Network


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

Hotspot


Dive into the research topics where Keesook Lee is active.

Publication


Featured researches published by Keesook Lee.


Molecular and Cellular Biology | 2004

Molecular Mechanism of Suppression of Testicular Steroidogenesis by Proinflammatory Cytokine Tumor Necrosis Factor Alpha

Cheol Yi Hong; Jin Hee Park; Ryun Seop Ahn; Suhn Young Im; Hueng-Sik Choi; Jaemog Soh; Synthia H. Mellon; Keesook Lee

ABSTRACT Tumor necrosis factor alpha (TNF-α) has been demonstrated to inhibit steroidogenesis in Leydig cells at the transcriptional level of steroidogenic enzymes. However, the molecular mechanism of this observed gene repression is not well understood. We now demonstrate that nuclear factor κB (NF-κB) activated by TNF-α inhibits the transactivation of orphan nuclear receptors, which regulate the expression of steroidogenic-enzyme genes. TNF-α treatment suppressed the luteinizing-hormone-induced or Nur77/SF-1-stimulated promoter activity of steroidogenic-enzyme genes in Leydig cells. The TNF-α-mediated gene suppression was blocked by treatment with an inhibitor of NF-κB. In addition, overexpression of the p65 (RelA) subunit of NF-κB showed the same effect as TNF-α and inhibited Nur77 transactivation, suggesting the involvement of NF-κB activation in the observed gene repression. Physical association of Nur77 with p65 was revealed by mammalian two-hybrid, GST pull-down, and coimmunoprecipitation analyses. The NF-κB inhibition of Nur77 transactivation was likely due to the competition of p65 for Nur77 binding with coactivators. Finally, chromatin immunoprecipitation assays revealed that TNF-α treatment caused the recruitment of NF-κB to the promoter of the steroidogenic-enzyme P450c17 gene, supporting the hypothesis that the TNF-α-mediated gene repression involves NF-κB inhibition of the transcriptional activity of Nur77 and other orphan nuclear receptors. These findings provide a molecular mechanism underlying the inhibition of testicular steroidogenesis by proinflammatory cytokines.


Molecular and Cellular Biology | 2001

Srg3, a Mouse Homolog of Yeast SWI3, Is Essential for Early Embryogenesis and Involved in Brain Development

Joong K. Kim; Sung-Oh Huh; Heonsik Choi; Keesook Lee; Dongho Shin; Changjin Lee; Ju-Suk Nam; Hyun Soo Kim; Heekyoung Chung; Han W. Lee; Sang D. Park; Rho Hyun Seong

ABSTRACT Srg3 (SWI3-related gene product) is a mouse homolog of yeast SWI3,Drosophila melanogaster MOIRA (also named MOR/BAP155), and human BAF155 and is known as a core subunit of SWI/SNF complex. This complex is involved in the chromatin remodeling required for the regulation of transcriptional processes associated with development, cellular differentiation, and proliferation. We generated mice with a null mutation in theSrg3 locus to examine its function in vivo. Homozygous mutants develop in the early implantation stage but undergo rapid degeneration thereafter. An in vitro outgrowth study revealed that mutant blastocysts hatch, adhere, and form a layer of trophoblast giant cells, but the inner cell mass degenerates after prolonged culture. Interestingly, about 20% of heterozygous mutant embryos display defects in brain development with abnormal organization of the brain, a condition known as exencephaly. Histological examination suggests that exencephaly is caused by the failure in neural fold elevation, resulting in severe brain malformation. Our findings demonstrate that Srg3 is essential for early embryogenesis and plays an important role in the brain development of mice.


Nature Genetics | 1999

A double-stranded RNA binding protein required for activation of repressed messages in mammalian germ cells.

Jun Zhong; Antoine H. F. M. Peters; Keesook Lee; Robert E. Braun

Chromatin packaging in mammalian spermatozoa requires an ordered replacement of the somatic histones by two classes of spermatid-specific basic proteins, the transition proteins and the protamines. Temporal expression of transition proteins and protamines during spermatid differentiation is under translational control, and premature translation of protamine 1 leads to precocious nuclear condensation and sterility. We have previously suggested that the double-stranded (ds) RNA binding protein Prbp (encoded by the gene Tarbp2) functions as a translational regulator during mouse spermatogenesis. Here we show that Prbp is required for proper translational activation of the mRNAs encoding the protamines. We generated mice that carry a targeted disruption of Tarbp2 and determined that they were sterile and severely oligospermic. Using immunohistological analysis, we determined that the endogenous Prm2 mRNA and a reporter mRNA carrying protamine 1 translational-control elements were translated in a mosaic pattern. We showed that failure to synthesize the protamines resulted in delayed replacement of the transition proteins and subsequent failure of spermiation. The timing of Prbp expression suggests that it may function as a chaperone in the assembly of specific translationally regulated ribonucleoprotein particles.


Molecular and Cellular Biology | 2000

Characterization and targeted disruption of murine Nup50, a p27(Kip1)-interacting component of the nuclear pore complex.

Matthew Smitherman; Keesook Lee; Jherek Swanger; Raj P. Kapur; Bruce E. Clurman

ABSTRACT p27Kip1 is a member of the Cip-Kip family of cyclin-dependent kinase (Cdk) inhibitors that binds to cyclin-Cdk complexes and inhibits their catalytic activity in response to antiproliferative stimuli. p27Kip1 is regulated by several posttranscriptional mechanisms, including subcellular localization. We have identified a component of the nuclear pore complex (NPC), termed Nup50, through its two-hybrid interactions with p27Kip1. Nup50 is a nucleoplasmically oriented component of the nuclear pore complex with a role in protein export (T. Guan, R. H. Kehlenbach, E. C. Schirmer, A. Kehlenbach, F. Fan, B. E. Clurman, N. Arnheim, and L. Gerace, Mol. Cell. Biol. 20:5619–5630, 2000). We found that murine Nup50 is a widely expressed nucleoporin and that Nup50 expression is highest in the developing neural tube and adult testes. We have also examined interactions between Nup50 and the NPC and found specific two-hybrid interactions between Nup50 and several well-defined components of the NPC, as well as coimmunoprecipitation of Nup50 with the nucleoporin Nup153 from transfected mammalian cells. In order to study Nup50 function in vivo, we cloned the mouse Nup50 genomic locus and created a targeted Nup50 deletion in the mouse germ line. Nup50 disruption resulted in a complex phenotype characterized by late embryonic lethality, neural tube defects, and intrauterine growth retardation. Although Nup50-null mouse embryo fibroblasts exhibited no defects in either cell cycle control or p27Kip1 regulation, Nup50 deletion was associated with abnormalities in p27Kip1expression and cell proliferation in the developing neuroepithelium. We conclude that Nup50 is a nucleoporin with essential functions during mouse development.


Molecular and Cellular Biology | 2003

Expression of MIS in the Testis Is Downregulated by Tumor Necrosis Factor Alpha through the Negative Regulation of SF-1 Transactivation by NF-κB

Cheol Yi Hong; Jin Hee Park; Kook Heon Seo; Jin-Man Kim; Suhn Young Im; Jae Woon Lee; Hueng Sik Choi; Keesook Lee

ABSTRACT The expression of Mullerian inhibiting substance (MIS), a key molecule in sex differentiation and reproduction, is tightly regulated. It has been suggested that meiotic germ cells repress MIS expression in testicular Sertoli cells, although the substance responsible for this cell-cell communication remains unknown. Here, we present the cytokine tumor necrosis factor alpha (TNF-α) as a strong candidate for such a substance and its downstream molecular events. TNF-α inhibited MIS expression in testis organ cultures, and TNF-α−/− testes showed high and prolonged MIS expression. Furthermore, in transient-transfection assays TNF-α suppressed the MIS promoter that was activated by steroidogenic factor 1 (SF-1), one of the major transcription factors that regulate MIS expression. The modulation of SF-1 transactivation by TNF-α is through the activation of NF-κB, which subsequently interacts with SF-1 and represses its transactivation. The physical association of NF-κB with SF-1 was shown by yeast two-hybrid protein interaction, glutathione S-transferase pull-down, and coimmunoprecipitation (ChIP) analyses. ChIP assays also revealed that endogenous NF-κB, as well as SF-1, is recruited to the MIS promoter upon TNF-α signaling. SF-1-bound NF-κB subsequently recruits histone deacetylases to inhibit the SF-1-activated gene expression. These results may identify, for the first time, the responsible substance and its action mechanism underlying the repression of MIS expression by meiotic germ cells in the testis.


Biochemical and Biophysical Research Communications | 2002

Differential expression of Prx I and II in mouse testis and their up-regulation by radiation.

Keesook Lee; Yun-Jeong Kim; Yong Soo Lee; Tae Sook Hwang; Dae-Joong Kim; Eun-Mi Park; Young-Mee Park

Testis is one of the most sensitive organs to ionizing radiation. The present study was designed to unravel the possible role of antioxidant proteins, peroxiredoxin I and II (Prx I and II) in the testis. Our results show that Prx I and II are constitutively expressed in the testis and their expression levels are decreased to some extent as the testis develops. Interestingly, immunohistochemical analysis revealed a preferential expression of Prx I and II in Leydig and Sertoli cells, respectively. Neither Prx I nor Prx II expression was obvious in the testicular germ cells including spermatogonia and spermatocytes. Ionizing radiation exerted oxidative stress on the testis and induced apoptosis primarily in the germ cells. When the irradiated testis was examined, the Prx system was found to be transiently up-regulated. Taken together, we suggest that the relative radiation-resistance of Leydig and Sertoli cells could be attributed in part to the antioxidant function of the Prx system in these cells.


Molecular and Cellular Biology | 2003

Activating Signal Cointegrator 2 Required for Liver Lipid Metabolism Mediated by Liver X Receptors in Mice

Seung Whan Kim; Keunhee Park; Eunyee Kwak; Eunho Choi; Seunghee Lee; Jungyeob Ham; Heonjoong Kang; Jong Man Kim; Seung Yong Hwang; Young-Yun Kong; Keesook Lee; Jae Woon Lee

ABSTRACT Activating signal cointegrator 2 (ASC-2), a cancer-amplified transcriptional coactivator of nuclear receptors and many other transcription factors, contains two LXXLL-type nuclear receptor interaction domains. Interestingly, the second LXXLL motif is highly specific to the liver X receptors (LXRs). In cotransfection, DN2, an ASC-2 fragment encompassing this motif, exerts a potent dominant-negative effect on transactivation by LXRs, which is rescued by ectopic coexpression of the full-length ASC-2 but not by other LXXLL-type coactivators, such as SRC-1 and TRAP220. In contrast, DN2/m, in which the LXXLL motif is mutated to LXXAA to abolish the interactions with LXRs, is without any effect. Accordingly, expression of DN2, but not DN2/m, in transgenic mice results in phenotypes that are highly homologous to those previously observed with LXRα−/− mice, including a rapid accumulation of large amounts of cholesterol and down-regulation of the known lipid-metabolizing target genes of LXRα in the liver upon being fed a high-cholesterol diet. These results identify ASC-2 as a physiologically important transcriptional coactivator of LXRs and demonstrate its pivotal role in the liver lipid metabolism.


Free Radical Biology and Medicine | 2009

ROS inhibit the expression of testicular steroidogenic enzyme genes via the suppression of Nur77 transactivation.

Seung-Yon Lee; Eun-Yeung Gong; Cheol Yi Hong; Keon-Hee Kim; Jung-Soo Han; Jae Chun Ryu; Ho Zoon Chae; Chul-Ho Yun; Keesook Lee

Steroidogenesis decreases with aging in the testis, whereas the levels of reactive oxygen species (ROS) increase. In addition, ROS have been reported to inhibit testicular steroidogenesis. Here, we investigated the effects of ROS on the transcriptional activity of Nur77, one of the major transcription factors that regulate the expression of steroidogenic enzyme genes. ROS signaling inhibited Nur77 transactivation, which was diminished by either treatment with c-Jun N-terminal kinase (JNK) inhibitor or the expression of a dominant negative form of JNK. This suggests the involvement of JNK signaling, which elevates the expression of c-Jun as well as its phosphorylation in Leydig cells. In transient transfection assays, c-Jun suppressed Nur77 transactivation in a dose-dependent manner. Further studies using c-Jun mutants revealed that the protein level of c-Jun, but not phosphorylation itself, was important for the suppression of Nur77 transactivation. Nur77 directly interacted with c-Jun in vivo, which blocked the DNA binding activity of Nur77. Together, these results suggest that ROS signaling-mediated c-Jun upregulation suppresses the expression of steroidogenic enzyme genes by inhibiting Nur77 transactivation, resulting in the reduction of testicular steroidogenesis. These findings may provide a mechanistic explanation for the age-related decline in testicular steroid hormone production.


Biology of Reproduction | 2005

Expression of the Putative Sterol Binding Protein Stard6 Gene Is Male Germ Cell Specific

Cynthia Gomes; Sung-Dug Oh; Jung-Woo Kim; Sang-Young Chun; Keesook Lee; Hyuk-Bang Kwon; Jaemog Soh

Abstract Mammalian spermatogenesis is orchestrated by many specific molecular and cellular events. To understand the detailed mechanism by which spermatogenesis is controlled, the specific genes involved in this process must be identified and studied. From the subtracted cDNA library of rat testis prepared using the representational difference analysis (RDA) method, we isolated the cDNA clone of steroidogenic acute regulatory (StAR) protein-related lipid transfer (START) protein 6 (Stard6). Stard6 cDNA consists of 1146 base pairs of nucleotides and has the longest open reading frame, of 227 amino acids. Northern blot analysis revealed Stard6 mRNA to be testis-specific. The mRNA transcript appeared from the third week of postnatal development, and the expression level increased up to adulthood. Moreover, in situ hybridization showed Stard6 mRNA expression to be germ cell-specific and expressed only during the maturation stages of round and elongated spermatids of adult rat testis. Western blot analysis with Stard6 antibody revealed a 28-kDa Stard6 protein only in testis. Immunohistochemistry further confirmed localization of Stard6 protein expressed in mature germ cells, in concert with the in situ hybridization result. Taken together, these results suggest that Stard6, a member of the START protein family, may play a role during germ cell maturation in adult rat testis.


Gene | 1999

Identification of ZFR, an ancient and highly conserved murine chromosome-associated zinc finger protein

Madeleine J. Meagher; Jill M. Schumacher; Keesook Lee; Robert W. Holdcraft; Susanne Edelhoff; Christine M. Disteche; Robert E. Braun

In a screen for RNA binding proteins expressed during murine spermatogenesis, we cloned a novel, ancient zinc finger protein possessing a region common to a small class of RNA binding proteins. Zfr (zinc finger RNA binding) encodes a protein of 1052 amino acids with three widely spaced Cys2His2 zinc fingers. Outside of the zinc fingers, ZFR shares a region that is highly conserved between several RNA binding proteins containing copies of the double-stranded RNA binding motif. By northern blotting, Zfr is expressed at highest levels within the testis, ovary and brain. Immunohistochemistry and confocal microscopy were used to show that ZFR is highly expressed during meiosis I in males and females and is chromosome associated. Zfr is also expressed in Sertoli cells in the testis and granulosa cells in the ovary where it is localized to the nucleus. Using fluorescent in situ hybridization we mapped Zfr to chromosome 15 region A. ZFR appears to be an ancient protein, as apparent homologs exist in invertebrates (D. melanogaster) nematodes (C. elegans) and humans (H. sapiens).

Collaboration


Dive into the Keesook Lee's collaboration.

Top Co-Authors

Avatar

Eun-Yeung Gong

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Eunsook Park

University of Tennessee

View shared research outputs
Top Co-Authors

Avatar

Cheol Yi Hong

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Hueng-Sik Choi

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryun Sup Ahn

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Chin-Hee Song

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Soma Chattopadhyay

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Hee-Sae Park

Chonnam National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge