Network


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

Hotspot


Dive into the research topics where Karan Bhuripanyo is active.

Publication


Featured researches published by Karan Bhuripanyo.


ACS Chemical Biology | 2012

Specificity of the E1-E2-E3 enzymatic cascade for ubiquitin C-terminal sequences identified by phage display

Bo Zhao; Karan Bhuripanyo; Jeffrey R. Schneider; Keya Zhang; Hermann Schindelin; David L. Boone; Jun Yin

Ubiquitin (UB) is a protein modifier that regulates many essential cellular processes. To initiate protein modification by UB, the E1 enzyme activates the C-terminal carboxylate of UB to launch its transfer through the E1-E2-E3 cascade onto target proteins. In this study, we used phage display to profile the specificity of the two human E1 enzymes, Ube1 and Uba6, toward the C-terminal sequence of UB ending with (71)LRLRGG(76). Phage selection revealed that while Arg72 of UB is absolutely required for E1 recognition, UB residues at positions 71, 73, and 74 can be replaced with bulky aromatic side chains, and Gly75 of UB can be changed to Ser, Asp, and Asn for efficient E1 activation. We have thus found that the E1 enzymes have substantial promiscuity regarding the UB C-terminal sequence. The UB variants from phage selection can also be transferred from E1 to E2 enzymes; however, they are blocked from further transfer to the E3 enzymes. This suggests that the C-terminal sequence of UB is important for its discharge from E2 and subsequent transfer to E3. In addition, we observed that the Leu73Phe and Leu73Tyr single mutants of UB are resistant to cleavage by deubiquitinating enzymes (DUBs), although they can be assembled by the E1-E2-E3 cascade into poly-UB chains, thus indicating differences in UB C-terminal specificities between the E1 and DUBs. Consequently these UB mutants may provide stability to UB polymers attached to cellular proteins and facilitate the elucidation of the biological signals encoded in the UB chains.


Organic Letters | 2012

Inhibiting the Protein Ubiquitination Cascade by Ubiquitin-Mimicking Short Peptides

Bo Zhao; Chan Hee J. Choi; Karan Bhuripanyo; Eric B. Villhauer; Keya Zhang; Hermann Schindelin; Jun Yin

Short heptapeptides were identified to function as ubiquitin (UB) mimics that are activated by E1 and form thioester conjugates with E1, E2, and HECT type E3 enzymes. The activities (k(cat)/K(1/2)) of E1 with the UB-mimicking peptides are 130-1,400-fold higher than the equally long peptide with the native C-terminal sequence of UB. By forming covalent conjugates with E1, E2, and E3 enzymes, the UB-mimicking peptides can block the transfer of native UB through the cascade.


Chemistry & Biology | 2012

Orthogonal Ubiquitin Transfer through Engineered E1-E2 Cascades for Protein Ubiquitination

Bo Zhao; Karan Bhuripanyo; Keya Zhang; Hiroaki Kiyokawa; Hermann Schindelin; Jun Yin

Protein modification by ubiquitin (UB) controls diverse cellular processes. UB is conjugated to cellular proteins by sequential transfer through an E1-E2-E3 enzymatic cascade. The cross-activities of 2 E1s, 50 E2s and thousands of E3s encoded by the human genome make it difficult to identify the substrate proteins of a specific E3 enzyme in the cell. One way to solve this problem is to engineer an orthogonal UB transfer (OUT) cascade in which the engineered UB (xUB) is relayed by engineered E1, E2 and E3 enzymes (xE1, xE2, xE3) to modify the substrate proteins of a specific E3. Here, we use phage display and mutagenesis to construct xUB-xE1 and xE1-xE2 pairs that are orthogonal to the native E1 and E2 enzymes. Our work on engineering the UB transfer cascades will enable us to use OUT to map the signal transduction networks mediated by protein ubiquitination.


ChemBioChem | 2013

Phage display to identify Nedd8-mimicking peptides as inhibitors of the Nedd8 transfer cascade.

Bo Zhao; Keya Zhang; Eric B. Villhauer; Karan Bhuripanyo; Hiroaki Kiyokawa; Hermann Schindelin; Jun Yin

The Nedd8 activating enzyme (NAE) launches the transfer of the ubiquitin‐like protein Nedd8 through an enzymatic cascade to covalently modify a diverse array of proteins, thus regulating their biological functions in the cell. The C‐terminal peptide of Nedd8 extends deeply into the active site of NAE and plays an important role in the specific recognition of Nedd8 by NAE. We used phage display to profile C‐terminal mutant sequences of Nedd8 that could be recognized by NAE for the activation reaction. We found that NAE can accommodate diverse changes in the Nedd8 C‐terminal sequence (71LALRGG76), including Arg and Ile replacing Leu71, Leu, Ser, and Gln replacing Ala72, and substitutions by bulky aromatic residues at positions 73 and 74. We also observed that short peptides corresponding to the C‐terminal sequences of the Nedd8 variants can be activated by NAE to form peptide∼NAE thioester conjugates. Once NAE is covalently loaded with these Nedd8‐mimicking peptides, it can no longer activate full‐length Nedd8 for transfer to the neddylation targets, such as the cullin subunits of cullin‐RING E3 ubiquitin ligases (CRLs). We have thus developed a new method to inhibit protein neddylation by Nedd8‐mimicking peptides.


Nature Communications | 2017

Orthogonal ubiquitin transfer identifies ubiquitination substrates under differential control by the two ubiquitin activating enzymes

Xianpeng Liu; Bo Zhao; Limin Sun; Karan Bhuripanyo; Yiyang Wang; Yingtao Bi; Ramana V. Davuluri; Duc M. Duong; Dhaval Nanavati; Jun Yin; Hiroaki Kiyokawa

Protein ubiquitination is mediated sequentially by ubiquitin activating enzyme E1, ubiquitin conjugating enzyme E2 and ubiquitin ligase E3. Uba1 was thought to be the only E1 until the recent identification of Uba6. To differentiate the biological functions of Uba1 and Uba6, we applied an orthogonal ubiquitin transfer (OUT) technology to profile their ubiquitination targets in mammalian cells. By expressing pairs of an engineered ubiquitin and engineered Uba1 or Uba6 that were generated for exclusive interactions, we identified 697 potential Uba6 targets and 527 potential Uba1 targets with 258 overlaps. Bioinformatics analysis reveals substantial differences in pathways involving Uba1- and Uba6-specific targets. We demonstrate that polyubiquitination and proteasomal degradation of ezrin and CUGBP1 require Uba6, but not Uba1, and that Uba6 is involved in the control of ezrin localization and epithelial morphogenesis. These data suggest that distinctive substrate pools exist for Uba1 and Uba6 that reflect non-redundant biological roles for Uba6.


Nature Communications | 2017

Identifying the ubiquitination targets of E6AP by orthogonal ubiquitin transfer

Yiyang Wang; Xianpeng Liu; Li Zhou; Duc M. Duong; Karan Bhuripanyo; Bo Zhao; Han Zhou; Ruochuan Liu; Yingtao Bi; Hiroaki Kiyokawa; Jun Yin

E3 ubiquitin (UB) ligases are the ending modules of the E1–E2-E3 cascades that transfer UB to cellular proteins and regulate their biological functions. Identifying the substrates of an E3 holds the key to elucidate its role in cell regulation. Here, we construct an orthogonal UB transfer (OUT) cascade to identify the substrates of E6AP, a HECT E3 also known as Ube3a that is implicated in cancer and neurodevelopmental disorders. We use yeast cell surface display to engineer E6AP to exclusively transfer an affinity-tagged UB variant (xUB) to its substrate proteins. Proteomic identification of xUB-conjugated proteins in HEK293 cells affords 130 potential E6AP targets. Among them, we verify that MAPK1, CDK1, CDK4, PRMT5, β-catenin, and UbxD8 are directly ubiquitinated by E6AP in vitro and in the cell. Our work establishes OUT as an efficient platform to profile E3 substrates and reveal the cellular circuits mediated by the E3 enzymes.E3 ubiquitin ligases regulate biological functions by ubiquitinating defined substrate proteins but overlapping specificities complicate the identification of E3-substrate relationships. Here, the authors construct an orthogonal UB transfer cascade and identify specific substrates of the E3 enzyme E6AP.


ChemBioChem | 2014

SUMO-Mimicking Peptides Inhibiting Protein SUMOylation

Bo Zhao; Eric B. Villhauer; Karan Bhuripanyo; Hiroaki Kiyokawa; Hermann Schindelin; Jun Yin

The ubiquitin‐like protein SUMO is transferred through a core E1–E2 cascade composed of the SUMO‐activating enzyme (SAE) and Ubc9 to modify cellular proteins and transmit important biological signals. SAE primarily recognizes the C‐terminal tail of SUMO and catalyzes ATP condensation with the SUMO C‐terminal carboxylate to activate its transfer through the cascade. Here, we used phage display to show that a broad profile of SUMO C‐terminal sequences could be activated by SAE. Based on this, we developed heptamer peptides that could 1) form thioester conjugates with SAE, 2) be transferred from SAE to Ubc9, and 3) be further transferred to the SUMOylation target protein RanGAP1. As these peptides recapitulate the action of SUMO in protein modification, we refer to them as “SUMO‐mimicking peptides”. We found that, once the peptides were conjugated to SAE and Ubc9, they blocked full‐length SUMO from entering the cascade. These peptides can thus function as mechanism‐based inhibitors of the protein SUMOylation reaction.


Science Advances | 2018

Identifying the substrate proteins of U-box E3s E4B and CHIP by orthogonal ubiquitin transfer

Karan Bhuripanyo; Yiyang Wang; Xianpeng Liu; Li Zhou; Ruochuan Liu; Duc M. Duong; Bo Zhao; Yingtao Bi; Han Zhou; Geng Chen; Nicholas T. Seyfried; Walter J. Chazin; Hiroaki Kiyokawa; Jun Yin

Engineering the ubiquitin transfer cascades by phage display enables an efficient way to profile E3 substrates. E3 ubiquitin (UB) ligases E4B and carboxyl terminus of Hsc70-interacting protein (CHIP) use a common U-box motif to transfer UB from E1 and E2 enzymes to their substrate proteins and regulate diverse cellular processes. To profile their ubiquitination targets in the cell, we used phage display to engineer E2-E4B and E2-CHIP pairs that were free of cross-reactivity with the native UB transfer cascades. We then used the engineered E2-E3 pairs to construct “orthogonal UB transfer (OUT)” cascades so that a mutant UB (xUB) could be exclusively used by the engineered E4B or CHIP to label their substrate proteins. Purification of xUB-conjugated proteins followed by proteomics analysis enabled the identification of hundreds of potential substrates of E4B and CHIP in human embryonic kidney 293 cells. Kinase MAPK3 (mitogen-activated protein kinase 3), methyltransferase PRMT1 (protein arginine N-methyltransferase 1), and phosphatase PPP3CA (protein phosphatase 3 catalytic subunit alpha) were identified as the shared substrates of the two E3s. Phosphatase PGAM5 (phosphoglycerate mutase 5) and deubiquitinase OTUB1 (ovarian tumor domain containing ubiquitin aldehyde binding protein 1) were confirmed as E4B substrates, and β-catenin and CDK4 (cyclin-dependent kinase 4) were confirmed as CHIP substrates. On the basis of the CHIP-CDK4 circuit identified by OUT, we revealed that CHIP signals CDK4 degradation in response to endoplasmic reticulum stress.


Methods of Molecular Biology | 2015

Phage Selection Assisted by Sfp Phosphopantetheinyl Transferase-Catalyzed Site-Specific Protein Labeling

Bo Zhao; Keya Zhang; Karan Bhuripanyo; Yiyang Wang; Han Zhou; Mengnan Zhang; Jun Yin

Phosphopantetheinyl transferases (PPTase) Sfp and AcpS catalyze a highly efficient reaction that conjugates chemical probes of diverse structures to proteins. PPTases have been widely used for site-specific protein labeling and live cell imaging of the target proteins. Here we describe the use of PPTase-catalyzed protein labeling in protein engineering by facilitating high-throughput phage selection.


PLOS ONE | 2013

Profiling the cross reactivity of ubiquitin with the Nedd8 activating enzyme by phage display.

Bo Zhao; Keya Zhang; Karan Bhuripanyo; Chan Hee J. Choi; Eric B. Villhauer; Heng Li; Ning Zheng; Hiroaki Kiyokawa; Hermann Schindelin; Jun Yin

The C-terminal peptides of ubiquitin (UB) and UB-like proteins (UBLs) play a key role in their recognition by the specific activating enzymes (E1s) to launch their transfer through the respective enzymatic cascades thus modifying cellular proteins. UB and Nedd8, a UBL regulating the activity of cullin-RING UB ligases, only differ by one residue at their C-termini; yet each has its specific E1 for the activation reaction. It has been reported recently that UAE can cross react with Nedd8 to enable its passage through the UB transfer cascade for protein neddylation. To elucidate differences in UB recognition by UAE and NAE, we carried out phage selection of a UB library with randomized C-terminal sequences based on the catalytic formation of UB∼NAE thioester conjugates. Our results confirmed the previous finding that residue 72 of UB plays a “gate-keeping” role in E1 selectivity. We also found that diverse sequences flanking residue 72 at the UB C-terminus can be accommodated by NAE for activation. Furthermore heptameric peptides derived from the C-terminal sequences of UB variants selected for NAE activation can function as mimics of Nedd8 to form thioester conjugates with NAE and the downstream E2 enzyme Ubc12 in the Nedd8 transfer cascade. Once the peptides are charged onto the cascade enzymes, the full-length Nedd8 protein is effectively blocked from passing through the cascade for the critical modification of cullin. We have thus identified a new class of inhibitors of protein neddylation based on the profiles of the UB C-terminal sequences recognized by NAE.

Collaboration


Dive into the Karan Bhuripanyo's collaboration.

Top Co-Authors

Avatar

Jun Yin

University of Chicago

View shared research outputs
Top Co-Authors

Avatar

Bo Zhao

University of Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yiyang Wang

Georgia State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Han Zhou

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Xianpeng Liu

Northwestern University

View shared research outputs
Researchain Logo
Decentralizing Knowledge