Network


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

Hotspot


Dive into the research topics where Yu-Jin Jo is active.

Publication


Featured researches published by Yu-Jin Jo.


Journal of Cell Science | 2015

Actin-capping proteins play essential roles in the asymmetric division of maturing mouse oocytes

Yu-Jin Jo; Woo-In Jang; Suk Namgoong; Nam-Hyung Kim

ABSTRACT Actin polymerization is essential for various stages of mammalian oocyte maturation, including spindle migration, actin cap formation, polar body extrusion and cytokinesis. The heterodimeric actin-capping protein is an essential element of the actin cytoskeleton. It binds to the fast-growing (barbed) ends of actin filaments and plays essential roles in various actin-mediated cellular processes. However, the roles of capping protein in mammalian oocyte maturation are poorly understood. We investigated the roles of capping protein in mouse oocytes and found that it is essential for correct asymmetric spindle migration and polar body extrusion. Capping protein mainly localized in the cytoplasm during maturation. By knocking down or ectopically overexpressing this protein, we revealed that it is crucial for efficient spindle migration and maintenance of the cytoplasmic actin mesh density. Expression of the capping-protein-binding region of CARMIL (also known as LRRC16A) impaired spindle migration and polar body extrusion during oocyte maturation and decreased the density of the cytoplasmic actin mesh. Taken together, these findings show that capping protein is an essential component of the actin cytoskeleton machinery that plays crucial roles in oocyte maturation, presumably by controlling the cytoplasmic actin mesh density.


Molecular Reproduction and Development | 2015

The Rho-GTPase effector ROCK regulates meiotic maturation of the bovine oocyte via myosin light chain phosphorylation and cofilin phosphorylation.

So-Rim Lee; Yong-Nan Xu; Yu-Jin Jo; Suk Namgoong; Nam-Hyung Kim

Oocyte meiosis involves a unique asymmetric division involving spindle movement from the central cytoplasm to the cortex, followed by polar body extrusion. ROCK is a Rho‐GTPase effector involved in various cellular functions in somatic cells as well as oocyte meiosis. ROCK was previously shown to promote actin organization by phosphorylating several downstream targets, including LIM domain kinase (LIMK), phosphorylated cofilin (p‐cofilin), and myosin light chain (MLC). In this study, we investigated the roles of ROCK and MLC during bovine oocyte meiosis. We found that ROCK was localized around the nucleus at the oocytes germinal‐vesicle (GV) stage, but spreads to the rest of the cytoplasm in later developmental stages. On the other hand, phosphorylated MLC (p‐MLC) localized at the cortex, and its abundance decreased by the metaphase‐II stage. Disrupting ROCK activity, via RNAi or the chemical inhibitor Y‐27632, blocked both cell cycle progression and polar body extrusion. ROCK inhibition also resulted in decreased cortical actin, p‐cofilin, and p‐MLC levels. Similar to the phenotype associated with inhibition of ROCK activity, inhibition of MLC kinase by the chemical inhibitor ML‐7 caused defects in polar body extrusion. Collectively, our results suggest that the ROCK/MLC/actomyosin as well as ROCK/LIMK/cofilin pathways regulate meiotic spindle migration and cytokinesis during bovine oocyte maturation. Mol. Reprod. Dev. 82: 849–858, 2015.


Cell Cycle | 2014

Non-muscle tropomyosin (Tpm3) is crucial for asymmetric cell division and maintenance of cortical integrity in mouse oocytes

Woo-In Jang; Yu-Jin Jo; Hak-Cheol Kim; Jia-Lin Jia; Suk Namgoong; Nam-Hyung Kim

Tropomyosins are actin-binding cytoskeletal proteins that play a pivotal role in regulating the function of actin filaments in muscle and non-muscle cells; however, the roles of non-muscle tropomyosins in mouse oocytes are unknown. This study investigated the expression and functions of non-muscle tropomyosin (Tpm3) during meiotic maturation of mouse oocytes. Tpm3 mRNA was detected at all developmental stages in mouse oocytes. Tpm3 protein was localized at the cortex during the germinal vesicle and germinal vesicle breakdown stages. However, the overall fluorescence intensity of Tpm3 immunostaining was markedly decreased in metaphase II oocytes. Knockdown of Tpm3 impaired asymmetric division of oocytes and spindle migration, considerably reduced the amount of cortical actin, and caused membrane blebbing during cytokinesis. Expression of a constitutively active cofilin mutant and Tpm3 overexpression confirmed that Tpm3 protects cortical actin from depolymerization by cofilin. The data indicate that Tpm3 plays crucial roles in maintaining cortical actin integrity and asymmetric cell division during oocyte maturation, and that dynamic regulation of cortical actin by Tpm3 is critical to ensure proper polar body protrusion.


PLOS ONE | 2015

Small Molecule Inhibitor of Formin Homology 2 Domains (SMIFH2) Reveals the Roles of the Formin Family of Proteins in Spindle Assembly and Asymmetric Division in Mouse Oocytes

Hak-Cheol Kim; Yu-Jin Jo; Nam-Hyung Kim; Suk Namgoong

Dynamic actin reorganization is the main driving force for spindle migration and asymmetric cell division in mammalian oocytes. It has been reported that various actin nucleators including Formin-2 are involved in the polarization of the spindle and in asymmetric cell division. In mammals, the formin family is comprised of 15 proteins. However, their individual roles in spindle migration and/or asymmetric division have not been elucidated yet. In this study, we employed a newly developed inhibitor for formin family proteins, small molecule inhibitor of formin homology 2 domains (SMIFH2), to assess the functions of the formin family in mouse oocyte maturation. Treatment with SMIFH2 during in vitro maturation of mouse oocytes inhibited maturation by decreasing cytoplasmic and cortical actin levels. In addition, treatment with SMIFH2, especially at higher concentrations (500 μM), impaired the proper formation of meiotic spindles, indicating that formins play a role in meiotic spindle formation. Knockdown of the mDia2 formins caused a similar decrease in oocyte maturation and abnormal spindle morphology, mimicking the phenotype of SMIFH2-treated cells. Collectively, these results suggested that besides Formin-2, the other proteins of the formin, including mDia family play a role in asymmetric division and meiotic spindle formation in mammalian oocytes.


Scientific Reports | 2016

Depletion of the LINC complex disrupts cytoskeleton dynamics and meiotic resumption in mouse oocytes

Yi-Bo Luo; In-Won Lee; Yu-Jin Jo; Suk Namgoong; Nam-Hyung Kim

The SUN (Sad-1/UNC-84) and KASH (Klarsicht/ANC-1/Syne/homology) proteins constitute the linker of nucleoskeleton and cytoskeleton (LINC) complex on the nuclear envelope. To date, the SUN1/KASH5 complex is known to function as meiotic-specific factors. In this study, gene-silencing methods were used to explore the roles of SUN1 and KASH5 in mouse oocytes after prophase. SUN1 was detected throughout the nucleus; however, KASH5 was dispersed through the cell. After germinal vesicle breakdown (GVBD), SUN1 and KASH5 migrated during spindle formation and localized to the spindle poles at the MII stage. Most oocytes were arrested at the germinal vesicle (GV) stage after depletion of either SUN1 or KASH5. The DNA damage response was triggered in SUN1-depleted oocytes and thus gave rise to the G2/M checkpoint protein, p-CHK1. Oocytes that underwent GVBD had relatively small and abnormal spindles and lower levels of cytoplasm F-actin mesh. Immunofluorescence results also indicated the dislocation of pericentrin and P150Glued after SUN1 or KASH5 depletion. Furthermore, KASH5 localized exclusively near the oocyte cortex after SUN1 depletion, but SUN1 localization was unaffected in KASH5-depleted oocytes. Taken together, the results suggest that SUN1 and KASH5 are essential factors in the regulation of meiotic resumption and spindle formation.


Oncotarget | 2017

Quercetin delays postovulatory aging of mouse oocytes by regulating SIRT expression and MPF activity

HaiYang Wang; Yu-Jin Jo; Jeong Su Oh; Nam-Hyung Kim

If no fertilization occurs at an appropriate time after ovulation, oocyte quality deteriorates rapidly as a process called postovulatory aging. Because the postovulatory aging of oocytes has detrimental effects on embryo development and offspring, many efforts have been made to prevent oocyte aging. Here we showed that quercetin prevented the decline in oocyte quality during postovulatory aging of oocytes. Quercetin treatment reduced aging-induced morphological changes and reactive oxygen species accumulation. Moreover, quercetin attenuated the aging-associated abnormalities in spindle organization and mitochondrial distribution, preventing decrease of SIRT expression and histone methylation. Quercetin also ameliorated the decrease in maturation-promoting factor activity and the onset of apoptosis during postovulatory aging. Furthermore, quercetin treatment during postovulatory aging improves early embryo development. Our results demonstrate that quercetin relieves deterioration in oocyte quality and improves subsequent embryo development.


Oncotarget | 2017

Protein phosphatase 2A regulatory subunit B55α functions in mouse oocyte maturation and early embryonic development

Shuang Liang; Jing Guo; Jeong-Woo Choi; Kyung-Tae Shin; HaiYang Wang; Yu-Jin Jo; Nam-Hyung Kim; Xiang-Shun Cui

Protein phosphatase 2A regulatory subunit B55α (PP2A-B55α) has been studied in mitosis. However, its functions in mammalian meiosis and early embryonic development remain unknown. Here, we report that PP2A-B55α is critical for mouse oocyte meiosis and preimplantation embryo development. Knockdown of PP2A-B55α in oocytes led to abnormal asymmetric division, disordered spindle dynamics, defects in chromosome congression, an increase in aneuploidy, and induction of the DNA damage response. Moreover, knockdown of PP2A-B55α in fertilized mouse zygotes impaired development to the blastocyst stage. The impairment of embryonic development might have been due to induction of sustained DNA damage in embryos, which caused apoptosis and inhibited cell proliferation and outgrowth potential at the blastocyst stage. Overall, these results provide a novel insight into the role of PP2A-B55α as a novel meiotic and embryonic competence factor at the onset of life.


Scientific Reports | 2016

Tropomodulin-3 is essential in asymmetric division during mouse oocyte maturation.

Yu-Jin Jo; Woo-In Jang; Nam-Hyung Kim; Suk Namgoong

The dynamic polymerization and depolymerization of actin filaments is essential for various cellular processes such as cell migration, rotation, cytokinesis, and mammalian oocyte maturation. Tropomodulin 3 (Tmod3) binds to the slow-growing (pointed) ends of the actin filament, thereby protecting the filament from depolymerization. However, the roles of Tmod3 in mammalian oocyte maturation remain elusive. Tmod3 mRNA and protein is present at all stages of mouse oocyte maturation. Tmod3 protein is mainly localized in the cytoplasm and appears enriched near the chromosome during maturation. By knocking down or ectopically overexpressing Tmod3, we confirmed that Tmod3 regulate the level of the intracytoplasmic actin mesh and asymmetric spindle migration. Expression of N-terminal Tmod3 (correspond to 1–155 amino acids), which contains the tropomyosin-binding site, results in decreased density of the actin mesh, thereby demonstrating the importance of the interaction between tropomyosin and tropomodulin for the maintenance of the actin mesh. Taken together, these findings indicate that Tmod3 plays crucial roles in oocyte maturation, presumably by protecting the actin filament from depolymerization and thereby controlling the density of the cytoplasmic actin mesh.


Molecular Human Reproduction | 2017

Spindlin 1 is essential for metaphase II stage maintenance and chromosomal stability in porcine oocytes

Jeong-Woo Choi; Ming-Hui Zhao; Shuang Liang; Jing Guo; Zi-Li Lin; Ying-Hua Li; Yu-Jin Jo; Nam-Hyung Kim; Xiang-Shun Cui

Study question What is the function of Spindlin 1 (Spin1) in metaphase II stage oocytes in pigs? Summary answer Depletion of Spin1 induces spontaneous oocyte activation and overexpression of Spin1 causes multinuclear formation through induction of DNA damage in porcine oocytes. What is known already Little is known about the function of Spin1 in oocytes and embryos. In mouse oocytes, Spin1 is specifically expressed during gametogenesis and is essential for meiotic resumption. In somatic cells, Spin1 promotes cancer cell proliferation and activates WNT/T-cell factor signaling. Study design size, duration After knockdown (KD) or overexpression of Spin1 in porcine MII-stage oocytes, MII maintenance was checked following additional culture for 24 h. Investigated parthenotes were cultured up to the four cell (72 h) or blastocyst (7 days) stages. Participants/materials, setting, methods Spin1 was knocked down in porcine oocytes and embryos via microinjection of pig Spin1-targeting siRNA. For Spin1 overexpression, porcine Spin1-eGFP cRNA was generated. Additionally, for rescue experiments, cRNA encoding siRNA-resistant mouse Spin1 was added to the pig Spin1-targeting siRNA. For the overexpression and rescue experiments, microinjection and culture were performed using the same methods as the KD experiments. Main results and the role of chance KD of Spin1 in MII-stage porcine oocytes reduced metaphase-promoting factor and mitogen-activated protein kinase activities, resulting in spontaneous pronuclear formation without calcium activation. However, the DNA damage response was triggered by Spin1 overexpression, generating the checkpoint protein γH2A.X. Furthermore, Spin1 overexpression blocked metaphase-anaphase transition and led to multinucleation in oocytes and embryos. Large scale data None. Limitations, reasons for caution This study is based on in vitro investigations with abnormal expression levels of Spin1. This may or may not accurately reflect the situation in vivo. Wider implications of the findings Spin1 is essential to maintain MII arrest, but a high level of Spin1 induces DNA damage in oocytes and embryos. Thus, a system to accurately regulate Spin1 expression operates in porcine MII-stage oocytes and embryos. Study funding and competing interest(s) This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (No. 2015R1D1A1A01057629). The authors declare no competing financial interests.


Molecular Reproduction and Development | 2016

Anillin controls cleavage furrow formation in the course of asymmetric division during mouse oocyte maturation

So-Rim Lee; Yu-Jin Jo; Suk Namgoong; Nam-Hyung Kim

Anillin is a scaffold protein that recruits several proteins involved in cleavage furrow formation during cytokinesis. The role of anilllin in symmetric cell divisions in somatic cells has been intensively studied, yet its involvement in cleavage furrow formation is still elusive. In this study, we investigated the role of anillin in mammalian oocyte maturation and cytokinesis. We found that anillin is localized around the nucleus during the oocyte germinal‐vesicle stage, and spreads to the cytoplasm after germinal vesicle breakdown. Thereafter, anillin concentrates at the site of the cleavage furrow from anaphase I to metaphase II. Disruption of anillin activity by microinjecting oocytes with specific siRNAs resulted in a failure of polar body extrusion and asymmetric division, and caused abnormal chromosome segregation during anaphase I. Furthermore, pharmacological inhibition of myosin light chain using Y‐27632 or ML‐7 resulted in decreased anillin expression. Collectively, our data suggest that anillin is an essential intracellular component that maintains the integrity of asymmetric division in mouse oocytes. Mol. Reprod. Dev. 83: 792–801, 2016

Collaboration


Dive into the Yu-Jin Jo's collaboration.

Top Co-Authors

Avatar

Nam-Hyung Kim

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Suk Namgoong

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

HaiYang Wang

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Woo-In Jang

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Xiang-Shun Cui

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Hak-Cheol Kim

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

In-Won Lee

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jeong Su Oh

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Jeong-Woo Choi

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jing Guo

Chungbuk National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge