Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hyuk Bang Kwon is active.

Publication


Featured researches published by Hyuk Bang Kwon.


Endocrinology | 2009

Molecular Evolution of Multiple Forms of Kisspeptins and GPR54 Receptors in Vertebrates

Yeo Reum Lee; Kenta Tsunekawa; Mi Jin Moon; Haet Nim Um; Jong-Ik Hwang; Tomohiro Osugi; Naohito Otaki; Yuya Sunakawa; Kyungjin Kim; Hubert Vaudry; Hyuk Bang Kwon; Jae Young Seong; Kazuyoshi Tsutsui

Kisspeptin and its receptor GPR54 play important roles in mammalian reproduction and cancer metastasis. Because the KiSS and GPR54 genes have been identified in a limited number of vertebrate species, mainly in mammals, the evolutionary history of these genes is poorly understood. In the present study, we have cloned multiple forms of kisspeptin and GPR54 cDNAs from a variety of vertebrate species. We found that fish have two forms of kisspeptin genes, KiSS-1 and KiSS-2, whereas Xenopus possesses three forms of kisspeptin genes, KiSS-1a, KiSS-1b, and KiSS-2. The nonmammalian KiSS-1 gene was found to be the ortholog of the mammalian KiSS-1 gene, whereas the KiSS-2 gene is a novel form, encoding a C-terminally amidated dodecapeptide in the Xenopus brain. This study is the first to identify a mature form of KiSS-2 product in the brain of any vertebrate. Likewise, fish possess two receptors, GPR54-1 and GPR54-2, whereas Xenopus carry three receptors, GPR54-1a, GPR54-1b, and GPR54-2. Sequence identity and genome synteny analyses indicate that Xenopus GPR54-1a is a human GPR54 ortholog, whereas Xenopus GPR54-1b is a fish GPR54-1 ortholog. Both kisspeptins and GPR54s were abundantly expressed in the Xenopus brain, notably in the hypothalamus, suggesting that these ligand-receptor pairs have neuroendocrine and neuromodulatory roles. Synthetic KiSS-1 and KiSS-2 peptides activated GPR54s expressed in CV-1 cells with different potencies, indicating differential ligand selectivity. These data shed new light on the molecular evolution of the kisspeptin-GPR54 system in vertebrates.


Journal of Biological Chemistry | 2008

Identification of Farnesyl Pyrophosphate and N-Arachidonylglycine as Endogenous Ligands for GPR92

Da Young Oh; Jung Min Yoon; Mi Jin Moon; Jong-Ik Hwang; Han Choe; Ju Yeon Lee; Jae Il Kim; Sunoh Kim; Hyewhon Rhim; David K. O'Dell; J. Michael Walker; Heung Sik Na; Min Goo Lee; Hyuk Bang Kwon; Kyungjin Kim; Jae Young Seong

A series of small compounds acting at the orphan G protein-coupled receptor GPR92 were screened using a signaling pathway-specific reporter assay system. Lipid-derived molecules including farnesyl pyrophosphate (FPP), N-arachidonylglycine (NAG), and lysophosphatidic acid were found to activate GPR92. FPP and lysophosphatidic acid were able to activate both Gq/11- and Gs-mediated signaling pathways, whereas NAG activated only the Gq/11-mediated signaling pathway. Computer-simulated modeling combined with site-directed mutagenesis of GPR92 indicated that Thr97, Gly98, Phe101, and Arg267 of GPR92 are responsible for the interaction of GPR92 with FPP and NAG. Reverse transcription-PCR analysis revealed that GPR92 mRNA is highly expressed in the dorsal root ganglia (DRG) but faint in other brain regions. Peripheral tissues including, spleen, stomach, small intestine, and kidney also expressed GPR92 mRNA. Immunohistochemical analysis revealed that GPR92 is largely co-localized with TRPV1, a nonspecific cation channel that responds to noxious heat, in mouse and human DRG. FPP and NAG increased intracellular Ca2+ levels in cultured DRG neurons. These results suggest that FPP and NAG play a role in the sensory nervous system through activation of GPR92.


Molecular and Cellular Endocrinology | 2004

Effect of ascorbic acid supplementation on testicular steroidogenesis and germ cell death in cadmium-treated male rats.

Ronojoy Sen Gupta; Jisun Kim; Cynthia Gomes; Sung-Dug Oh; Juran Park; Wook Bin Im; Jae Young Seong; Ryun Sup Ahn; Hyuk Bang Kwon; Jaemog Soh

Cadmium (Cd) is one of the environmental pollutants affecting various tissues and organs including testis. Harmful effect of Cd in testis is known to be germ cell degeneration and impairment of testicular steroidogenesis. Animals treated with high doses of Cd (0.2 and 0.3 mg/100g BW) showed a significant decrease in serum testosterone (T) level, but a significant induction of testicular lipid peroxidation levels. TUNEL assay showed that low doses of Cd (0.13 and 0.15 mg/100g BW) exhibited typical characteristics of apoptosis while high doses of Cd caused more necrosis than apoptosis. In contrast, supplementation with ascorbic acid reduced testicular lipid peroxidation levels. Ascorbic acid supplementation restored testicular 3beta-hydroxysteroiddehydrogenase (HSD) and 17beta-HSD enzyme activities, 3beta-HSD and cytochrome P450 side chain cleavage (P450(scc)) mRNA levels and serum T concentration to normal in Cd-administered rats. Moreover, administration of ascorbic acid prevented germ cell apoptosis as demonstrated by the reduced number of TUNEL-positive cells in germinal epithelium and inhibited Cd-induced necrosis. These results indicate that ascorbic acid have protective roles in vivo on the Cd-induced overall testicular damage including impaired steroidogenesis and germ cell death possibly through scavenging the reactive oxygen species generated by Cd administration.


Molecular and Cellular Endocrinology | 2000

Molecular cloning, distribution and pharmacological characterization of a novel gonadotropin-releasing hormone ([Trp8] GnRH) in frog brain☆

Myung Sik Yoo; Hae Mook Kang; Hueng Sik Choi; Jung Woo Kim; Brigitte E. Troskie; Robert P. Millar; Hyuk Bang Kwon

To date nine structural variants of GnRH have been identified in vertebrates and two additional forms have been isolated from a tunicate. In amphibians only mammalian GnRH ([Arg8] GnRH) and type II GnRH (chicken GnRH II, [His5, Trp7, Tyr8] GnRH) have been identified. In the present study, a full-length cDNA encoding a novel type of GnRH was isolated from pituitary of Rana dybowskii. The GnRH gene encodes a GnRH peptide ([Trp8] GnRH) in which tryptophan is substituted for arginine of mammalian GnRH Northern blot analysis revealed the presence of a single 500 bp transcript for the [Trp8] GnRH precursor in forebrain but its absence in testis, ovary, kidney and liver. Restriction digests of genomic DNA demonstrated a single copy of the gene. The [Trp8] GnRH immunoreactive cells were identified in the preoptic area of the frog brain. Synthetic [Trp8] GnRH was tested for its ability to stimulate inositol phosphate production by COS-1 cells transfected with the cloned Xenopus pituitary GnRH receptor and the cloned human GnRH receptor. [Trp8] GnRH had a potency of about 60% compared with mammalian GnRH ([Arg8] GnRH) for the Xenopus receptor, whereas the potency of [Trp8] GnRH was approximately 5% compared with mammalian GnRH for the human receptor. Both mammalian GnRH and [Trp8] GnRH were 1000-fold less potent than type II GnRH for the Xenopus GnRH receptor. The similar potency of [Arg8] GnRH and the novel [Trp8] GnRH for the Xenopus pituitary receptor indicates that, unlike the human receptor, the Xenopus receptor does not discriminate between these amino acids in position eight thereby allowing substitution of the arginine in the mammalian GnRH.


International Review of Cytology-a Survey of Cell Biology | 2006

Cellular and Molecular Biology of Orphan G Protein-Coupled Receptors

Da Young Oh; Kyungjin Kim; Hyuk Bang Kwon; Jae Young Seong

The superfamily of G protein-coupled receptors (GPCRs) is the largest and most diverse group of membrane-spanning proteins. It plays a variety of roles in pathophysiological processes by transmitting extracellular signals to cells via heterotrimeric G proteins. Completion of the human genome project revealed the presence of approximately 168 genes encoding established nonsensory GPCRs, as well as 207 genes predicted to encode novel GPCRs for which the natural ligands remained to be identified, the so-called orphan GPCRs. Eighty-six of these orphans have now been paired to novel or previously known molecules, and 121 remain to be deorphaned. A better understanding of the GPCR structures and classification; knowledge of the receptor activation mechanism, either dependent on or independent of an agonist; increased understanding of the control of GPCR-mediated signal transduction; and development of appropriate ligand screening systems may improve the probability of discovering novel ligands for the remaining orphan GPCRs.


Peptides | 2009

Molecular cloning of the bullfrog kisspeptin receptor GPR54 with high sensitivity to Xenopus kisspeptin

Jung Sun Moon; Yeo Reum Lee; Da Young Oh; Jong Ik Hwang; Ju Yeon Lee; Jae Il Kim; Hubert Vaudry; Hyuk Bang Kwon; Jae Young Seong

Kisspeptin and its receptor, GPR54, play important roles in mammalian reproduction and cancer development. However, little is known about their function in nonmammalian species. In the present study, we have isolated the cDNA encoding the kisspeptin receptor, GPR54, from the bullfrog, Rana catesbeiana. The bullfrog GPR54 (bfGPR54) cDNA encodes a 379-amino acid heptahelical G protein-coupled receptor. bfGPR54 exhibits 45-46% amino acid identity with mammalian GPR54s and 70-74% identity with fish GPR54s. RT-PCR analysis showed that bfGPR54 mRNA is highly expressed in the forebrain, hypothalamus and pituitary. Upon stimulation by synthetic human kisspeptin-10 with Phe-amide residue at the C-terminus (h-Kiss-10F), bfGPR54 induces SRE-luc activity, a PKC-specific reporter, evidencing the PKC-linked signaling pathway of bfGPR54. Using a blast search, we found a gene encoding a kisspeptin-like peptide in Xenopus. The C-terminal decapeptide of Xenopus kisspeptin shows higher amino acid sequence identity to fish Kiss-10s than mammalian Kiss-10s. A synthetic Xenopus kisspeptin peptide (x-Kiss-12Y) showed a higher potency than mammalian Kiss-10s in the activation of bfGPR54. This study expands our understanding of the physiological roles and molecular evolution of kisspeptins and their receptors.


Molecular Reproduction and Development | 1996

Differential expression of laminin chain-specific mRNA transcripts during mouse preimplantation embryo development

Chanseob Shim; Hyuk Bang Kwon; Kyungjin Kim

Laminin is the first extracellular matrix protein that has been shown to be synthesized in preimplantation mouse embryos. In the present study, chain‐specific expression patterns of laminin mRNAs were examined by semiquantitative reverse transcription‐polymerase chain reaction (RT‐PCR). During preimplantation mouse embryo development, temporal expression patterns of laminin chain mRNAs were somewhat differential: B1 chain mRNA was first detectable at the late two‐cell stage and its level was gradually increased by the blastocyst stage. In contrast, B2 and A chain mRNAs first appeared at the morula and blastocyst stages, respectively. At the blastocyst stage, all of the laminin chain mRNAs were highly detected compared to the earlier stages. When embryos were flushed at the morula stage and cultured in vitro, all laminin chain mRNA levels were decreased or not changed in the process of blastocoele expansion. In contrast, in the in vivo condition where embryos at different stages of blastocyst were flushed at different time points, laminin chain mRNA levels were increased as a function of blastocoele expansion. These changes in laminin mRNAs were parallel with its receptors such as integrin α3 and α6. 3‐Isobutyl‐1‐methylxanthine (IBMX), which is known to be a potent activator of blastocoele expansion and regulates cAMP metabolism, upregulated laminin expression (except B1 chain) in blastocysts cultured in vitro. In vitro cultured embryos normally developed up to the late blastocyst, although their development was delayed compared with the in vivo condition where laminin gene expression was gradually increased as the blastocoele expanded. These results indicate that laminin expression may not be involved directly in the regulation of blastocoele expansion. The uterine environment enclosing the preimplantation embryos appears, therefore, to play an important role in the regulation of laminin gene expression during blastocyst development.


Cancer Research | 2009

A gonadotropin-releasing hormone-II antagonist induces autophagy of prostate cancer cells.

Dong-Ki Kim; Ji Sook Yang; Kaushik Maiti; Jong-Ik Hwang; Kyungjin Kim; Dong-Seung Seen; Younghee Ahn; Cheolju Lee; Byeong-Cheol Kang; Hyuk Bang Kwon; Jun Cheon; Jae Young Seong

Gonadotropin-releasing hormone-I (GnRH-I) is known to directly regulate prostate cancer cell proliferation. However, the role of GnRH-II in prostate cancer is unclear. Here, we investigated the effect of the GnRH-II antagonist trptorelix-1 (Trp-1) on growth of PC3 prostate cancer cells. Trp-1 induced growth inhibition of PC3 cells in vitro and inhibited growth of PC3 cells xenografted into nude mice. FITC-N3, an FITC-conjugated Trp-1 analogue, was largely present in the mitochondria of prostate cancer cells, but not in other cells that are not derived from the prostate. Trp-1-induced PC3 growth inhibition was associated with decreased mitochondrial membrane potential and increased levels of mitochondrial and cytosolic reactive oxygen species (ROS). Growth inhibition was partially prevented by cotreating cells with N-acetyl cysteine, an antioxidant. Cytochrome c release and caspase-3 activation were not detected in Trp-1-treated cells. However, Trp-1 induced autophagosome formation, as seen by increased LysoTracker staining and recruitment of microtubule-associated protein 1 light chain 3 to these new lysosomal compartments. Trp-1-induced autophagy was accompanied by decreased AKT phosphorylation and increased c-Jun NH(2) terminal kinase phosphorylation, two events known to be linked to autophagy. Taken together, these data suggest that Trp-1 directly induces mitochondrial dysfunction and ROS increase, leading to autophagy of prostate cancer cells. GnRH-II antagonists may hold promise in the treatment of prostate cancer.


Molecular and Cellular Endocrinology | 2003

Differential G protein coupling preference of mammalian and nonmammalian gonadotropin-releasing hormone receptors.

Da Young Oh; Li Wang; Ryun Sup Ahn; Jae Yong Park; Jae Young Seong; Hyuk Bang Kwon

Recently, we have identified three distinct types of gonadotropin-releasing hormone receptor (GnRHR) in the bullfrog (designated bfGnRHR-1, bfGnRHR-2, and bfGnRHR-3). In the present study, we compared G protein coupling preference of mammalian and nonmammalian GnRHRs. In a transient expression system, stimulation of either bfGnRHRs or rat GnRHR by GnRH significantly increased both inositol phosphates (IP) and cAMP productions, but ratios of IP to cAMP induction levels were quite different among the receptors, indicating differential G protein coupling preference. Using cAMP-dependent protein kinase A (PKA)-specific (CRE-luc) or protein kinase C (PKC)-specific reporter (c-fos-luc) systems, we further examined G(s) and G(q/11) coupling preference of these GnRHRs. Since activities of CRE-luc and c-fos-luc were highly dependent on cell types, GnRH-induced CRE-luc or c-fos-luc activity was normalized by forskolin-induced CRE-luc or 12-O-tetradecanoylphenol-13-acetate (TPA)-induced c-fos-luc activity, respectively. This normalized result indicated that bfGnRHR-2 couples to G(s) more actively than G(q/11), while bfGnRHR-1 and -3 couple to G(s) and G(q/11) with similar strength. However, the rat GnRHR appeared to couple to G(q/11) more efficiently than G(s). This study was further confirmed by an experiment in which GnRH augmented CRE-driven luciferase activity in alphaT3-1 cells when CRE-luc was cotransfected with bfGnRHRs but not with vehicle or rat GnRHR. Collectively, these results indicate that mammalian and nonmammalian GnRHRs may induce diverse cellular and physiological responses through differential activation of PKA and PKC signaling pathways.


Molecular Brain Research | 1998

Differential regulation of gonadotropin-releasing hormone (GnRH) receptor expression in the posterior mediobasal hypothalamus by steroid hormones : implication of GnRH neuronal activity

Jae Young Seong; Sang Soo Kang; Kyungyoon Kam; Young Goo Han; Hyuk Bang Kwon; Kyungza Ryu; Kyungjin Kim

The present study is designed to evaluate the relationship between gonadotropin-releasing hormone (GnRH) and GnRH receptor (GnRHR) gene expression during the steroid-induced LH surge. One week after ovariectomy (OVX), a capsule containing 17beta-estradiol (E) or vehicle (V) was implanted into OVX rats, and 2 days later a single injection of progesterone (P) or V was administered s.c. at 10:00 h. Poly(A)-rich RNA samples were isolated from the micropunches of the preoptic area (POA) and the posterior mediobasal hypothalamus (pMBH) from both sides of individual brain slices. Using competitive reverse transcription-polymerase chain reaction (RT-PCR) procedures, three parameters (POA GnRH, pMBH GnRHR) and pituitary GnRHR mRNA levels were simultaneously determined in each individual animal. POA GnRH mRNA and pituitary GnRHR mRNA levels were decreased by treatment with E, but increased by a combination of E and P. In contrast, pMBH GnRHR mRNA levels were clearly augmented by treatment with E, and decreased by the combination of E and P. Temporal changes in such parameters were determined in OVX+E+V- and OVX+E+P-treated rats. P augmented POA GnRH mRNA levels at the time of the LH surge (17:00 h) and the increased GnRH mRNA levels were remained until 22:00 h, while E alone failed to alter POA GnRH mRNA levels. In the pMBH micropunch samples, P substantially decreased E-induced increase in GnRHR mRNA levels at 17:00 h and further lowered those until 22:00 h. Antisense oligonucleotides of GnRHR mRNA administered into the lateral ventricle of OVX+E-treated rats blocked the E-induced increase in pMBH GnRHR mRNA levels. The antisense oligonucleotides also prevented the LH surge as well as the increase in pituitary GnRHR mRNA levels in the OVX+E+P-treated group. However, administration of this antisense oligonucleotides failed to alter POA GnRH mRNA levels. In conclusion, the present study demonstrated that there is an inverse relationship between POA GnRH mRNA levels and pMBH GnRHR mRNA levels in response to E and/or P, and that the blockade of the E-induced increase in pMBH GnRHR mRNA levels effectively nullified the P-induced LH surge. These results indicate that pMBH GnRHR gene expression is involved in synchronizing the GnRH neuronal activity, which is crucial for the generation of the LH surge.

Collaboration


Dive into the Hyuk Bang Kwon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kyungjin Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ryun Sup Ahn

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Da Young Oh

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Kaushik Maiti

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Wook-Bin Im

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Hua Li

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Sujata Acharjee

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Hueng Sik Choi

Chonnam National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge