌細胞である。短期間に種々の生理現象が起こるため、卵管の分泌機能は緻密

に制御される必要がある。卵管の分泌機能は、1) 卵巣から分泌されるホルモン、およ

び 2) 卵管上皮細胞が分泌する生理活性物質によって制御されると考えられる。卵巣

から分泌される estradiol-17β (E2) ならびに progesterone (P4) は血液を介して内分泌

的に卵管機能を支配し、その血中濃度の変化によって卵管内腔の微細環境を大きく変

化させる。これまでにこれらの卵巣ステロイドホルモンは prostaglandin (PG) E2、 PGF2α および endothelin-1 (EDN1) などの卵管上皮細胞から分泌され平滑筋運動を調

節する因子の分泌活性を調節することが報告されている。これら卵管上皮細胞から分

泌される生理活性物質は、さらに卵管上皮細胞自身に自己分泌的に作用することで卵

管内腔の緻密な環境を生み出すと考えられる。本研究では第一に、卵管分泌機能制御

機構の一端解明を目的として、卵管上皮細胞の分泌する一酸化窒素 (NO) 合成に及ぼ

す卵胞由来ホルモンならびに卵管由来の EDN1 の影響を検討した。次に、卵管分泌

機能に及ぼす外的な要因に着目した。ウシにおいて、夏季の暑熱ストレスが発情兆候

の微弱化、卵胞の発育障害などを引き起こし、受胎率を低下させることがこれまでに

明らかにされてきた。また、暑熱ストレスが主席卵胞の卵胞液中 E2 濃度を減少させ

ること、子宮由来の黄体退行因子である PGF2α 分泌を刺激し、妊娠維持に必須の黄

体を退行させる可能性が示されている。暑熱ストレスは初期胚への影響も報告されて

おり、高温環境下での培養は初期胚の死滅を招く。その一方で、卵管機能に及ぼす暑

熱ストレスの影響に関して検討された報告はない。本研究では第二に、冬季および

夏季に採取したウシ卵管組織における PG 合成酵素発現を比較することで、夏季に

おいて卵管生理機能に及ぶ影響を推察した。

【方法】1) Day 0 (排卵日)、Days 2-3、Days 5-6、Days 8-12、Days 15-17 および Days 19-21 の卵管膨大部および峡部の組織を採取し、内皮型 NO 合成酵素 (eNOS)、誘導

型 NOS (iNOS)、estrogen receptor α (ESR1)、ESR2、P4 receptor、E-prostanoid receptors

Page 69: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

67

(PTGER2 および PTGER4) および F-prostanoid receptor mRNA 発現量を検討した。

2) 排卵時の卵胞液中各ホルモン濃度を調べるために、過排卵処理を施したウシより

排卵直前の卵胞液を採取し、卵胞液中の E2, P4, PGE2 および PGF2α 濃度を測定した。

3) 単離した卵管膨大部および峡部上皮細胞に E2 (0.1,1,10 nM)、P4 (1, 10, 100 nM)、PGE2 (0.01,0.1,1 nM) または PGF2α (0.01,0.1,1 nM) をそれぞれ添加し、iNOS mRNA 発現量を検討した。各分子の添加濃度は血中および卵胞液中の濃度を模した。また、

E2 (10 nM) を ESR1 または ESR2 それぞれの特異的拮抗剤と、PGE2 (1 μM) を PTGER2 または PTGER4 それぞれの特異的拮抗剤と組み合わせて添加し、iNOS mRNA 発現量を検討した。4) 単離した培養細胞の iNOS mRNA およびタンパク質

発現に及ぼす ET-1 の影響を検討した。5) ウシ卵管膨大部および峡部組織の ET 受

容体 A (EDNRA) および B (EDNRB) の局在および発現量を検討した。6) 冬季 (11月下旬-3 月上旬) および夏季 (7 月下旬-9 月上旬) において、排卵後 0-6 日のウシ

卵管組織を採取し、組織中の PG 合成酵素発現を測定した。7) 単離した卵管上皮細

胞を 38.5°C および 40.5°C で培養した後、培養液中の PGE2 および PGF2α 濃度を測

定した。

【結果】1) 卵管膨大部の iNOS mRNA 発現量は他の周期と比較し排卵日に高かっ

た。一方峡部の iNOS mRNA 発現量は黄体形成期に最も高く、卵胞期に低かった。

峡部の ERα mRNA 発現量は他の周期と比較し卵胞期に高かった。2) 卵胞液中の各

物質の濃度は E2: 106.3-409.9 nM, P4: 771.0-2763 nM, PGE2: 0.006-0.04 μM、PGF2α: 0.06-0.83 μM であった。3) 峡部上皮細胞の iNOS mRNA 発現量は E2 の 24 h 感作によ

って抑制された。この E2 による iNOS mRNA 発現量の抑制作用は ERα 特異的拮

抗剤によって低減された。膨大部上皮細胞の iNOS mRNA 発現量は PGE2 および PGF2α の 1 h 感作によって刺激された。PGE2 による iNOS 刺激作用は PTGER2 特異的拮抗剤によって抑制された。4) 卵管膨大部上皮細胞において EDN1 は iNOS mRNA およびタンパク質発現を刺激した。5) EDNRA および EDNRB は主に卵管上

皮細胞に局在した。NO 合成誘導作用を持つ受容体である EDNRB は、膨大部にお

いては排卵後に発現上昇が認められた一方峡部においては有意な変化が認められな

かった。6) 夏季ウシ卵管膨大部組織において、cytosolic PGES (cPGES) mRNA が冬

季と比べて高い発現を、carbonyl reductase 1 (CBR1) mRNA は冬季と比べて低い発現

を示した。7) 卵管膨大部上皮細胞において、高温培養 (40.5°C) が PGE2 濃度を刺

激する一方、PGF2α 濃度には影響を及ぼさなかった。

【考察】本研究より、排卵後の卵胞液中に含まれる PGs が卵管膨大部の iNOS 発現/NO 分泌を誘導する可能性が示された。この NO は、排卵後卵管に進入した卵母

細胞の生存に関与する可能性が考えられる。一方卵管峡部では排卵前 (卵胞期) に血

中濃度の増加する E2 により NO 合成が抑制されることが示唆された。NO は精子

Page 70: REGULATION OF MICRO ENVIRONMENT WITHIN THE OVIDUCT

68

の運動性を刺激することが知られることから、この時期に卵管に精子が存在すれば

NO により運動性を調節されるかもしれない。また、卵管上皮由来因子である EDN1 が自己分泌的に NO 分泌を調節すること、さらに排卵後の時期に特に膨大部におい

て NO 分泌を亢進し、卵管内腔の微細環境構築に寄与することが示唆された。夏季

の高温環境においては PGE2 合成酵素の発現が亢進される一方、PGF2α 合成酵素の

発現が抑えられることで PGE2 分泌が異常促進され、PGF2α とのバランスが崩れる可

能性が示された。PGE2 は平滑筋弛緩、PGF2α は収縮誘導因子であることから、夏季

の暑熱が卵管の収縮、弛緩のバランスを乱し、卵母細胞や初期胚が適切に輸送され

ず、受胎率の低下を招くことが考えられる。