Network


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

Hotspot


Dive into the research topics where Yousang Hwang is active.

Publication


Featured researches published by Yousang Hwang.


Nature | 2008

The structural basis of protein acetylation by the p300/CBP transcriptional coactivator

Xin Liu; Ling Wang; Kehao Zhao; Paul R. Thompson; Yousang Hwang; Ronen Marmorstein; Philip A. Cole

The transcriptional coactivator p300/CBP (CREBBP) is a histone acetyltransferase (HAT) that regulates gene expression by acetylating histones and other transcription factors. Dysregulation of p300/CBP HAT activity contributes to various diseases including cancer. Sequence alignments, enzymology experiments and inhibitor studies on p300/CBP have led to contradictory results about its catalytic mechanism and its structural relation to the Gcn5/PCAF and MYST HATs. Here we describe a high-resolution X-ray crystal structure of a semi-synthetic heterodimeric p300 HAT domain in complex with a bi-substrate inhibitor, Lys-CoA. This structure shows that p300/CBP is a distant cousin of other structurally characterized HATs, but reveals several novel features that explain the broad substrate specificity and preference for nearby basic residues. Based on this structure and accompanying biochemical data, we propose that p300/CBP uses an unusual ‘hit-and-run’ (Theorell–Chance) catalytic mechanism that is distinct from other characterized HATs. Several disease-associated mutations can also be readily accounted for by the p300 HAT structure. These studies pave the way for new epigenetic therapies involving modulation of p300/CBP HAT activity.


Science | 2010

Glucose and Weight Control in Mice with a Designed Ghrelin O-Acyltransferase Inhibitor

Brad P. Barnett; Yousang Hwang; Martin S. Taylor; Henriette Kirchner; Paul T. Pfluger; Vincent Bernard; Yu Yi Lin; Erin M. Bowers; Chandrani Mukherjee; Woo Jin Song; Patti A. Longo; Daniel J. Leahy; Mehboob A. Hussain; Matthias H. Tschöp; Jef D. Boeke; Philip A. Cole

Metabolism Without Modification Obesity-associated metabolic disease has rapidly become a public health priority in the developed world and is being addressed through prevention strategies aimed at lifestyle changes and through pharmacological approaches. Barnett et al. (p. 1689, published online 18 November) designed a drug that inhibits the action of ghrelin, a circulating peptide hormone that increases fat mass and food intake. The drug, a bisubstrate analog called GO-CoA-Tat, is a selective antagonist of ghrelin O-acyltransferase (GOAT), an enzyme that catalyzes a posttranslational modification that is essential for ghrelin activity. Injection of GO-CoA-Tat into wild-type mice on a high-fat diet improved glucose tolerance and reduced weight gain, probably through changes in metabolic activity. Because GO-CoA-Tat is a peptide-based drug that requires repeated injection, it is unsuitable for clinical use, but GOAT does represent a potentially valuable target for future drug development efforts in metabolic disease. A drug inhibiting the activation of ghrelin, a peptide that promotes weight gain, has beneficial metabolic effects in mice. Ghrelin is a gastric peptide hormone that stimulates weight gain in vertebrates. The biological activities of ghrelin require octanoylation of the peptide on Ser3, an unusual posttranslational modification that is catalyzed by the enzyme ghrelin O-acyltransferase (GOAT). Here, we describe the design, synthesis, and characterization of GO-CoA-Tat, a peptide-based bisubstrate analog that antagonizes GOAT. GO-CoA-Tat potently inhibits GOAT in vitro, in cultured cells, and in mice. Intraperitoneal administration of GO-CoA-Tat improves glucose tolerance and reduces weight gain in wild-type mice but not in ghrelin-deficient mice, supporting the concept that its beneficial metabolic effects are due specifically to GOAT inhibition. In addition to serving as a research tool for mapping ghrelin actions, GO-CoA-Tat may help pave the way for clinical targeting of GOAT in metabolic diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Electrophilic tuning of the chemoprotective natural product sulforaphane

Young Hoon Ahn; Yousang Hwang; Hua Liu; Xiu Jun Wang; Ying Zhang; Katherine K. Stephenson; Tatiana Boronina; Robert N. Cole; Albena T. Dinkova-Kostova; Paul Talalay; Philip A. Cole

Sulforaphane [1-isothiocyanato-4-(methylsulfinyl)butane], a naturally occurring isothiocyanate derived from cruciferous vegetables, is a highly potent inducer of phase 2 cytoprotective enzymes and can protect against electrophiles including carcinogens, oxidative stress, and inflammation. The mechanism of action of sulforaphane is believed to involve modifications of critical cysteine residues of Keap1, which lead to stabilization of Nrf2 to activate the antioxidant response element of phase 2 enzymes. However, the dithiocarbamate functional group formed by a reversible reaction between isothiocyanate of sulforaphane and sulfhydryl nucleophiles of Keap1 is kinetically labile, and such modification in intact cells has not yet been demonstrated. Here we designed sulforaphane analogs with replacement of the reactive isothiocyanate by the more gentle electrophilic sulfoxythiocarbamate group that also selectively targets cysteine residues in proteins but forms stable thiocarbamate adducts. Twenty-four sulfoxythiocarbamate analogs were synthesized that retain the structural features important for high potency in sulforaphane analogs: the sulfoxide or keto group and its appropriate distance to electrophilic functional group. Evaluation in various cell lines including hepatoma cells, retinal pigment epithelial cells, and keratinocytes as well as in mouse skin shows that these analogs maintain high potency and efficacy for phase 2 enzyme induction as well as the inhibitory effect on lipopolysaccharide-induced nitric oxide formation like sulforaphane. We further show in living cells that a sulfoxythiocarbamate analog can label Keap1 on several key cysteine residues as well as other cellular proteins offering new insights into the mechanism of chemoprotection.


Journal of the American Chemical Society | 2010

Site-specific introduction of an acetyl-lysine mimic into peptides and proteins by cysteine alkylation.

Rong Huang; Marc A. Holbert; Mary Katherine Tarrant; Sandrine Curtet; David R. Colquhoun; Beverley M. Dancy; Blair C. R. Dancy; Yousang Hwang; Yong Tang; Katrina Meeth; Ronen Marmorstein; Robert N. Cole; Saadi Khochbin; Philip A. Cole

Protein acetylation on Lys residues is recognized as a significant post-translational modification in cells, but it is often difficult to discern the direct structural and functional effects of individual acetylation events. Here we describe a new tool, methylthiocarbonyl-aziridine, to install acetyl-Lys mimics site-specifically into peptides and proteins by alkylation of Cys residues. We demonstrate that the resultant thiocarbamate modification can be recognized by the Brdt bromodomain and site-specific antiacetyl-Lys antibodies, is resistant to histone deacetylase cleavage, and can confer activation of the histone acetyltransferase Rtt109 by simulating autoacetylation. We also use this approach to obtain functional evidence that acetylation of CK2 protein kinase on Lys102 can stimulate its catalytic activity.


Journal of Biological Chemistry | 2013

Architectural Organization of the Metabolic Regulatory Enzyme Ghrelin O-Acyltransferase

Martin S. Taylor; Travis R. Ruch; Po Yuan Hsiao; Yousang Hwang; Pingfeng Zhang; Lixin Dai; Cheng Ran Lisa Huang; Christopher E. Berndsen; Min Sik Kim; Akhilesh Pandey; Cynthia Wolberger; Ronen Marmorstein; Carolyn E. Machamer; Jef D. Boeke; Philip A. Cole

Background: Ghrelin O-acyltransferase (GOAT) is a membrane protein that is responsible for octanoylating the metabolism-regulating peptide hormone ghrelin. Results: We have used a combination of approaches to determine the topology of GOAT. Conclusion: We have shown that GOAT has 11 transmembrane-spanning domains and one reentrant loop. Significance: These findings serve as a reference for other membrane-bound O-acyltransferase family members. Ghrelin O-acyltransferase (GOAT) is a polytopic integral membrane protein required for activation of ghrelin, a secreted metabolism-regulating peptide hormone. Although GOAT is a potential therapeutic target for the treatment of obesity and diabetes and plays a key role in other physiologic processes, little is known about its structure or mechanism. GOAT is a member of the membrane-bound O-acyltransferase (MBOAT) family, a group of polytopic integral membrane proteins involved in lipid-biosynthetic and lipid-signaling reactions from prokaryotes to humans. Here we use phylogeny and a variety of bioinformatic tools to predict the topology of GOAT. Using selective permeabilization indirect immunofluorescence microscopy in combination with glycosylation shift immunoblotting, we demonstrate that GOAT contains 11 transmembrane helices and one reentrant loop. Development of the V5Glyc tag, a novel, small, and sensitive dual topology reporter, facilitated these experiments. The MBOAT family invariant residue His-338 is in the ER lumen, consistent with other family members, but conserved Asn-307 is cytosolic, making it unlikely that both are involved in catalysis. Photocross-linking of synthetic ghrelin analogs and inhibitors demonstrates binding to the C-terminal region of GOAT, consistent with a role of His-338 in the active site. This knowledge of GOAT architecture is important for a deeper understanding of the mechanism of GOAT and other MBOATs and could ultimately advance the discovery of selective inhibitors for these enzymes.


Methods in Enzymology | 2012

Ghrelin O-acyltransferase assays and inhibition.

Martin S. Taylor; Yousang Hwang; Po Yuan Hsiao; Jef D. Boeke; Philip A. Cole

Ghrelin O-acyltransferase (GOAT) is responsible for catalyzing the attachment of the eight-carbon fatty acid octanoyl to the Ser3 side chain of the peptide ghrelin to generate the active form of this metabolic hormone. As such, GOAT is viewed as a potential therapeutic target for the treatment of obesity and diabetes mellitus. Here, we review recent progress in the development of cell and in vitro assays to measure GOAT action and the identification of several synthetic GOAT inhibitors. In particular, we discuss the design, synthesis, and characterization of the bisubstrate analog GO-CoA-Tat and its ability to modulate weight and blood glucose in mice. We also highlight current challenges and future research directions in our biomedical understanding of this fascinating ghrelin processing enzyme.


Nature Methods | 2016

Enzyme-catalyzed expressed protein ligation

Samuel Henager; Nam Chu; Zan Chen; David M. Bolduc; Daniel R. Dempsey; Yousang Hwang; James A. Wells; Philip A. Cole

Expressed protein ligation is a valuable method for protein semisynthesis that involves the reaction of recombinant protein C-terminal thioesters with N-terminal cysteine (N-Cys)-containing peptides, but the requirement of a Cys residue at the ligation junction can limit the utility of this method. Here we employ subtiligase variants to efficiently ligate Cys-free peptides to protein thioesters. Using this method, we have more accurately determined the effect of C-terminal phosphorylation on the tumor suppressor protein PTEN.


Expert Opinion on Therapeutic Patents | 2010

METHODS FOR SYNTHESIS AND USES OF INHIBITORS OF GHRELIN O-ACYLTRANSFERASE AS POTENTIAL THERAPEUTIC AGENTS FOR OBESITY AND DIABETES

Philip A. Cole; Bradley Powers Barnett; Yousang Hwang; Jef D. Boeke

The patent claims inhibitors of ghrelin-O-acyltransferase enzyme. These inhibitors were designed as bisubstrate analogs of the enzymatic reaction and were coupled with one of the peptides able to cross cell membranes, the 11-mer Tat peptide. The compound GO-CoA-Tat was studied in vitro and in vivo; after intraperitoneal administration, it enhances insulin response to a glucose load and leads to a statistically significant weight loss in mouse fed with a high fat diet.


Bioorganic Chemistry | 2015

Mechanistic analysis of ghrelin-O-acyltransferase using substrate analogs.

Martin S. Taylor; Daniel R. Dempsey; Yousang Hwang; Zan Chen; Nam Chu; Jef D. Boeke; Philip A. Cole

Ghrelin-O-Acyltransferase (GOAT) is an 11-transmembrane integral membrane protein that octanoylates the metabolism-regulating peptide hormone ghrelin at Ser3 and may represent an attractive target for the treatment of type II diabetes and the metabolic syndrome. Protein octanoylation is unique to ghrelin in humans, and little is known about the mechanism of GOAT or of related protein-O-acyltransferases HHAT or PORC. In this study, we explored an in vitro microsomal ghrelin octanoylation assay to analyze its enzymologic features. Measurement of Km for 10-mer, 27-mer, and synthetic Tat-peptide-containing ghrelin substrates provided evidence for a role of charge interactions in substrate binding. Ghrelin substrates with amino-alanine in place of Ser3 demonstrated that GOAT can catalyze the formation of an octanoyl-amide bond at a similar rate compared with the natural reaction. A pH-rate comparison of these substrates revealed minimal differences in acyltransferase activity across pH 6.0-9.0, providing evidence that these reactions may be relatively insensitive to the basicity of the substrate nucleophile. The conserved His338 residue was required both for Ser3 and amino-Ala3 ghrelin substrates, suggesting that His338 may have a key catalytic role beyond that of a general base.


Journal of the American Chemical Society | 2018

Hydrazide Mimics for Protein Lysine Acylation to Assess Nucleosome Dynamics and Deubiquitinase Action

Shridhar Bhat; Yousang Hwang; Matthew D. Gibson; Michael T Morgan; Sean D. Taverna; Yingming Zhao; Cynthia Wolberger; Michael G. Poirier; Philip A. Cole

A range of acyl-lysine (acyl-Lys) modifications on histones and other proteins have been mapped over the past decade but for most, their functional and structural significance remains poorly characterized. One limitation in the study of acyl-Lys containing proteins is the challenge of producing them or their mimics in site-specifically modified forms. We describe a cysteine alkylation-based method to install hydrazide mimics of acyl-Lys post-translational modifications (PTMs) on proteins. We have applied this method to install mimics of acetyl-Lys, 2-hydroxyisobutyryl-Lys, and ubiquityl-Lys that could be recognized selectively by relevant acyl-Lys modification antibodies. The acyl-Lys modified histone H3 proteins were reconstituted into nucleosomes to study nucleosome dynamics and stability as a function of modification type and site. We also installed a ubiquityl-Lys mimic in histone H2B and generated a diubiquitin analog, both of which could be cleaved by deubiquitinating enzymes. Nucleosomes containing the H2B ubiquityl-Lys mimic were used to study the SAGA deubiquitinating modules molecular recognition. These results suggest that acyl-Lys mimics offer a relatively simple and promising strategy to study the role of acyl-Lys modifications in the function, structure, and regulation of proteins and protein complexes.

Collaboration


Dive into the Yousang Hwang's collaboration.

Top Co-Authors

Avatar

Philip A. Cole

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel R. Dempsey

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Nam Chu

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ronen Marmorstein

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Zan Chen

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Cynthia Wolberger

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

James A. Wells

University of California

View shared research outputs
Top Co-Authors

Avatar

Paul R. Thompson

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge