Network


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

Hotspot


Dive into the research topics where Kyung Bon Lee is active.

Publication


Featured researches published by Kyung Bon Lee.


Biology of Reproduction | 2008

Identification of Novel Bovine Cumulus Cell Molecular Markers Predictive of Oocyte Competence: Functional and Diagnostic Implications

Anilkumar Bettegowda; Osman V. Patel; Kyung Bon Lee; Ki Eun Park; Mohamed Salem; Jianbo Yao; James J. Ireland; George W. Smith

Abstract The present study was undertaken to discover molecular markers in bovine cumulus cells predictive of oocyte competence and to elucidate their functional significance. Differences in RNA transcript abundance in cumulus cells harvested from oocytes of adult versus prepubertal animals (a model of poor oocyte quality) were identified by microarray analysis. Four genes of interest encoding for the lysosomal cysteine proteinases cathepsins B, S, K, and Z and displaying greater transcript abundance in cumulus cells surrounding oocytes harvested from prepubertal animals were chosen for further investigation. Greater mRNA abundance for such genes in cumulus cells of prepubertal oocytes was confirmed by real-time RT-PCR. Elevated transcript abundance for cathepsins B, S, and Z also was observed in cumulus cells surrounding adult metaphase II oocytes that developed to the blastocyst stage at a low percentage following parthenogenetic activation versus those that developed at a high percentage. Functional significance of cumulus cell cathepsin expression to oocyte competence was confirmed by treatment of cumulus-oocyte complexes during in vitro oocyte maturation with a cell-permeable cysteine proteinase (cathepsin) inhibitor. Inhibitor treatment decreased apoptotic nuclei in the cumulus layer and enhanced development of parthenogenetically activated and in vitro-fertilized adult oocytes to the blastocyst stage. Stimulatory effects of inhibitor treatment during meiotic maturation on subsequent embryonic development were not observed when oocytes were matured in the absence of cumulus cells. The present results support a functional role for cumulus cell cathepsins in compromised oocyte competence and suggest that cumulus cell cathepsin mRNA abundance may be predictive of oocyte quality.


Endocrinology | 2009

Molecular determinants of oocyte competence: potential functional role for maternal (oocyte-derived) follistatin in promoting bovine early embryogenesis.

Kyung Bon Lee; Anilkumar Bettegowda; Gabbine Wee; James J. Ireland; George W. Smith

Previous studies established a positive relationship between oocyte competence and follistatin mRNA abundance. Herein, we used the bovine model to test the hypothesis that follistatin plays a functional role in regulation of early embryogenesis. Treatment of early embryos with follistatin during in vitro culture (before embryonic genome activation) resulted in a dose-dependent decrease in time to first cleavage, increased numbers of blastocysts, and increased blastocyst total and trophectoderm cell numbers. To determine the requirement of endogenous follistatin for early embryogenesis, follistatin ablation/replacement studies were performed. Microinjection of follistatin small interfering RNA into zygotes reduced follistatin mRNA and protein and was accompanied by a reduction in number of embryos developing to eight- to 16-cell and blastocyst stages and reduced blastocyst total and trophectoderm cell numbers. Effects of follistatin ablation were rescued by culture of follistatin small interfering RNA-injected embryos in the presence of exogenous follistatin. To investigate whether follistatin regulation of early embryogenesis is potentially mediated via inhibition of endogenous activin activity, the effects of treatment of embryos with exogenous activin, SB-431542 (inhibitor of activin, TGF-beta, and nodal type I receptor signaling) and follistatin plus SB-431542 were investigated. Activin treatment mimicked positive effects of follistatin on time to first cleavage and blastocyst development, whereas negative effects of SB-431542 treatment were observed. Stimulatory effects of follistatin on embryogenesis were not blocked by SB-431542 treatment. Results support a functional role for oocyte-derived follistatin in bovine early embryogenesis and suggest that observed effects of follistatin are likely not mediated by classical inhibition of activin activity.


Biology of Reproduction | 2009

Role of Importin Alpha8, a New Member of the Importin Alpha Family of Nuclear Transport Proteins, in Early Embryonic Development in Cattle

Jyothsna Tejomurtula; Kyung Bon Lee; Swamy K. Tripurani; George W. Smith; Jianbo Yao

Abstract Nuclear proteins such as transcription and chromatin remodeling factors are required for initiation of transcription in early embryos before embryonic genome activation. The nuclear transport of these proteins is mediated by transport factors such as importins. Through analysis of expressed sequence tags from a bovine oocyte cDNA library, we identified a new member of the importin alpha family (named importin alpha8). The cloned cDNA for bovine importin alpha8 (KPNA7) is 1817 base pair in length, encoding a protein of 522 amino acids that contains a conserved importin beta-binding domain and seven armadillo motifs. The RT-PCR analysis revealed that KPNA7 mRNA is specifically expressed in ovaries and mature oocytes. Real-time PCR demonstrated that KPNA7 expression in germinal vesicle (GV) oocytes is 33 to 2396 times higher than that of other importin alpha genes and that KPNA7 mRNA is abundant in GV and metaphase II oocytes, as well as in early-stage embryos collected before embryonic genome activation, but is barely detectable in morula- and blastocyst-stage embryos. Similarly, expression of KPNA7 protein is very high in oocytes and early embryos but is low in blastocysts. A glutathione S-transferase pull-down assay revealed that KPNA7 has a strong binding affinity for the nuclear protein nucleoplasmin 2 relative to that of other importin alphas. RNA interference experiments demonstrated that knockdown of KPNA7 in early embryos results in a decreased proportion of embryos developing to the blastocyst stage. These results suggest that KPNA7 may have an important role in the transport of essential nuclear proteins required for early embryogenesis.


Reproduction, Fertility and Development | 2008

Cytoplasmic and nuclear determinants of the maternal-to-embryonic transition

Anilkumar Bettegowda; Kyung Bon Lee; George W. Smith

Although improvements in culture systems have greatly enhanced in vitro embryo production, success rates under the best conditions are still far from ideal. The reasons for developmental arrest of the majority of in vitro produced embryos are unclear, but likely attributable, in part, to intrinsic and extrinsic influences on the cytoplasmic and/or nuclear environment of an oocyte and/or early embryo that impede normal progression through the maternal-to-embryonic transition. The maternal-to-embryonic transition is the time period during embryonic development spanning from fertilisation until when control of early embryogenesis changes from regulation by oocyte-derived factors to regulation by products of the embryonic genome. The products of numerous maternal effect genes transcribed and stored during oogenesis mediate this transition. Marked epigenetic changes to chromatin during this window of development significantly modulate embryonic gene expression. Depletion of maternal mRNA pools is also an obligatory event during the maternal-to-embryonic transition critical to subsequent development. An increased knowledge of the fundamental mechanisms and mediators of the maternal-to-embryonic transition is foundational to understanding the regulation of oocyte quality and future breakthroughs relevant to embryo production.


BMC Developmental Biology | 2011

MicroRNA-196a regulates bovine newborn ovary homeobox gene (NOBOX) expression during early embryogenesis

Swamy K. Tripurani; Kyung Bon Lee; Gabbine Wee; George W. Smith; Jianbo Yao

BackgroundOocyte-derived maternal RNAs drive early embryogenesis when the newly formed embryo is transcriptionally inactive. Recent studies in zebrafish have identified the role of microRNAs during the maternal-to-embryonic transition (MET). MicroRNAs are short RNAs that bind to the 3 UTR of target mRNAs to repress their translation and accelerate their decay. Newborn ovary homeobox gene (NOBOX) is a transcription factor that is preferentially expressed in oocytes and essential for folliculogenesis in mice. NOBOX knockout mice are infertile and lack of NOBOX disrupts expression of many germ-cell specific genes and microRNAs. We recently reported the cloning and expression of bovine NOBOX during early embryonic development and our gene knockdown studies indicate that NOBOX is a maternal effect gene essential for early embryonic development. As NOBOX is a maternal transcript critical for development and NOBOX is depleted during early embryogenesis, we hypothesized that NOBOX is targeted by microRNAs for silencing and/or degradation.ResultsUsing an algorithm MicroInspector, a potential microRNA recognition element (MRE) for miR-196a was identified in the 3 UTR of the bovine NOBOX mRNA. Expression analysis of miR-196a in bovine oocytes and during early embryonic development indicated that it is expressed both in oocytes and embryos and tends to increase at the four-cell and eight-cell stages. Ectopic expression of NOBOX and miR-196a in HeLa cells inhibited the expression of NOBOX protein compared to the control cells without miR-196a. Similarly, the activity of a luciferase construct containing the entire 3 UTR of bovine NOBOX was suppressed, and the regulation was abolished by mutations in the miR-196a binding site indicating that the predicted MRE is critical for the direct and specific binding of miR-196a to the NOBOX mRNA. Furthermore, ectopic expression of miR-196a mimic in bovine early embryos significantly reduced the NOBOX expression at the both mRNA and protein levels.ConclusionCollectively, our results demonstrate that miR-196a is a bona fide negative regulator of NOBOX during bovine early embryogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2007

JY-1, an oocyte-specific gene, regulates granulosa cell function and early embryonic development in cattle

Anilkumar Bettegowda; Jianbo Yao; Aritro Sen; Qinglei Li; Kyung Bon Lee; Yasuhiro Kobayashi; Osman V. Patel; Paul M. Coussens; James J. Ireland; George W. Smith

Oocyte-specific gene products play a key role in regulation of fertility in mammals. Here, we describe the discovery, molecular characterization, and function of JY-1, a bovine oocyte-expressed gene shown to regulate both function of ovarian granulosa cells and early embryogenesis in cattle and characteristics of JY-1 loci in other species. The JY-1 gene encodes for a secreted protein with multiple mRNA transcripts containing an identical ORF but differing lengths of 3′ UTR. JY-1 mRNA and protein are oocyte-specific and detectable throughout folliculogenesis. Recombinant JY-1 protein regulates function of follicle-stimulating hormone-treated ovarian granulosa cells, resulting in enhanced progesterone synthesis accompanied by reduced cell numbers and estradiol production. JY-1 mRNA of maternal origin is also present in early bovine embryos, temporally regulated during the window from meiotic maturation through embryonic genome activation, and is required for blastocyst development. The JY-1 gene has three exons and is located on bovine chromosome 29. JY-1-like sequences are present on syntenic chromosomes of other vertebrate species, but lack exons 1 and 2, including the protein-coding region, suggestive of species specificity in evolution and function of this oocyte-specific gene.


Endocrinology | 2011

A Novel Functional Role for the Oocyte-Specific Transcription Factor Newborn Ovary Homeobox (NOBOX) during Early Embryonic Development in Cattle

Swamy K. Tripurani; Kyung Bon Lee; Lei Wang; Gabbine Wee; George W. Smith; Young S. Lee; Keith E. Latham; Jianbo Yao

Newborn ovary homeobox (NOBOX) is an oocyte-specific transcription factor essential for folliculogenesis and expression of many germ cell-specific genes in mice. Here we report the characterization of the bovine NOBOX gene and its role in early embryogenesis. The cloned cDNA for bovine NOBOX contains an open reading frame encoding a protein of 500 amino acids with a conserved homeodomain. mRNA for NOBOX is preferentially expressed in ovaries and undetectable by RT-PCR in somatic tissues examined. NOBOX protein is present in oocytes throughout folliculogenesis. NOBOX is expressed in a stage-specific manner during oocyte maturation and early embryonic development and of maternal origin. Knockdown of NOBOX in early embryos using small interfering RNA demonstrated that NOBOX is required for embryonic development to the blastocyst stage. Depletion of NOBOX in early embryos caused significant down-regulation of genes associated with transcriptional regulation, signal transduction, and cell cycle regulation during embryonic genome activation. In addition, NOBOX depletion in early embryos reduced expression of pluripotency genes (POU5F1/OCT4 and NANOG) and number of inner cell mass cells in embryos that reached the blastocyst stage. This study demonstrates that NOBOX is an essential maternal-derived transcription factor during bovine early embryogenesis, which functions in regulation of embryonic genome activation, pluripotency gene expression, and blastocyst cell allocation.


Biology of Reproduction | 2009

Evidence Supporting a Role for Cocaine- and Amphetamine-Regulated Transcript (CARTPT) in Control of Granulosa Cell Estradiol Production Associated with Dominant Follicle Selection in Cattle

Lihua Lv; Fermin Jimenez-Krassel; Aritro Sen; Anilkumar Bettegowda; Mohan Mondal; Joseph K. Folger; Kyung Bon Lee; James J. Ireland; George W. Smith

We demonstrated previously a negative association of granulosa cell cocaine- and amphetamine-regulated transcript (CARTPT) expression with follicle health status and inhibitory effects of the mature CARTPT peptide (CART) on follicle-stimulating hormone (FSH) signal transduction in vitro, resulting in reduced bovine granulosa cell CYP19A1 mRNA and estradiol production. The objectives of this study were to investigate temporal regulation of granulosa cell CARTPT expression (granulosa cell mRNA and follicular fluid CART peptide concentrations) during follicular waves, CART regulation of androstenedione production (precursor for estradiol biosynthesis) by thecal tissue collected at specific stages of a follicular wave, FSH regulation of granulosa cell CARTPT mRNA expression, and the ability of CART to inhibit granulosa cell estradiol production and CYP19A1 mRNA expression when administered in vivo. CART concentrations in healthy, estrogen-active follicles (estradiol greater than progesterone in follicular fluid) decreased after dominant follicle selection, and CARTPT mRNA was lower in healthy, estrogen-active versus estrogen-inactive atretic follicles (progesterone greater than estradiol) collected at the predeviation and early dominance stages. CART treatment reduced luteinizing hormone-induced androstenedione production by thecal tissue collected at predeviation and early dominance stages but not at later stages of a follicular wave. The FSH or insulin-like growth factor 1 treatment in vitro reduced granulosa cell CARTPT mRNA in a dose-dependent fashion. Administration of CART in vivo into follicles at the early dominance stage reduced follicular fluid estradiol concentrations and granulosa cell CYP19A1 mRNA. Collectively, results support a potential stage-specific regulatory role for CART in negative regulation of estradiol production associated with selection of the dominant follicle.


Biology of Reproduction | 2015

Evidence Supporting a Role for SMAD2/3 in Bovine Early Embryonic Development: Potential Implications for Embryotropic Actions of Follistatin

Kun Zhang; Sandeep K. Rajput; Kyung Bon Lee; Dongliang Wang; Juncheng Huang; Joseph K. Folger; Jason G. Knott; Jiuzhen Zhang; George W. Smith

ABSTRACT The TGF-beta-SMAD signaling pathway is involved in regulation of various aspects of female reproduction. However, the intrinsic functional role of SMADs in early embryogenesis remains poorly understood. Previously, we demonstrated that treatment with follistatin, an activin (TGF-beta superfamily ligand)-binding protein, is beneficial for bovine early embryogenesis and specific embryotropic actions of follistatin are dependent on SMAD4. Because SMAD4 is a common SMAD that can bind both SMAD2/3 and SMAD1/5, the objective of this study was to further determine the intrinsic role of SMAD2/3 in the control of early embryogenesis and delineate if embryotropic actions of follistatin in early embryos are SMAD2/3 dependent. By using a combination of pharmacological and small interfering RNA-mediated inhibition of SMAD2/3 signaling in the presence or absence of follistatin treatment, our results indicate that SMAD2 and SMAD3 are both required for bovine early embryonic development and stimulatory actions of follistatin on 8- to 16-cell and that blastocyst rates, but not early cleavage, are muted when SMAD2/3 signaling is inhibited. SMAD2 deficiency also results in reduced expression of the bovine trophectoderm cell-specific gene CTGF. In conclusion, the present work provides evidence supporting a functional role of SMAD2/3 in bovine early embryogenesis and that specific stimulatory actions of follistatin are not observed in the absence of SMAD2/3 signaling.


Biology of Reproduction | 2014

Evidence Supporting a Functional Requirement of SMAD4 for Bovine Preimplantation Embryonic Development: A Potential Link to Embryotrophic Actions of Follistatin

Kyung Bon Lee; Kun Zhang; Joseph K. Folger; Jason G. Knott; George W. Smith

ABSTRACT Transforming growth factor beta (TGFbeta) superfamily signaling controls various aspects of female fertility. However, the functional roles of the TGFbeta-superfamily cognate signal transduction pathway components (e.g., SMAD2/3, SMAD4, SMAD1/5/8) in early embryonic development are not completely understood. We have previously demonstrated pronounced embryotrophic actions of the TGFbeta superfamily member-binding protein, follistatin, on oocyte competence in cattle. Given that SMAD4 is a common SMAD required for both SMAD2/3- and SMAD1/5/8-signaling pathways, the objectives of the present studies were to determine the temporal expression and functional role of SMAD4 in bovine early embryogenesis and whether embryotrophic actions of follistatin are SMAD4 dependent. SMAD4 mRNA is increased in bovine oocytes during meiotic maturation, is maximal in 2-cell stage embryos, remains elevated through the 8-cell stage, and is decreased and remains low through the blastocyst stage. Ablation of SMAD4 via small interfering RNA microinjection of zygotes reduced proportions of embryos cleaving early and development to the 8- to 16-cell and blastocyst stages. Stimulatory effects of follistatin on early cleavage, but not on development to 8- to 16-cell and blastocyst stages, were observed in SMAD4-depleted embryos. Therefore, results suggest SMAD4 is obligatory for early embryonic development in cattle, and embryotrophic actions of follistatin on development to 8- to 16-cell and blastocyst stages are SMAD4 dependent.

Collaboration


Dive into the Kyung Bon Lee's collaboration.

Top Co-Authors

Avatar

George W. Smith

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianbo Yao

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Gabbine Wee

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lihua Lv

Shanxi Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Aritro Sen

University of Rochester

View shared research outputs
Top Co-Authors

Avatar

Jason G. Knott

Michigan State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge