Network


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

Hotspot


Dive into the research topics where Ki Jun Yoon is active.

Publication


Featured researches published by Ki Jun Yoon.


Nature | 2014

Synaptic dysregulation in a human iPS cell model of mental disorders

Zhexing Wen; Ha Nam Nguyen; Ziyuan Guo; Matthew A. Lalli; Xinyuan Wang; Yijing Su; Nam Shik Kim; Ki Jun Yoon; Jaehoon Shin; Ce Zhang; Georgia Makri; David Nauen; Huimei Yu; Elmer Guzman; Cheng Hsuan Chiang; Nadine Yoritomo; Kozo Kaibuchi; Jizhong Zou; Kimberly M. Christian; Linzhao Cheng; Christopher A. Ross; Russell L. Margolis; Gong Chen; Kenneth S. Kosik; Hongjun Song; Guo Li Ming

Dysregulated neurodevelopment with altered structural and functional connectivity is believed to underlie many neuropsychiatric disorders, and ‘a disease of synapses’ is the major hypothesis for the biological basis of schizophrenia. Although this hypothesis has gained indirect support from human post-mortem brain analyses and genetic studies, little is known about the pathophysiology of synapses in patient neurons and how susceptibility genes for mental disorders could lead to synaptic deficits in humans. Genetics of most psychiatric disorders are extremely complex due to multiple susceptibility variants with low penetrance and variable phenotypes. Rare, multiply affected, large families in which a single genetic locus is probably responsible for conferring susceptibility have proven invaluable for the study of complex disorders. Here we generated induced pluripotent stem (iPS) cells from four members of a family in which a frameshift mutation of disrupted in schizophrenia 1 (DISC1) co-segregated with major psychiatric disorders and we further produced different isogenic iPS cell lines via gene editing. We showed that mutant DISC1 causes synaptic vesicle release deficits in iPS-cell-derived forebrain neurons. Mutant DISC1 depletes wild-type DISC1 protein and, furthermore, dysregulates expression of many genes related to synapses and psychiatric disorders in human forebrain neurons. Our study reveals that a psychiatric disorder relevant mutation causes synapse deficits and transcriptional dysregulation in human neurons and our findings provide new insight into the molecular and synaptic etiopathology of psychiatric disorders.


Development | 2005

Mind bomb 1 is essential for generating functional Notch ligands to activate Notch.

Bon-Kyoung Koo; Hyoung Soo Lim; Ran Song; Mi Jeong Yoon; Ki Jun Yoon; Jin Sook Moon; Young Kim; Min Chul Kwon; Kyeong Won Yoo; Myung Phil Kong; Jinie Lee; Ajay B. Chitnis; Cheol-Hee Kim; Young-Yun Kong

The Delta-Notch signaling pathway is an evolutionarily conserved intercellular signaling mechanism essential for cell fate specification. Mind bomb 1 (Mib1) has been identified as a ubiquitin ligase that promotes the endocytosis of Delta. We now report that mice lacking Mib1 die prior to embryonic day 11.5, with pan-Notch defects in somitogenesis, neurogenesis, vasculogenesis and cardiogenesis. The Mib1–/– embryos exhibit reduced expression of Notch target genes Hes5, Hey1, Hey2 and Heyl, with the loss of N1icd generation. Interestingly, in the Mib1–/– mutants, Dll1 accumulated in the plasma membrane, while it was localized in the cytoplasm near the nucleus in the wild types, indicating that Mib1 is essential for the endocytosis of Notch ligand. In accordance with the pan-Notch defects in Mib1–/– embryos, Mib1 interacts with and regulates all of the Notch ligands, jagged 1 and jagged 2, as well as Dll1, Dll3 and Dll4. Our results show that Mib1 is an essential regulator, but not a potentiator, for generating functional Notch ligands to activate Notch signaling.


Neuron | 2008

Mind Bomb 1-Expressing Intermediate Progenitors Generate Notch Signaling to Maintain Radial Glial Cells

Ki Jun Yoon; Bon-Kyoung Koo; Sun Kyoung Im; Hyun Woo Jeong; Jaewang Ghim; Min Chul Kwon; Jin Sook Moon; Takaki Miyata; Young-Yun Kong

Notch signaling is critical for the stemness of radial glial cells (RGCs) during embryonic neurogenesis. Although Notch-signal-receiving events in RGCs have been well characterized, the signal-sending mechanism by the adjacent cells is poorly understood. Here, we report that conditional inactivation of mind bomb-1 (mib1), an essential component for Notch ligand endocytosis, in mice using the nestin and hGFAP promoters resulted in complete loss of Notch activation, which leads to depletion of RGCs, and premature differentiation into intermediate progenitors (IPs) and finally neurons, which were reverted by the introduction of active Notch1. Interestingly, Mib1 expression is restricted in the migrating IPs and newborn neurons, but not in RGCs. Moreover, sorted Mib1+ IPs and neurons can send the Notch signal to neighboring cells. Our results reveal that not only newborn neurons but also IPs are essential Notch-ligand-presenting cells for maintaining RGC stemness during both symmetric and asymmetric divisions.


PLOS ONE | 2007

An Obligatory Role of Mind Bomb-1 in Notch Signaling of Mammalian Development

Bon-Kyoung Koo; Mi Jeong Yoon; Ki Jun Yoon; Sun Kyoung Im; Yoon Young Kim; Cheol-Hee Kim; Pann Ghill Suh; Yuh Nung Jan; Young-Yun Kong

Background The Notch signaling pathway is an evolutionarily conserved intercellular signaling module essential for cell fate specification that requires endocytosis of Notch ligands. Structurally distinct E3 ubiquitin ligases, Neuralized (Neur) and Mind bomb (Mib), cooperatively regulate the endocytosis of Notch ligands in Drosophila. However, the respective roles of the mammalian E3 ubiquitin ligases, Neur1, Neur2, Mib1, and Mib2, in mammalian development are poorly understood. Methodology/Principal Findings Through extensive use of mammalian genetics, here we show that Neur1 and Neur2 double mutants and Mib2−/− mice were viable and grossly normal. In contrast, conditional inactivation of Mib1 in various tissues revealed the representative Notch phenotypes: defects of arterial specification as deltalike4 mutants, abnormal cerebellum and skin development as jagged1 conditional mutants, and syndactylism as jagged2 mutants. Conclusions/Significance Our data provide the first evidence that Mib1 is essential for Jagged as well as Deltalike ligand-mediated Notch signaling in mammalian development, while Neur1, Neur2, and Mib2 are dispensable.


Journal of Biological Chemistry | 2006

Neuralized-2 Regulates a Notch Ligand in Cooperation with Mind Bomb-1

Ran Song; Bon-Kyoung Koo; Ki Jun Yoon; Mi Jeong Yoon; Kyeong Won Yoo; Hyun Taek Kim; Hyeon Jeong Oh; Yoon Young Kim; Jin-Kwan Han; Cheol-Hee Kim; Young-Yun Kong

Mutations in Drosophila neuralized (Dneur) result in a variety of developmental defects that closely resemble those of Notch mutants and other Notch pathway mutants. However, mice with disrupted neur1 do not show any aberrant cell fate specifications in neurogenesis and somitogenesis. Thus, we speculated that other vertebrate neur homolog(s) might compensate for loss of the neur gene. Here, we report the paralog of mouse Neur1, named Neuralized-2 (Neur2), which is a ubiquitin-protein isopeptide ligase (E3) that interacts with and ubiquitinates Delta. Both murine Neur1 and Neur2 have similar degrees of homology to DNeur, and neur2 is expressed in patterns similar to those of neur1 in embryos, suggesting potential functional redundancy. Interestingly, two distinct classes of E3 ligases, Mind bomb-1 (Mib1) and Neur2, have cooperative but distinct roles in Delta endocytosis to Hrs-positive vesicles, i.e. Mib1 functions in the initial step of Delta endocytosis, and Neur2 is required for targeting endocytosed Delta to Hrs-positive vesicles. Thus, our study provides a new insight into how distinct E3 ligases work together in the endocytic pathways for Notch signaling.


The EMBO Journal | 2008

Crif1 is a novel transcriptional coactivator of STAT3

Min Chul Kwon; Bon-Kyoung Koo; Jin Sook Moon; Yoon Young Kim; Ki Cheol Park; Nam Kim; Mi Yi Kwon; Myung Phil Kong; Ki Jun Yoon; Sun Kyoung Im; Jaewang Ghim; Yong Mahn Han; Sung Key Jang; Minho Shong; Young-Yun Kong

Signal transducer and activator of transcription 3 (STAT3) is a transcriptional factor that performs a broad spectrum of biological functions in response to various stimuli. However, no specific coactivator that regulates the transcriptional activity of STAT3 has been identified. Here we report that CR6‐interacting factor 1 (Crif1) is a specific transcriptional coactivator of STAT3, but not of STAT1 or STAT5a. Crif1 interacts with STAT3 and positively regulates its transcriptional activity. Crif1−/− embryos were lethal around embryonic day 6.5, and manifested developmental arrest accompanied with defective proliferation and massive apoptosis. The expression of STAT3 target genes was markedly reduced in a Crif1−/− blastocyst culture and in Oncostatin M‐stimulated Crif1‐deficient MEFs. Importantly, the key activities of constitutively active STAT3‐C, such as transcription, DNA binding, and cellular transformation, were abolished in the Crif1‐null MEFs, suggesting the essential role of Crif1 in the transcriptional activity of STAT3. Our results reveal that Crif1 is a novel and essential transcriptional coactivator of STAT3 that modulates its DNA binding ability, and shed light on the regulation of oncogenic STAT3.


Journal of Experimental Medicine | 2008

Mind bomb 1 in the lymphopoietic niches is essential for T and marginal zone B cell development

Ran Song; Young Kim; Bon-Kyoung Koo; Hyun Woo Jeong; Mi Jeong Yoon; Ki Jun Yoon; Dong Jae Jun; Sun Kyoung Im; Juhee Shin; Myoung Phil Kong; Kyong-Tai Kim; Keejung Yoon; Young-Yun Kong

Notch signaling regulates lineage decisions at multiple stages of lymphocyte development, and Notch activation requires the endocytosis of Notch ligands in the signal-sending cells. Four E3 ubiquitin ligases, Mind bomb (Mib) 1, Mib2, Neuralized (Neur) 1, and Neur2, regulate the Notch ligands to activate Notch signaling, but their roles in lymphocyte development have not been defined. We show that Mib1 regulates T and marginal zone B (MZB) cell development in the lymphopoietic niches. Inactivation of the Mib1 gene, but not the other E3 ligases, Mib2, Neur1, and Neur2, abrogated T and MZB cell development. Reciprocal bone marrow (BM) transplantation experiments revealed that Mib1 in the thymic and splenic niches is essential for T and MZB cell development. Interestingly, when BM cells from transgenic Notch reporter mice were transplanted into Mib1-null mice, the Notch signaling was abolished in the double-negative thymocytes. In addition, the endocytosis of Dll1 was impaired in the Mib1-null microenvironment. Moreover, the block in T cell development and the failure of Dll1 endocytosis were also observed in coculture system by Mib1 knockdown. Our study reveals that Mib1 is the essential E3 ligase in T and MZB cell development, through the regulation of Notch ligands in the thymic and splenic microenvironments.


FEBS Letters | 2006

Snx5, as a Mind bomb-binding protein, is expressed in hematopoietic and endothelial precursor cells in zebrafish.

Kyeong Won Yoo; Eun Kim; Seung Hyun Jung; Myungchull Rhee; Bon-Kyoung Koo; Ki Jun Yoon; Young-Yun Kong; Cheol-Hee Kim

Notch signaling has an evolutionarily conserved function for cell fate determination and stem cell maintenance. Previously, we identified a novel component of the Notch signaling pathway in zebrafish, mind bomb, which encodes an E3 ubiquitin ligase essential for Notch signal activation. Further studies showed that Mind bomb −/− mouse embryos exhibited pan‐Notch phenotypes in various tissues, suggesting that Mind bomb function is conserved in mammals. Therefore we sought to understand the various molecular partners of Mind bomb using yeast two‐hybrid screening. In this search we identified Sorting nexin 5 (Snx5) as a novel interacting partner of Mind bomb. Furthermore we demonstrated that Snx5 colocalizes with Mind bomb in early endosomal compartments, suggesting that Snx5 is important for Mind bomb trafficking. In addition, we identified zebrafish orthologue of Snx5 and showed that snx5 is predominantly expressed in hematopoietic and endothelial precursor cells in zebrafish. We also found defects in hematopoiesis and blood vessel development in snx5 morpholino‐injected embryos. Taken together, we show that Snx5, a novel interacting partner of Mind bomb, may have an essential role for cell fate determination in early development.


Nucleic Acids Research | 2009

Molecule-level imaging of Pax6 mRNA distribution in mouse embryonic neocortex by molecular interaction force microscopy

Yu Jin Jung; Yu Shin Park; Ki Jun Yoon; Young-Yun Kong; Joon Won Park; Hong Gil Nam

Detection of the cellular and tissue distributions of RNA species is critical in our understanding of the regulatory mechanisms underlying cellular and tissue differentiation. Here, we show that an atomic force microscope tip modified with 27-acid dendron, a cone shaped molecule with 27 monomeric units forming its base, can be successfully used to map the spatial distribution of mouse Pax6 mRNA on sectioned tissues of the mouse embryonic neocortex. Scanning of the sectioned tissue with a 30-mer DNA probe attached to the apex of the dendron resulted in detection of the target mRNA on the tissue section, permitting mapping of the mRNA distribution at nanometer resolution. The unprecedented sensitivity and resolution of this process should be applicable to identification of molecular level distribution of various RNAs in a cell.


Molecular Brain | 2012

Mind bomb-1 is an essential modulator of long-term memory and synaptic plasticity via the Notch signaling pathway

Ki Jun Yoon; Hye Ryeon Lee; Yong Sang Jo; Kyongman An; Sang Yong Jung; Min Woo Jeong; Seok-Kyu Kwon; Nam Kim; Hyun Woo Jeong; Seo Hee Ahn; Kyong-Tai Kim; Kyungmin Lee; Eunjoon Kim; Joung Hun Kim; June Seek Choi; Bong-Kiun Kaang; Young-Yun Kong

BackgroundNotch signaling is well recognized as a key regulator of the neuronal fate during embryonic development, but its function in the adult brain is still largely unknown. Mind bomb-1 (Mib1) is an essential positive regulator in the Notch pathway, acting non-autonomously in the signal-sending cells. Therefore, genetic ablation of Mib1 in mature neuron would give valuable insight to understand the cell-to-cell interaction between neurons via Notch signaling for their proper function.ResultsHere we show that the inactivation of Mib1 in mature neurons in forebrain results in impaired hippocampal dependent spatial memory and contextual fear memory. Consistently, hippocampal slices from Mib1-deficient mice show impaired late-phase, but not early-phase, long-term potentiation and long-term depression without change in basal synaptic transmission at SC-CA1 synapses.ConclusionsThese data suggest that Mib1-mediated Notch signaling is essential for long-lasting synaptic plasticity and memory formation in the rodent hippocampus.

Collaboration


Dive into the Ki Jun Yoon's collaboration.

Top Co-Authors

Avatar

Young-Yun Kong

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Bon-Kyoung Koo

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Cheol-Hee Kim

Chungnam National University

View shared research outputs
Top Co-Authors

Avatar

Kyeong Won Yoo

Chungnam National University

View shared research outputs
Top Co-Authors

Avatar

Mi Jeong Yoon

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Min Chul Kwon

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Nam Kim

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Ran Song

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Sun Kyoung Im

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Yoon Young Kim

Pohang University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge