• Title/Summary/Keyword: PACAP

Search Result 15, Processing Time 0.03 seconds

Control Mechanisms of Ovulation by Pituitary Adenylate Cyclase-Activating Polypeptide (Pituitary Adenylate Cyclase-Activating Polypeptide에 의한 배란 조절에 관한 연구)

  • Lee, Yu-Il;Kim, Hyoung-Choon;Kim, Mi-Young;Chun, Sang-Young
    • Clinical and Experimental Reproductive Medicine
    • /
    • v.32 no.2
    • /
    • pp.101-111
    • /
    • 2005
  • 배 경: Pituitary adenylate cyclase-activating polypeptide (PACAP)은 양의 시상하부에서 추출된 신경펩타이드 호르몬으로 난소에도 존재하여 배양된 과립막 세포에서 스테로이드합성과 cyclic AMP 형성을 촉진함이 보고되었다. 목 적: 흰쥐 난소를 실험 모델로 사용하여 배란시 황체화호르몬 (luteinizing hormone; LH)에 의해 유도된 PACAP과 PACAP 수용체의 유전자 발현양상과 신호 전달경로를 규명하고자 하였다. 재료 및 방법: 미성숙 흰쥐의 배란전 난포를 체외 배양하면서 LH로 처리하고 PACAP 및 PACAP수용체의 유전자 발현을 보기 위해서는 Northern blot 분석과 in situ hybridization (ISH)을, 그리고 단백질 수준의 PACAP 검색을 위해서는 enzyme linked immunosorbent assay (ELISA) 분석을 이용하였다. 결 과: LH 처리 후 Northern blot상의 PACAP 유전자 발현은 6~9시간에 일시적으로 최고치에 도달하였으며 ISH로 보아 과립막 세포에서 발현됨을 알 수 있었다. ELISA 분석 상 PACAP 단백질도 LH처리 후 6~12시간에 최고치를 나타내었으며, PACAP 수용체 mRNA 역시 3~9시간에 최고치로 과립막 세포에서 발현되었다. Adenylate cyclase (AC) 억제제인 MDL12330A 처리시 LH로 발현된 PACAP mRNA가 감소되며, AC의 활성제인 forskolin 처리에는 LH시와 유사한 PACAP mRNA의 발현양상을 나타내었다. 그러나 protein kinase C (PKC)의 억제제인 chelerythrine과 2-0-tetradecanolphorbol-13-acetate (TPA) 처리로는 PACAP 의 유전자 발현에 영향을 주지 못하였다. 5-lipoxygenase의 억제제인 MK886이나 nordihydroguaiaretic acid (NDGA)로 처리한 결과 LH로 유도된 PACAP 유전자의 발현이 감소되었으나, cyclooxygenase의 억제제인 indomethacin은 별로 영향을 주지 못하였다. MEK와 p38의 억제제인 PD98059와 SB203580도 LH로 촉진 된 PACAP의 유전자 발현을 농도 의존적으로 억제하였다. 결 론 : 배란전 난포에서 PACAP과 PACAP 수용체의 유전자 발현은 모두 LH의 폭발적 분비에 의해 유도되어 일시적으로 과립막 세포에서 나타나 배란을 위한 국소적인 조절 작용을 할 것으로 추정되며, LH로 촉진된 PACAP 유전자 발현을 위한 신호전달은 cAMP-PKA, lipoxygenase 및 MAP kinase 경로를 통하는 것으로 사료된다.

Expression of pituitary adenylate cyclase activating polypeptide in the adult rat testis by in situ hybridization and immunohistochemistry (In situ hybridization법과 면역조직화학적법을 이용한 성숙한 흰쥐고환에서의 pituitary adenylate cyclase activating polypeptide의 발현)

  • Koh, Phil-ok;Kwak, Soo-dong
    • Korean Journal of Veterinary Research
    • /
    • v.41 no.1
    • /
    • pp.1-6
    • /
    • 2001
  • Pituitary adenyl ate cyclase activating polypeptide (PACAP) was originally isolated from the ovine hypothalamus and stimulated cAMP production in anterior pituitary cells. It is known that PACAP stimulates cAMP accumulation and contributes to the spermatogenesis and steroidogenesis in rat testis. The principal aim of this study is to determinate the distribution of PACAP mRNA and protein in adult rat testis. For this study, we used in situ hybridization and immunohistochemistry techniques in adult rat testis. PACAP mRNA was stage specifically expressed in seminiferous tubules. Positive signals of PACAP mRNA were detected in the developing germ cells at stages HI-VII of the epithelial cycle. The strongest signals of PACAP mRNA and protein were detected in round spermatids at stages V to early VII of the cycle. These results demonstrate that PACAP which is synthesised in the developing germ cells contributes to the spermatogenesis in rat testis. Thus, we suggest that PACAP plays a critical role in the function of testis.

  • PDF

The use of pituitary adenylate cyclase-activating polypeptide in the pre-maturation system improves in vitro developmental competence from small follicles of porcine oocytes

  • Park, Kyu-Mi;Kim, Kyu-Jun;Jin, Minghui;Han, Yongquan;So, Kyoung-Ha;Hyun, Sang-Hwan
    • Asian-Australasian Journal of Animal Sciences
    • /
    • v.32 no.12
    • /
    • pp.1844-1853
    • /
    • 2019
  • Objective: We investigated how pituitary adenylate cyclase-activating polypeptide (PACAP) affects embryonic development during pre-in vitro maturation (pre-IVM) using porcine oocytes isolated from small follicles. Methods: We divided the follicles into the experimental groups by size (SF, small follicles; MF, medium follicles) and treated with and without PACAP and cultured for 18 hours (PreSF[-]PACAP; without PACAP, Pre-SF[+]PACAP; with PACAP) before undergoing IVM. The gene expression related to extracellular matrix formation (amphiregulin, epiregulin, and hyaluronan synthase 2 [HAS2]) and apoptosis (Bcl-2-associated X [BAX], B-cell lymphoma 2, and cysteine-aspartic acid protease 3) was investigated after maturation. The impact on developmental competence was assessed by the cleavage and blastocyst rate and total cell number of blastocysts in embryos generated from parthenogenesis (PA) and in vitro fertilization (IVF). Results: Cleavage rates in the Pre-SF(+)PACAP after PA were significantly higher than SF and Pre-SF(-)PACAP (p<0.05). The cleavage rates between MF and Pre- SF(+)PACAP groups yielded no notable differences after IVF. Pre-SF(+)PACAP displayed the higher rate of blastocyst formation and greater total cell number than SF and Pre-SF(-)PACAP (p<0.05). Cumulus cells showed significant upregulation of HAS2 mRNA in the Pre-SF(+)PACAP compared to the SF (p<0.05). In comparison to other groups, the Pre-SF(+)PACAP group displayed a downregulation in mRNA expression of BAX in matured oocytes (p<0.05). Conclusion: The PACAP treatment during pre-IVM improved the developmental potential of porcine oocytes derived from SF by regulating cumulus expansion and apoptosis of oocytes.

Control Mechanisms of Ovarian Follicle Development by Follicle Stimulating Hormone and Pituitary Adenylate Cyclase-activating Polypeptide (난포자극호르몬과 Pituitary Adenylate Cyclase-activating Polypeptide에 의한 난소의 난포성장)

  • Lee, Yu-Il;Shin, Jin-Ok;Kim, Mi-Young;Chun, Sang-Young
    • Clinical and Experimental Reproductive Medicine
    • /
    • v.33 no.1
    • /
    • pp.15-23
    • /
    • 2006
  • Objective: Pituitary adenylate cyclase-activating polypeptide (PACAP), a novel hypothalamic neuropeptide, has been suggested to play a role in ovarian folliculogenesis. The present study evaluated the effect of PACAP on the growth of preantral follicles. Methods: Preantral follicles were mechanically isolated from ovaries of 21-day-old rats and cultured in groups for 3 days in serum-free medium in the absence or presence of PACAP-38 ($10^{-6}M$). Results: Treatment with PACAP-38 resulted in an increase in follicle diameter by 75% whereas treatment with follicle stimulating hormone (FSH) increased follicle diameter by 65%. PACAP-38 treatment enhanced the granulosa cell proliferation as measured by thymidine incorporation analysis. Furthermore, the production of progesterone by cultured granulosa cells and GFSHR-17 cell line was stimulated by PACAP-38. Interestingly, PACAP enhanced FSH action on stimulation of SF-1 and aromatase gene expression. Conclusion: The present results demonstrate that PACAP stimulated preantral follicle growth by potentiating proliferation and by stimulating steroidogenesis.

Expressions of Pituitary Adenylate Cyclase-Activating Polypeptide and Its Receptor Gene in the Rat Uterus (흰쥐 자궁에서 Pituitary Adenylate Cyclase-Activating Polypeptide와 수용체 유전자의 발현)

  • 이성호
    • Development and Reproduction
    • /
    • v.2 no.1
    • /
    • pp.21-27
    • /
    • 1998
  • The present study was performed to analyze the gene expressions of pituitary adenylate cyclase-activating polypeptide(PACAP) and its receptor in the rat uterus, a candidate for novel extrahypothalamic source and target. The PACAP cDNA fragments corresponding to the common exon region which is found in both the rat hypothalamus and testis were produced from all tissue samples including the rat uterus by reverse transcriptionpolymerase chain reaction (RT-PCR). No PCR product was amplified from the rat hypothalamic, pituitary, ovarian and uterine samples when the 5' primer corresponding to the testis-specific exon 1 region was used, while the predicted size of product was detected from the testis sample. RT-PCR using the uterine RNA and specific primers for the PACAP receptor yielded products with predicted sizes. Transcripts for the rat uterine PACAP receptor were identified as type I isoforms with hip-hop and hip- or hop-type inserts. After pregnant mare's serum gonadotropin (15 IU) treatment of immature rats (day 25), the level of PACAP mRNA was increased in 24 h and 48 h group, and was declined to the lowest in 72 h group. The present study shows the presence of transcripts for PACAP and its receptor isoform in the rat uterus. These finding ssuggest that the uterine PACAP ight act as a novel autocrine and/or paracrine factor via its specific receptors on the reglulation of rat uterine function and physiology during the reproductive cycle.

  • PDF

Pituitary Adenylate Cyclase-activating Polypeptide (PACAP) Treatment during Pre-maturation Increases the Maturation of Porcine Oocytes Derived from Small Follicles

  • Park, Kyu-Mi;So, Kyoung-Ha;Hyun, Sang-Hwan
    • Journal of Embryo Transfer
    • /
    • v.33 no.1
    • /
    • pp.1-11
    • /
    • 2018
  • Cellular cyclic adenosine-3' 5'-monophosphate (cAMP) modulator is known as meiotic inhibitor and can delays spontaneous maturation in IVM experiment. Among many cAMP modulators, the role of Pituitary adenylate cyclase activating polypeptide (PACAP) on IVM isn't known. The purpose of this study is to improve the maturation of oocytes derived from follicles ${\leq}3mm$ in diameter through PACAP as meiotic inhibitor during pre-in vitro maturation (pre-IVM). First, we checked PACAP and its receptors in cumulus cells and, to establish the optimal phase and concentration of PACAP for pre-IVM, we conducted chromatin configuration assessments. As a result, the rate of GV (Germinal Vesicle) according to duration of pre-IVM was significantly decreased 12 h and 18 h after IVM (87.1 and 84.1%, respectively) compared to 0 h (99.4%). When COC was cultured for 18 h, the GV rate in the $1{\mu}M$ of PACAP treatment group (82.1%) was significantly higher than any other PACAP treatment groups (60.5, 64.1, 74.4 and 69.9 %, respectively). So, we divided into four groups as follows; MF (the conventional IVM group, obtained from follicle from 3 to 6 mm in diameter), SF (the conventional IVM group, obtained from follicle ${\leq}3mm$ in diameter), Pre-SF(-)PACAP (IVM group including 18 h pre-IVM without $1{\mu}M$ of PACAP, obtained from follicle ${\leq}3mm$ in diameter) and Pre-SF(+)PACAP (IVM group including 18 h pre-IVM with $1{\mu}M$ of PACAP, obtained from follicle ${\leq}3mm$ in diameter). To examine the effect of PACAP during pre-IVM, we investigated analysis of nuclear maturation, intracellular glutathione (GSH) and reactive oxygen species (ROS) levels. In cumulus cells, PACAP receptors, ADCYAP1R1 and VIPR1 were detected but were not detected in oocytes. After IVM, the Pre-SF(+)PACAP had the highest Metaphase II rate (91.7%) among all groups (P<0.05). The GSH levels in the MF and Pre-SF(+)PACAP were significantly higher than in the other groups (P<0.05) and ROS levels was no significant difference among all groups. In conclusion, these results indicated that even though the oocytes were derived from SF, pre-IVM application of PACAP improved meiotic and cytoplasmic maturation by regulating intracellular oxidative stress.

TTF-1 Expression in PACAP-expressing Retinal Ganglion Cells

  • Son, Young June;Park, Jeong Woo;Lee, Byung Ju
    • Molecules and Cells
    • /
    • v.23 no.2
    • /
    • pp.215-219
    • /
    • 2007
  • In mammals light input resets the central clock of the suprachiasmatic nucleus by inducing secretion of pituitary adenylate cyclase-activating polypeptide (PACAP) from retinal ganglion cells (RGCs). We previously showed that thyroid transcription factor 1 (TTF-1), a homeodomain-containing transcription factor, specifically regulates PACAP gene expression in the rat hypothalamus. In the present study we examined the expression of TTF-1 in PACAP-synthesizing retinal cells. Fluorescence in situ hybridization (FISH) showed that it is abundantly expressed in RGCs of the superior region of the retina, but in only a small subset of RGCs in the inferior region. Double FISH experiments revealed that TTF-1 is exclusively expressed in PACAP-producing RGCs. These results suggest that TTF-1 plays a regulatory role in PACAP-expressing retinal ganglion cells.

Neuroprotective roles of pituitary adenylate cyclase-activating polypeptide in neurodegenerative diseases

  • Lee, Eun Hye;Seo, Su Ryeon
    • BMB Reports
    • /
    • v.47 no.7
    • /
    • pp.369-375
    • /
    • 2014
  • Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic bioactive peptide that was first isolated from an ovine hypothalamus in 1989. PACAP belongs to the secretin/glucagon/vasoactive intestinal polypeptide (VIP) superfamily. PACAP is widely distributed in the central and peripheral nervous systems and acts as a neurotransmitter, neuromodulator, and neurotrophic factor via three major receptors (PAC1, VPAC1, and VPAC2). Recent studies have shown a neuroprotective role of PACAP using in vitro and in vivo models. In this review, we briefly summarize the current findings on the neurotrophic and neuroprotective effects of PACAP in different brain injury models, such as cerebral ischemia, Parkinson's disease (PD), and Alzheimer's disease (AD). This review will provide information for the future development of therapeutic strategies in treatment of these neurodegenerative diseases.

Epac2 contributes to PACAP-induced astrocytic differentiation through calcium ion influx in neural precursor cells

  • Seo, Hyunhyo;Lee, Kyungmin
    • BMB Reports
    • /
    • v.49 no.2
    • /
    • pp.128-133
    • /
    • 2016
  • Astrocytes play a critical role in normal brain functions and maintaining the brain microenvironment, and defects in astrocytogenesis during neurodevelopment could give rise to severe mental illness and psychiatric disorders. During neuro-embryogenesis, astrocytogenesis involves astrocytic differentiation of neural precursor cells (NPCs) induced by signals from ciliary neurotrophic factor (CNTF) or pituitary adenylate cyclase-activating peptide (PACAP). However, in contrast to the CNTF signaling pathway, the exact mechanism underlying astrocytic differentiation induced by PACAP is unknown. In the present study, we aimed to verify a signaling pathway specific to PACAP-induced astrocytogenesis, using exchange protein directly activated by cAMP2 (Epac2)-knockout mice. We found that PACAP could trigger astrocytic differentiation of NPCs via Epac2 activation and an increase in the intracellular calcium concentration via a calcium ion influx. Taken together, we concluded that astrocytogenesis stimulated by PACAP occurs through a novel signaling pathway independent from CNTF-JAK/STAT signaling, that is the well-known pathway of astrocytogenesis.

Pituitary Adenylate Cyclase-activating Polypeptide Inhibits Pacemaker Activity of Colonic Interstitial Cells of Cajal

  • Wu, Mei Jin;Kee, Keun Hong;Na, Jisun;Kim, Seok Won;Bae, Youin;Shin, Dong Hoon;Choi, Seok;Jun, Jae Yeoul;Jeong, Han-Seong;Park, Jong-Seong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.19 no.5
    • /
    • pp.435-440
    • /
    • 2015
  • This study aimed to investigate the effect of pituitary adenylate cyclase-activating peptide (PACAP) on the pacemaker activity of interstitial cells of Cajal (ICC) in mouse colon and to identify the underlying mechanisms of PACAP action. Spontaneous pacemaker activity of colonic ICC and the effects of PACAP were studied using electrophysiological recordings. Exogenously applied PACAP induced hyperpolarization of the cell membrane and inhibited pacemaker frequency in a dose-dependent manner (from 0.1 nM to 100 nM). To investigate cyclic AMP (cAMP) involvement in the effects of PACAP on ICC, SQ-22536 (an inhibitor of adenylate cyclase) and cell-permeable 8-bromo-cAMP were used. SQ-22536 decreased the frequency of pacemaker potentials, and cell-permeable 8-bromo-cAMP increased the frequency of pacemaker potentials. The effects of SQ-22536 on pacemaker potential frequency and membrane hyperpolarization were rescued by co-treatment with glibenclamide (an ATP-sensitive $K^+$ channel blocker). However, neither $N^G$-nitro-L-arginine methyl ester (L-NAME, a competitive inhibitor of NO synthase) nor 1H-[1,2,4]oxadiazolo[4,3-${\alpha}$]quinoxalin-1-one (ODQ, an inhibitor of guanylate cyclase) had any effect on PACAP-induced activity. In conclusion, this study describes the effects of PACAP on ICC in the mouse colon. PACAP inhibited the pacemaker activity of ICC by acting through ATP-sensitive $K^+$ channels. These results provide evidence of a physiological role for PACAP in regulating gastrointestinal (GI) motility through the modulation of ICC activity.