Network


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

Hotspot


Dive into the research topics where Donghwa Kim is active.

Publication


Featured researches published by Donghwa Kim.


Cancer Research | 2005

Action of the Src Family Kinase Inhibitor, Dasatinib (BMS-354825), on Human Prostate Cancer Cells

Sangkil Nam; Donghwa Kim; Jin Q. Cheng; Shumin M. Zhang; Ji-Hyun Lee; Ralf Buettner; Janni Mirosevich; Francis Y. Lee; Richard Jove

Src family kinases (SFK) are currently being investigated as targets for treatment strategies in various cancers. The novel SFK/Abl inhibitor, dasatinib (BMS-354825), is a promising therapeutic agent with oral bioavailability. Dasatinib has been shown to inhibit growth of Bcr-Abl-dependent chronic myeloid leukemia xenografts in nude mice. Dasatinib also has been shown to have activity against cultured human prostate and breast cancer cells. However, the molecular mechanism by which dasatinib acts on epithelial tumor cells remains unknown. In this study, we show that dasatinib blocks the kinase activities of the SFKs, Lyn, and Src, in human prostate cancer cells at low nanomolar concentrations. Moreover, focal adhesion kinase and Crk-associated substrate (p130(CAS)) signaling downstream of SFKs are also inhibited at similar concentrations of dasatinib. Consistent with inhibition of these signaling pathways, dasatinib suppresses cell adhesion, migration, and invasion of prostate cancer cells at low nanomolar concentrations. Therefore, dasatinib has potential as a therapeutic agent for metastatic prostate cancers harboring activated SFK and focal adhesion kinase signaling.


Proceedings of the National Academy of Sciences of the United States of America | 2005

Indirubin derivatives inhibit Stat3 signaling and induce apoptosis in human cancer cells

Sangkil Nam; Ralf Buettner; James Turkson; Donghwa Kim; Jin Q. Cheng; Stephan Muehlbeyer; Frankie Hippe; Sandra Vatter; Karl-Heinz Merz; Gerhard Eisenbrand; Richard Jove

Stat3 protein has an important role in oncogenesis and is a promising anticancer target. Indirubin, the active component of a traditional Chinese herbal medicine, has been shown previously to inhibit cyclin-dependent kinases, resulting in cell cycle arrest. Here, we show that the indirubin derivatives E564, E728, and E804 potently block constitutive Stat3 signaling in human breast and prostate cancer cells. In addition, E804 directly inhibits Src kinase activity (IC50 = 0.43 μM) in an in vitro kinase assay. Levels of tyrosyl phosphorylation of c-Src are also reduced in cultured cells 30 min after E804 treatment. Tyrosyl phosphorylation of Stat3, which is known to be phosphorylated by c-Src, was decreased, and constitutive Stat3 DNA binding-activity was suppressed in cells 30 min after E804 treatment. The antiapoptotic proteins Mcl-1 and Survivin, which are encoded in target genes of Stat3, were down-regulated by indirubin derivatives, followed by induction of apoptosis. These results demonstrate that E804 directly blocks the Src-Stat3 signaling pathway, suggesting that the antitumor activity of indirubin compounds is at least partially due to inhibition of this pathway.


Frontiers in Bioscience | 2005

AKT/PKB signaling mechanisms in cancer and chemoresistance.

Donghwa Kim; Han C. Dan; Sungman Park; Lin Yang; Qiyuan Liu; Satoshi Kaneko; Jinying Ning; Lili He; Hua Yang; Mei Sun; Santo V. Nicosia; Jin Q. Cheng

During the past decade, Akt (also known as protein kinase B, PKB) has been extensively studied. It regulates a variety of cellular processes by mediating extracellular (mitogenic growth factor, insulin and stress) and intracellular (altered tyrosine receptor kinases, Ras and Src) signals. Activation of Akt by these signals is via its pleckstrin homology (PH) domain binding to products of phosphatidylinositol 3-kinase (PI3K). This process is negatively regulated by a dual phosphatase PTEN tumor suppressor. Today, more than 30 Akt substrates have been identified. These phosphorylation events mediate the effects of Akt on cell survival, growth, differentiation, angiogenesis, migration and metabolism. Further, PI3K/PTEN/Akt pathway is frequently altered in many human malignancies and overexpression of Akt induces malignant transformation and chemoresistance. Thus, the Akt pathway is a major target for anti-cancer drug development. This review focuses on Akt signaling mechanism in oncogenesis and chemoresistance, and ongoing translational efforts to therapeutically target Akt.


Journal of Biological Chemistry | 2010

Phosphoinositide 3-Kinase/Akt Inhibits MST1-Mediated Pro-apoptotic Signaling through Phosphorylation of Threonine 120

Zengqiang Yuan; Donghwa Kim; Shaokun Shu; Junbing Wu; Jianping Guo; Lei Xiao; Satoshi Kaneko; Domenico Coppola; Jin Q. Cheng

The protein kinase mammalian sterile 20-like kinase 1 (MST1) is a mammalian homologue of the Drosophila hippo and plays a critical role in regulation of programmed cell death. MST1 exerts pro-apoptotic function through cleavage, autophosphorylation-Thr(183) and subsequent translocation to the nucleus where it phosphorylates a number of molecules, including LATS1/2, FOXO, JNK, and histone H2B. Here, we show that the cleavage of MST1 is inhibited by the phosphatidylinositol 3-kinase/Akt pathway. Akt interacts with MST1 and phosphorylates a highly conserved residue threonine 120 of MST1, which leads to inhibition of its kinase activity and nuclear translocation as well as the autophosphorylation of Thr(183). Phospho-MST1-Thr(120) failed to activate downstream targets FOXO3a and JNK. Further, inverse correlation between pMST1-Thr(120) and pMST1-Thr(183) was observed in human ovarian tumors. These findings indicate that the phosphorylation of MST1-Thr(120) by Akt could be a major mechanism of regulation of the Hippo/MST1 pathway by cell survival signaling.The protein kinase mammalian sterile 20-like kinase 1 (MST1) is a mammalian homologue of the Drosophila hippo and plays a critical role in regulation of programmed cell death. MST1 exerts pro-apoptotic function through cleavage, autophosphorylation-Thr183 and subsequent translocation to the nucleus where it phosphorylates a number of molecules, including LATS1/2, FOXO, JNK, and histone H2B. Here, we show that the cleavage of MST1 is inhibited by the phosphatidylinositol 3-kinase/Akt pathway. Akt interacts with MST1 and phosphorylates a highly conserved residue threonine 120 of MST1, which leads to inhibition of its kinase activity and nuclear translocation as well as the autophosphorylation of Thr183. Phospho-MST1-Thr120 failed to activate downstream targets FOXO3a and JNK. Further, inverse correlation between pMST1-Thr120 and pMST1-Thr183 was observed in human ovarian tumors. These findings indicate that the phosphorylation of MST1-Thr120 by Akt could be a major mechanism of regulation of the Hippo/MST1 pathway by cell survival signaling.


Nature Structural & Molecular Biology | 2012

PHF20 is an effector protein of p53 double lysine methylation that stabilizes and activates p53.

Gaofeng Cui; Sungman Park; Aimee I. Badeaux; Donghwa Kim; James J. Lee; James R. Thompson; Fei Yan; Satoshi Kaneko; Zengqiang Yuan; Maria Victoria Botuyan; Mark T. Bedford; Jin Q. Cheng; Georges Mer

PHF20 is a multidomain protein and subunit of a lysine acetyltransferase complex that acetylates histone H4 and p53 but whose function is unclear. Using biochemical, biophysical and cellular approaches, we determined that PHF20 is a direct regulator of p53. A Tudor domain in PHF20 recognized p53 dimethylated at Lys370 or Lys382 and a homodimeric form of this Tudor domain could associate with the two dimethylated sites on p53 with enhanced affinity, indicating a multivalent interaction. Association with PHF20 promotes stabilization and activation of p53 by diminishing Mdm2-mediated p53 ubiquitylation and degradation. PHF20 contributes to upregulation of p53 in response to DNA damage, and ectopic expression of PHF20 in different cell lines leads to phenotypic changes that are hallmarks of p53 activation. Overall our work establishes that PHF20 functions as an effector of p53 methylation that stabilizes and activates p53.


Journal of Biological Chemistry | 2010

A Small Molecule Inhibits Akt through Direct Binding to Akt and Preventing Akt Membrane Translocation

Donghwa Kim; Mei Sun; Lili He; Qing-Hua Zhou; Jun Chen; Xia-meng Sun; Gerold Bepler; Said M. Sebti; Jin Q. Cheng

The Akt pathway is frequently hyperactivated in human cancer and functions as a cardinal nodal point for transducing extracellular and intracellular oncogenic signals and, thus, presents an exciting target for molecular therapeutics. Here we report the identification of a small molecule Akt/protein kinase B inhibitor, API-1. Although API-1 is neither an ATP competitor nor substrate mimetic, it binds to pleckstrin homology domain of Akt and blocks Akt membrane translocation. Furthermore, API-1 treatment of cancer cells results in inhibition of the kinase activities and phosphorylation levels of the three members of the Akt family. In contrast, API-1 had no effects on the activities of the upstream Akt activators, phosphatidylinositol 3-kinase, phosphatidylinositol-dependent kinase-1, and mTORC2. Notably, the kinase activity and phosphorylation (e.g. Thr(P)308 and Ser(P)473) levels of constitutively active Akt, including a naturally occurring mutant AKT1-E17K, were inhibited by API-1. API-1 is selective for Akt and does not inhibit the activation of protein kinase C, serum and glucocorticoid-inducible kinase, protein kinase A, STAT3, ERK1/2, or JNK. The inhibition of Akt by API-1 resulted in induction of cell growth arrest and apoptosis selectively in human cancer cells that harbor constitutively activated Akt. Furthermore, API-1 inhibited tumor growth in nude mice of human cancer cells in which Akt is elevated but not of those cancer cells in which it is not. These data indicate that API-1 directly inhibits Akt through binding to the Akt pleckstrin homology domain and blocking Akt membrane translocation and that API-1 has anti-tumor activity in vitro and in vivo and could be a potential anti-cancer agent for patients whose tumors express hyperactivated Akt.


Journal of Biological Chemistry | 2005

Molecular cloning and characterization of the human AKT1 promoter uncovers its up-regulation by the Src/Stat3 pathway.

Sungman Park; Donghwa Kim; Satoshi Kaneko; Kristen M. Szewczyk; Santo V. Nicosia; Hua Yu; Richard Jove; Jin Q. Cheng

Akt1, also known as protein kinase B (PKB) α, is frequently activated in human cancers and has been implicated in many cell processes by phosphorylation of downstream molecules. However, transcriptional regulation of Akt1 has not been documented. Here, we report the isolation and characterization of the human AKT1 promoter and demonstrate transcriptional up-regulation of AKT1 by the Src/Stat3 pathway. Protein and mRNA levels of AKT1 are elevated in cells expressing constitutively active Stat3 as well as in v-Src-transformed NIH3T3 cells. Knockdown of Stat3 reduces AKT1 expression induced by v-Src. Although the 4.2-kb region upstream of the transcription start site of the AKT1 promoter contains five putative Stat3-binding motifs, the promoter failed to be induced by Stat3 and/or Src. Further analysis reveals that major Stat3 response elements are located within exon 1 and intron 1 regions of the AKT1 gene, which is upstream of the AKT1 translation initiation site. In addition, ectopic expression of wild type AKT1 in Stat3-/- MEF cells largely rescues serum starvation-induced cell death. These findings indicate that the AKT1 promoter comprises exon 1 and intron 1, in addition to the sequence upstream of transcriptional start site. Our data further show that AKT1 is a direct target gene of Stat3 and contributes to Stat3 anti-apoptotic function.


PLOS ONE | 2010

Regulation of Proapoptotic Mammalian ste20–Like Kinase MST2 by the IGF1-Akt Pathway

Donghwa Kim; Shaokun Shu; Marc D. Coppola; Satoshi Kaneko; Zengqiang Yuan; Jin Q. Cheng

Background Hippo, a Drosophila serine/threonine kinase, promotes apoptosis and restricts cell growth and proliferation. Its mammalian homolog MST2 has been shown to play similar role and be regulated by Raf-1 via a kinase-independent mechanism and by RASSF family proteins through forming complex with MST2. However, regulation of MST2 by cell survival signal remains largely unknown. Methodology/Principal Findings Using immunoblotting, in vitro kinase and in vivo labeling assays, we show that IGF1 inhibits MST2 cleavage and activation induced by DNA damage through the phosphatidylinosotol 3-kinase (PI3K)/Akt pathway. Akt phosphorylates a highly conserved threonine-117 residue of MST2 in vitro and in vivo, which leads to inhibition of MST2 cleavage, nuclear translocation, autophosphorylation-Thr180 and kinase activity. As a result, MST2 proapoptotic and growth arrest function was significantly reduced. Further, inverse correlation between pMST2-T117/pAkt and pMST2-T180 was observed in human breast tumors. Conclusions/Significance Our findings demonstrate for the first time that extracellular cell survival signal IGF1 regulates MST2 and that Akt is a key upstream regulator of MST2.


Journal of Biological Chemistry | 2005

ArgBP2γ Interacts with Akt and p21-activated Kinase-1 and Promotes Cell Survival

Zengqiang Yuan; Donghwa Kim; Satoshi Kaneko; Melissa Sussman; Gary M. Bokoch; Gary D. Kruh; Santo V. Nicosia; Joseph R. Testa; Jin Q. Cheng

Akt/protein kinase B is a major cell survival pathway through phosphorylation of proapoptotic proteins Bad and Bax and of additional apoptotic pathways linked to Forkhead proteins glycogen synthase kinase-3β and ASK1. To further explore the mechanism by which Akt regulates cell survival, we identified an Akt interaction protein by yeast two-hybrid screening. It is highly homologous to ARG-binding protein 2 (ArgBP2) with splicing exon 8 of the coding region of the ArgBP2. As two splicing isoforms (ArgBP2α and -β) of ArgBP2 have been identified (Wang, B., Golemis, E. A., and Kruh, G. D. (1997) J. Biol. Chem. 272, 17542–17550), it was named ArgBP2γ. ArgBP2γ contains four Akt phosphorylation consensus sites, a SoHo motif, and three Src homology (SH) 3 domains and binds to C-terminal proline-rich motifs of Akt through its first and second SH3 domains. It also interacts with p21-activated protein kinase (PAK1) via its first and third SH3 domains, indicating the SH3 domains of ArgBP2γ as docking sites for Akt and PAK1. Akt phosphorylates ArgBP2γ in vitro and in vivo. Expression of ArgBP2γ induces PAK1 activity and overrides apoptosis induced by ectopic expression of Bad or DNA damage. Nonphosphorylatable ArgBP2γ-4A and SH3 domain-truncated mutant ArgBP2γ inhibit Akt-induced PAK1 activation and reduce Akt and PAK1 phosphorylation of Bad and antiapoptotic function. These data indicate that ArgBP2γ is a physiological substrate of Akt, functions as an adaptor for Akt and PAK1, and plays a role in Akt/PAK1 cell survival pathway.


World Journal of Gastroenterology | 2011

MicroRNAs in pancreatic ductal adenocarcinoma

Jong Y Park; James F. Helm; Domenico Coppola; Donghwa Kim; Mokenge P. Malafa; Seung Joon Kim

Ductal adenocarcinoma of the pancreas is a lethal cancer for which the only chance of long-term survival belongs to the patient with localized disease in whom a potentially curative resection can be done. Therefore, biomarkers for early detection and new therapeutic strategies are urgently needed. miRNAs are a recently discovered class of small endogenous non-coding RNAs of about 22 nucleotides that have gained attention for their role in downregulation of mRNA expression at the post-transcriptional level. miRNAs regulate proteins involved in critical cellular processes such as differentiation, proliferation, and apoptosis. Evidence suggests that deregulated miRNA expression is involved in carcinogenesis at many sites, including the pancreas. Aberrant expression of miRNAs may upregulate the expression of oncogenes or downregulate the expression of tumor suppressor genes, as well as play a role in other mechanisms of carcinogenesis. The purpose of this review is to summarize our knowledge of deregulated miRNA expression in pancreatic cancer and discuss the implication for potential translation of this knowledge into clinical practice.

Collaboration


Dive into the Donghwa Kim's collaboration.

Top Co-Authors

Avatar

Jin Q. Cheng

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Domenico Coppola

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jianping Guo

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sungman Park

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Satoshi Kaneko

University of the Ryukyus

View shared research outputs
Top Co-Authors

Avatar

Santo V. Nicosia

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Zengqiang Yuan

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Eric B. Haura

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jong Y. Park

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge