Network


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

Hotspot


Dive into the research topics where Joungjoa Park is active.

Publication


Featured researches published by Joungjoa Park.


The New England Journal of Medicine | 2011

A common MUC5B promoter polymorphism and pulmonary fibrosis.

Max A. Seibold; Anastasia L. Wise; Marcy C. Speer; Mark P. Steele; Kevin K. Brown; James E. Loyd; Tasha E. Fingerlin; Weiming Zhang; Gunnar Gudmundsson; Steve D. Groshong; Christopher M. Evans; Stavros Garantziotis; Kenneth B. Adler; Burton F. Dickey; Roland M. du Bois; Ivana V. Yang; Aretha Herron; Dolly Kervitsky; Janet Talbert; Cheryl Markin; Joungjoa Park; Anne L. Crews; Susan Slifer; Scott S. Auerbach; Michelle G. Roy; Jia Lin; Corinne E. Hennessy; Marvin I. Schwarz; David A. Schwartz

BACKGROUND The mutations that have been implicated in pulmonary fibrosis account for only a small proportion of the population risk. METHODS Using a genomewide linkage scan, we detected linkage between idiopathic interstitial pneumonia and a 3.4-Mb region of chromosome 11p15 in 82 families. We then evaluated genetic variation in this region in gel-forming mucin genes expressed in the lung among 83 subjects with familial interstitial pneumonia, 492 subjects with idiopathic pulmonary fibrosis, and 322 controls. MUC5B expression was assessed in lung tissue. RESULTS Linkage and fine mapping were used to identify a region of interest on the p-terminus of chromosome 11 that included gel-forming mucin genes. The minor-allele of the single-nucleotide polymorphism (SNP) rs35705950, located 3 kb upstream of the MUC5B transcription start site, was present at a frequency of 34% among subjects with familial interstitial pneumonia, 38% among subjects with idiopathic pulmonary fibrosis, and 9% among controls (allelic association with familial interstitial pneumonia, P=1.2×10(-15); allelic association with idiopathic pulmonary fibrosis, P=2.5×10(-37)). The odds ratios for disease among subjects who were heterozygous and those who were homozygous for the minor allele of this SNP were 6.8 (95% confidence interval [CI], 3.9 to 12.0) and 20.8 (95% CI, 3.8 to 113.7), respectively, for familial interstitial pneumonia and 9.0 (95% CI, 6.2 to 13.1) and 21.8 (95% CI, 5.1 to 93.5), respectively, for idiopathic pulmonary fibrosis. MUC5B expression in the lung was 14.1 times as high in subjects who had idiopathic pulmonary fibrosis as in those who did not (P<0.001). The variant allele of rs35705950 was associated with up-regulation in MUC5B expression in the lung in unaffected subjects (expression was 37.4 times as high as in unaffected subjects homozygous for the wild-type allele, P<0.001). MUC5B protein was expressed in lesions of idiopathic pulmonary fibrosis. CONCLUSIONS A common polymorphism in the promoter of MUC5B is associated with familial interstitial pneumonia and idiopathic pulmonary fibrosis. Our findings suggest that dysregulated MUC5B expression in the lung may be involved in the pathogenesis of pulmonary fibrosis. (Funded by the National Heart, Lung, and Blood Institute and others.).


Nature Medicine | 2004

A MARCKS-related peptide blocks mucus hypersecretion in a mouse model of asthma

Monique Singer; Linda D. Martin; B. Boris Vargaftig; Joungjoa Park; Achim D Gruber; Yuehua Li; Kenneth B. Adler

Mucus hypersecretion is a crucial feature of pulmonary diseases such as asthma, chronic bronchitis and cystic fibrosis. Despite much research, there is still no effective therapy for this condition. Recently, we showed that the myristoylated, alanine-rich C-kinase substrate (MARCKS) protein is required for mucus secretion by human bronchial epithelial cells in culture. Having synthesized a peptide corresponding to the N-terminal domain of MARCKS, we now show that the intratracheal instillation of this peptide blocks mucus hypersecretion in a mouse model of asthma. A missense peptide with the same amino acid composition has no effect. Based on quantitative histochemical analysis of the mouse airways, the peptide seems to act by blocking mucus release from goblet cells, possibly by inhibiting the attachment of MARCKS to membranes of intracellular mucin granules. These results support a pivotal role for MARCKS protein, specifically its N-terminal region, in modulating this secretory process in mammalian airways. Intratracheal administration of this MARCKS-related peptide could therapeutically reduce mucus secretion in the airways of human patients with asthma, chronic bronchitis and cystic fibrosis.


American Journal of Respiratory Cell and Molecular Biology | 2010

Myristoylated Alanine-Rich C-Kinase Substrate (MARCKS) Protein Regulation of Human Neutrophil Migration

Rachael E. Eckert; Laura E. Neuder; Joungjoa Park; Kenneth B. Adler; Samuel L. Jones

Neutrophil migration into infected tissues is essential for host defense, but products of activated neutrophils can be quite damaging to host cells. Neutrophil influx into the lung and airways and resultant inflammation characterizes diseases such as chronic obstructive pulmonary disease, bronchiectasis, and cystic fibrosis. To migrate, neutrophils must reorganize the actin cytoskeleton to establish a leading edge pseudopod and a trailing edge uropod. The actin-binding protein myristoylated alanine-rich C-kinase substrate (MARCKS) has been shown to bind and cross-link actin in a variety of cell types and to co-localize with F-actin in the leading edge lamellipodium of migrating fibroblasts. The hypothesis that MARCKS has a role in the regulation of neutrophil migration was tested using a cell-permeant peptide derived from the MARCKS myristoylated aminoterminus (MANS peptide). Treatment of isolated human neutrophils with MANS significantly inhibited both their migration and beta2 integrin-dependent adhesion in response to N-formyl-methionyl-leucyl-phenylalanine (fMLF), IL-8, or leukotriene (LT)B(4). The IC(50) for fMLF-induced migration and adhesion was 17.1 microM and 12.5 microM, respectively. MANS significantly reduced the F-actin content in neutrophils 30 seconds after fMLF stimulation, although the peptide did not alter the ability of cells to polarize or spread. MANS did not alter fMLF-induced increases in surface beta2 integrin expression. These results suggest that MARCKS, via its myristoylated aminoterminus, is a key regulator of neutrophil migration and adhesion.


American Journal of Respiratory Cell and Molecular Biology | 2008

MARCKS Regulation of Mucin Secretion by Airway Epithelium in Vitro: Interaction with Chaperones

Joungjoa Park; Shijing Fang; Anne L. Crews; Ko-Wei Lin; Kenneth B. Adler

We have reported previously that myristoylated alanine-rich C kinase substrate (MARCKS) is a key regulatory molecule controlling mucin secretion by airway epithelial cells in vitro and in vivo. The results of those studies supported a mechanism whereby MARCKS, upon phosphorylation by protein kinase C (PKC), translocates from plasma membrane to cytoplasm, where its binding to membranes of intracellular mucin granules is a key component of the secretory pathway. It remains unknown how MARCKS is targeted to and/or preferentially attaches to mucin granule membranes. We hypothesized that the chaperone cysteine string protein (CSP) may play an important role in this process. CSP was shown to associate with membranes of intracellular mucin granules in well-differentiated normal human bronchial epithelial (NHBE) cells in vitro, as determined by ultrastructural immunohistochemistry and Western blotting of isolated granule membranes. CSP in these cells complexed with MARCKS, as shown by co-immunoprecipitation. Given reported associations between CSP and a second chaperone, heat shock protein 70 (HSP70), a role for HSP70 in the MARCKS-dependent secretory mechanism also was investigated. HSP70 appeared to form a trimeric complex with MARCKS and CSP associated with mucin granule membranes within airway epithelial cells. Transfection of the HBE1 human bronchial epithelial cell line with siRNAs targeting sequences of MARCKS, CSP, or HSP70 resulted, in each case, in significant knockdown of expression of these proteins and subsequent attenuation of mucin secretion. The results provide the first evidence that CSP and HSP70, and their interactions with MARCKS, are involved in mucin secretion.


Biochimica et Biophysica Acta | 2011

Regulation of mucin secretion and inflammation in asthma: A role for MARCKS protein?

Teresa D. Green; Anne L. Crews; Joungjoa Park; Shijing Fang; Kenneth B. Adler

BACKGROUND A major characteristic of asthmatic airways is an increase in mucin (the glycoprotein component of mucus) producing and secreting cells, which leads to increased mucin release that further clogs constricted airways and contributes markedly to airway obstruction and, in the most severe cases, to status asthmaticus. Asthmatic airways show both a hyperplasia and metaplasia of goblet cells, mucin-producing cells in the epithelium; hyperplasia refers to enhanced numbers of goblet cells in larger airways, while metaplasia refers to the appearance of these cells in smaller airways where they normally are not seen. With the number of mucin-producing and secreting cells increased, there is a coincident hypersecretion of mucin which characterizes asthma. On a cellular level, a major regulator of airway mucin secretion in both in vitro and in vivo studies has been shown to be MARCKS (myristoylated alanine-rich C kinase substrate) protein, a ubiquitous substrate of protein kinase C (PKC). GENERAL SIGNIFICANCE In this review, properties of MARCKS and how the protein may regulate mucin secretion at a cellular level will be discussed. In addition, the roles of MARCKS in airway inflammation related to both influx of inflammatory cells into the lung and release of granules containing inflammatory mediators by these cells will be explored. This article is part of a Special Issue entitled: Biochemistry of Asthma.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2013

MARCKS and HSP70 interactions regulate mucin secretion by human airway epithelial cells in vitro

Shijing Fang; Anne L. Crews; Wei Chen; Joungjoa Park; Qi Yin; Xiu-Rong Ren; Kenneth B. Adler

Myristoylated alanine-rich C kinase substrate (MARCKS) protein has been recognized as a key regulatory molecule controlling mucin secretion by airway epithelial cells in vitro and in vivo. We recently showed that two intracellular chaperones, heat shock protein 70 (HSP70) and cysteine string protein (CSP), associate with MARCKS in the secretory mechanism. To elucidate more fully MARCKS-HSP70 interactions in this process, studies were performed in well-differentiated normal human bronchial epithelial (NHBE) cells maintained in air-liquid interface culture utilizing specific pharmacological inhibition of HSP70 with pyrimidinone MAL3-101 and siRNA approaches. The results indicate that HSP70 interaction with MARCKS is enhanced after exposure of the cells to the protein kinase C activator/mucin secretagogue, phorbol 12-myristate 13-acetate (PMA). Pretreatment of NHBEs with MAL3-101 attenuated in a concentration-dependent manner PMA-stimulated mucin secretion and interactions among HSP70, MARCKS, and CSP. In additional studies, trafficking of MARCKS in living NHBE cells was investigated after transfecting cells with fluorescently tagged DNA constructs: MARCKS-yellow fluorescent protein, and/or HSP70-cyan fluorescent protein. Cells were treated with PMA 48 h posttransfection, and trafficking of the constructs was examined by confocal microscopy. MARCKS translocated rapidly from plasma membrane to cytoplasm, whereas HSP70 was observed in the cytoplasm and appeared to associate with MARCKS after PMA exposure. Pretreatment of cells with either MAL3-101 or HSP70 siRNA inhibited translocation of MARCKS. These results provide evidence of a role for HSP70 in mediating mucin secretion via interactions with MARCKS and that these interactions are critical for the cytoplasmic translocation of MARCKS upon its phosphorylation.


American Journal of Respiratory Cell and Molecular Biology | 2010

MARCKS and Related Chaperones Bind to Unconventional Myosin V Isoforms in Airway Epithelial Cells

Ko-Wei Lin; Shijing Fang; Joungjoa Park; Anne L. Crews; Kenneth B. Adler

We have shown previously that myristoylated alanine-rich C kinase substrate (MARCKS) is a key regulatory molecule in the process of mucin secretion by airway epithelial cells, and that part of the secretory mechanism involves intracellular associations of MARCKS with specific chaperones: heat shock protein 70 (Hsp70) and cysteine string protein (CSP). Here, we report that MARCKS also interacts with unconventional myosin isoforms within these cells, and further molecular interactions between MARCKS and these chaperones/cytoskeletal proteins are elucidated. Primary human bronchial epithelial cells and the HBE1 cell line both expressed myosin V and VI proteins, and both MARCKS and CSP were shown to bind to myosin V, specifically Va and Vc. This binding was enhanced by exposing the cells to phorbol-12-myristate-13-acetate, an activator of protein kinase C and stimulator of mucin secretion. Binding of MARCKS, Hsp70, and CSP was further investigated by His-tagged pull down assays of purified recombinant proteins and multiple transfections of HBE1 cells with fusion proteins (MARCKS-HA; Flag-Hsp70; c-Myc-CSP) and immunoprecipitation. The results showed that MARCKS binds directly to Hsp70, and that Hsp70 binds directly to CSP, but that MARCKS binding to CSP appears to require the presence of Hsp70. Interrelated binding(s) of MARCKS, chaperones, and unconventional myosin isoforms may be integral to the mucin secretion process.


Journal of Epithelial Biology & Pharmacology | 2011

Aerosolized BIO-11006, a Novel MARCKS- Related Peptide, Improves Airway Obstruction in a Mouse Model of Mucus Hypersecretion

Anurag A. Agrawal; Edwin C. Murphy; Joungjoa Park; Kenneth B. Adler; Indu Parikh

Rationale: Mucus hypersecretion has been shown to be an important cause of airway obstruction in COPD and asthma, and has been associated with fatal asthma. Previous in-vitro and in-vivo studies have provided direct evidence that the myristoylated alanine-rich C kinase substrate (MARCKS) is a central regulatory molecule linking secretagogue stimu- lation at the goblet cell surface to mucin granule release. BIO-11006 is a novel, highly soluble, aerosolized peptide that inhibits MARCKS function and results in reduced mucus secretion and improved airway obstruction. BIO-11006 is being tested in Phase 2 clinical studies of COPD. Objective: To determine access of BIO-11006 to the intracellular compartment of normal human bronchial epithelial (NHBE) cells, and assess the effectiveness of BIO-11006 inhalation solution in inhibiting airway obstruction caused by mucus hypersecretion as a function of dose and time. Methods: Biotin labeled BIO-11006 was incubated with NHBE cells and visualized with fluorescein-labeled streptavidin. Ovalbumin-sensitized and challenged mice with airway inflammation and hyperresponsiveness to methacholine (MCh) were treated with aerosolized BIO-11006 solution prior to MCh challenge, and effects on airway obstruction and mucus secretion were measured. Results: A 30-minute aerosol exposure to a 10 mM BIO-11006 solution was maximally effective in inhibiting MCh- induced airway obstruction and mucus hypersecretion. There was a long-lasting effect on inhibition of airway obstruction after a single treatment with BIO-11006 (t 1/2 ~4 hrs). The duration of effect on inhibition of mucin secretion was shorter (t 1/2 ~2 hrs). Conclusion: The pharmacodynamic properties of aerosolized BIO-11006 Inhalation Solution are suitable for therapeutic use. Inhaled BIO-11006 effectively inhibits mucin secretion and improves airway obstruction.


American Journal of Respiratory Cell and Molecular Biology | 2006

A Peptide Against the N-Terminus of Myristoylated Alanine-Rich C Kinase Substrate Inhibits Degranulation of Human Leukocytes In Vitro

Shuji Takashi; Joungjoa Park; Shijing Fang; Sekiya Koyama; Indu Parikh; Kenneth B. Adler


American Journal of Pathology | 2007

Protein Kinase Cδ Regulates Airway Mucin Secretion via Phosphorylation of MARCKS Protein

Jin-Ah Park; Anne L. Crews; William R. Lampe; Shijing Fang; Joungjoa Park; Kenneth B. Adler

Collaboration


Dive into the Joungjoa Park's collaboration.

Top Co-Authors

Avatar

Kenneth B. Adler

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Anne L. Crews

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Shijing Fang

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Ko-Wei Lin

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samuel L. Jones

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Anastasia L. Wise

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Burton F. Dickey

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge