Network


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

Hotspot


Dive into the research topics where Hea-Jin Jung is active.

Publication


Featured researches published by Hea-Jin Jung.


Molecular Biology of the Cell | 2011

Deficiencies in lamin B1 and lamin B2 cause neurodevelopmental defects and distinct nuclear shape abnormalities in neurons

Catherine Coffinier; Hea-Jin Jung; Chika Nobumori; Sandy Y. Chang; Yiping Tu; Richard H. Barnes; Yuko Yoshinaga; Pieter J. de Jong; Laurent Vergnes; Karen Reue; Loren G. Fong; Stephen G. Young

Lamin B1 is essential for neuronal migration and progenitor proliferation during the development of the cerebral cortex. The observation of distinct phenotypes of Lmnb1- and Lmnb2-knockout mice and the differences in the nuclear morphology of cortical neurons in vivo suggest that lamin B1 and lamin B2 play distinct functions in the developing brain.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Regulation of prelamin A but not lamin C by miR-9, a brain-specific microRNA

Hea-Jin Jung; Catherine Coffinier; Youngshik Choe; Anne P. Beigneux; Brandon S. J. Davies; Shao H. Yang; Richard H. Barnes; Janet Hong; Tao Sun; Samuel J. Pleasure; Stephen G. Young; Loren G. Fong

Lamins A and C, alternatively spliced products of the LMNA gene, are key components of the nuclear lamina. The two isoforms are found in similar amounts in most tissues, but we observed an unexpected pattern of expression in the brain. Western blot and immunohistochemistry studies showed that lamin C is abundant in the mouse brain, whereas lamin A and its precursor prelamin A are restricted to endothelial cells and meningeal cells and are absent in neurons and glia. Prelamin A transcript levels were low in the brain, but this finding could not be explained by alternative splicing. In lamin A-only knockin mice, where alternative splicing is absent and all the output of the gene is channeled into prelamin A transcripts, large amounts of lamin A were found in peripheral tissues, but there was very little lamin A in the brain. Also, in knockin mice expressing exclusively progerin (a toxic form of prelamin A found in Hutchinson–Gilford progeria syndrome), the levels of progerin in the brain were extremely low. Further studies showed that prelamin A expression, but not lamin C expression, is down-regulated by a brain-specific microRNA, miR-9. Expression of miR-9 in cultured cells reduced lamin A expression, and this effect was abolished when the miR-9–binding site in the prelamin A 3′ UTR was mutated. The down-regulation of prelamin A expression in the brain could explain why mouse models of Hutchinson–Gilford progeria syndrome are free of central nervous system pathology.


Journal of Biological Chemistry | 2010

Direct Synthesis of Lamin A, Bypassing Prelamin A Processing, Causes Misshapen Nuclei in Fibroblasts but No Detectable Pathology in Mice

Catherine Coffinier; Hea-Jin Jung; Ziwei Li; Chika Nobumori; Ui Jeong Yun; Emily Farber; Brandon S. J. Davies; Michael M. Weinstein; Shao H. Yang; Jan Lammerding; Javad N. Farahani; Laurent A. Bentolila; Loren G. Fong; Stephen G. Young

Lamin A, a key component of the nuclear lamina, is generated from prelamin A by four post-translational processing steps: farnesylation, endoproteolytic release of the last three amino acids of the protein, methylation of the C-terminal farnesylcysteine, and finally, endoproteolytic release of the last 15 amino acids of the protein (including the farnesylcysteine methyl ester). The last cleavage step, mediated by ZMPSTE24, releases mature lamin A. This processing scheme has been conserved through vertebrate evolution and is widely assumed to be crucial for targeting lamin A to the nuclear envelope. However, its physiologic importance has never been tested. To address this issue, we created mice with a “mature lamin A-only” allele (LmnaLAO), which contains a stop codon immediately after the last codon of mature lamin A. Thus, LmnaLAO/LAO mice synthesize mature lamin A directly, bypassing prelamin A synthesis and processing. The levels of mature lamin A in LmnaLAO/LAO mice were indistinguishable from those in “prelamin A-only” mice (LmnaPLAO/PLAO), where all of the lamin A is produced from prelamin A. LmnaLAO/LAO exhibited normal body weights and had no detectable disease phenotypes. A higher frequency of nuclear blebs was observed in LmnaLAO/LAO embryonic fibroblasts; however, the mature lamin A in the tissues of LmnaLAO/LAO mice was positioned normally at the nuclear rim. We conclude that prelamin A processing is dispensable in mice and that direct synthesis of mature lamin A has little if any effect on the targeting of lamin A to the nuclear rim in mouse tissues.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Farnesylation of lamin B1 is important for retention of nuclear chromatin during neuronal migration.

Hea-Jin Jung; Chika Nobumori; Chris N. Goulbourne; Yiping Tu; John M. Lee; Angelica Tatar; Daniel Wu; Yuko Yoshinaga; Pieter J. de Jong; Catherine Coffinier; Loren G. Fong; Stephen G. Young

Significance Both lamin B1 and lamin B2 have farnesyl lipid anchors, but the importance of this lipid modification has been unclear. We addressed that issue with knock-in mouse models. Mice expressing nonfarnesylated lamin B2 developed normally and were healthy. In contrast, mice expressing nonfarnesylated lamin B1 exhibited a severe neurodevelopmental abnormality accompanied by a striking defect in the cell nucleus. During the migration of neurons, the nuclear lamina was pulled free of the chromatin. Thus, farnesylation of lamin B1—but not lamin B2—is crucial for neuronal migration in the brain and for the retention of chromatin within the nuclear lamina. The role of protein farnesylation in lamin A biogenesis and the pathogenesis of progeria has been studied in considerable detail, but the importance of farnesylation for the B-type lamins, lamin B1 and lamin B2, has received little attention. Lamins B1 and B2 are expressed in nearly every cell type from the earliest stages of development, and they have been implicated in a variety of functions within the cell nucleus. To assess the importance of protein farnesylation for B-type lamins, we created knock-in mice expressing nonfarnesylated versions of lamin B1 and lamin B2. Mice expressing nonfarnesylated lamin B2 developed normally and were free of disease. In contrast, mice expressing nonfarnesylated lamin B1 died soon after birth, with severe neurodevelopmental defects and striking nuclear abnormalities in neurons. The nuclear lamina in migrating neurons was pulled away from the chromatin so that the chromatin was left “naked” (free from the nuclear lamina). Thus, farnesylation of lamin B1—but not lamin B2—is crucial for brain development and for retaining chromatin within the bounds of the nuclear lamina during neuronal migration.


Journal of Biological Chemistry | 2012

Understanding the Roles of Nuclear A- and B-type Lamins in Brain Development

Stephen G. Young; Hea-Jin Jung; Catherine Coffinier; Loren G. Fong

The nuclear lamina is composed mainly of lamins A and C (A-type lamins) and lamins B1 and B2 (B-type lamins). Dogma has held that lamins B1 and B2 play unique and essential roles in the nucleus of every eukaryotic cell. Recent studies have raised doubts about that view but have uncovered crucial roles for lamins B1 and B2 in neuronal migration during the development of the brain. The relevance of lamins A and C in the brain remains unclear, but it is intriguing that prelamin A expression in the brain is low and is regulated by miR-9, a brain-specific microRNA.


Journal of Clinical Investigation | 2016

Modulation of LMNA splicing as a strategy to treat prelamin A diseases

John M. Lee; Chika Nobumori; Yiping Tu; Catherine Choi; Shao H. Yang; Hea-Jin Jung; Timothy A. Vickers; Frank Rigo; C. Frank Bennett; Stephen G. Young; Loren G. Fong

The alternatively spliced products of LMNA, lamin C and prelamin A (the precursor to lamin A), are produced in similar amounts in most tissues and have largely redundant functions. This redundancy suggests that diseases, such as Hutchinson-Gilford progeria syndrome (HGPS), that are caused by prelamin A-specific mutations could be treated by shifting the output of LMNA more toward lamin C. Here, we investigated mechanisms that regulate LMNA mRNA alternative splicing and assessed the feasibility of reducing prelamin A expression in vivo. We identified an exon 11 antisense oligonucleotide (ASO) that increased lamin C production at the expense of prelamin A when transfected into mouse and human fibroblasts. The same ASO also reduced the expression of progerin, the mutant prelamin A protein in HGPS, in fibroblasts derived from patients with HGPS. Mechanistic studies revealed that the exon 11 sequences contain binding sites for serine/arginine-rich splicing factor 2 (SRSF2), and SRSF2 knockdown lowered lamin A production in cells and in murine tissues. Moreover, administration of the exon 11 ASO reduced lamin A expression in wild-type mice and progerin expression in an HGPS mouse model. Together, these studies identify ASO-mediated reduction of prelamin A as a potential strategy to treat prelamin A-specific diseases.


Science Translational Medicine | 2013

Targeting Protein Prenylation in Progeria

Stephen G. Young; Shao H. Yang; Brandon S. J. Davies; Hea-Jin Jung; Loren G. Fong

Results of a recent clinical trial in children with Hutchinson-Gilford progeria spur scientists to consider new therapeutic strategies. A clinical trial of a protein farnesyltransferase inhibitor (lonafarnib) for the treatment of Hutchinson-Gilford progeria syndrome (HGPS) was recently completed. Here, we discuss the mutation that causes HGPS, the rationale for inhibiting protein farnesyltransferase, the potential limitations of this therapeutic approach, and new potential strategies for treating the disease.


Nucleus | 2011

Are B-type lamins essential in all mammalian cells?

Shao H. Yang; Hea-Jin Jung; Catherine Coffinier; Loren G. Fong; Stephen G. Young

The B-type lamins are widely assumed to be essential for mammalian cells. In part, this assumption is based on a highly cited study that found that RNAi-mediated knockdown of lamin B1 or lamin B2 in HeLa cells arrested cell growth and led to apoptosis. Studies indicating that B-type lamins play roles in DNA replication, the formation of the mitotic spindle, chromatin organization and regulation of gene expression have fueled the notion that B-type lamins must be essential. But surprisingly, this idea had never been tested with genetic approaches. Earlier this year, a research group from UCLA reported the development of genetically modified mice that lack expression of both Lmnb1 and Lmnb2 in skin keratinocytes (a cell type that proliferates rapidly and participates in complex developmental programs). They reasoned that if lamins B1 and B2 were truly essential, then keratinocyte-specific lamin B1/lamin B2 knockout mice would exhibit severe pathology. Contrary to expectations, the skin and hair of lamin B1/lamin B2-deficient mice were quite normal, indicating that the B-type lamins are dispensable in some cell types. The same UCLA research group has gone on to show that lamin B1 and lamin B2 are critical for neuronal migration in the developing brain and for neuronal survival. The absence of either lamin B1 or lamin B2, or the absence of both B-type lamins, results in severe neurodevelopmental abnormalities.


Molecular and Cellular Biology | 2014

Nuclear lamins and neurobiology.

Stephen G. Young; Hea-Jin Jung; John M. Lee; Loren G. Fong

ABSTRACT Much of the work on nuclear lamins during the past 15 years has focused on mutations in LMNA (the gene for prelamin A and lamin C) that cause particular muscular dystrophy, cardiomyopathy, partial lipodystrophy, and progeroid syndromes. These disorders, often called “laminopathies,” mainly affect mesenchymal tissues (e.g., striated muscle, bone, and fibrous tissue). Recently, however, a series of papers have identified important roles for nuclear lamins in the central nervous system. Studies of knockout mice uncovered a key role for B-type lamins (lamins B1 and B2) in neuronal migration in the developing brain. Also, duplications of LMNB1 (the gene for lamin B1) have been shown to cause autosome-dominant leukodystrophy. Finally, recent studies have uncovered a peculiar pattern of nuclear lamin expression in the brain. Lamin C transcripts are present at high levels in the brain, but prelamin A expression levels are very low—due to regulation of prelamin A transcripts by microRNA 9. This form of prelamin A regulation likely explains why “prelamin A diseases” such as Hutchinson-Gilford progeria syndrome spare the central nervous system. In this review, we summarize recent progress in elucidating links between nuclear lamins and neurobiology.


Journal of Lipid Research | 2012

Inhibitors of protein geranylgeranyltransferase-I lead to prelamin A accumulation in cells by inhibiting ZMPSTE24.

Sandy Y. Chang; Sarah E. Hudon-Miller; Shao H. Yang; Hea-Jin Jung; John M. Lee; Emily Farber; Thangaiah Subramanian; Douglas A. Andres; H. Peter Spielmann; Christine A. Hrycyna; Stephen G. Young; Loren G. Fong

Protein farnesyltransferase (FTase) inhibitors, generally called “FTIs,” block the farnesylation of prelamin A, inhibiting the biogenesis of mature lamin A and leading to an accumulation of prelamin A within cells. A recent report found that a GGTI, an inhibitor of protein geranylgeranyltransferase-I (GGTase-I), caused an exaggerated accumulation of prelamin A in the presence of low amounts of an FTI. This finding was interpreted as indicating that prelamin A can be alternately prenylated by GGTase-I and that inhibiting both protein prenyltransferases leads to more prelamin A accumulation than blocking FTase alone. Here, we tested an alternative hypothesis—GGTIs are not specific for GGTase-I, and they lead to prelamin A accumulation by inhibiting ZMPSTE24 (a zinc metalloprotease that converts farnesyl–prelamin A to mature lamin A). In our studies, commonly used GGTIs caused prelamin A accumulation in human fibroblasts, but the prelamin A in GGTI-treated cells exhibited a more rapid electrophoretic mobility than prelamin A from FTI-treated cells. The latter finding suggested that the prelamin A in GGTI-treated cells might be farnesylated (which would be consistent with the notion that GGTIs inhibit ZMPSTE24). Indeed, metabolic labeling studies revealed that the prelamin A in GGTI-treated fibroblasts is farnesylated. Moreover, biochemical assays of ZMPSTE24 activity showed that ZMPSTE24 is potently inhibited by a GGTI. Our studies show that GGTIs inhibit ZMPSTE24, leading to an accumulation of farnesyl–prelamin A. Thus, caution is required when interpreting the effects of GGTIs on prelamin A processing.

Collaboration


Dive into the Hea-Jin Jung's collaboration.

Top Co-Authors

Avatar

Loren G. Fong

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shao H. Yang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chika Nobumori

University of California

View shared research outputs
Top Co-Authors

Avatar

John M. Lee

University of California

View shared research outputs
Top Co-Authors

Avatar

Yiping Tu

University of California

View shared research outputs
Top Co-Authors

Avatar

Angelica Tatar

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Wu

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge