Network


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

Hotspot


Dive into the research topics where Yanke Liang is active.

Publication


Featured researches published by Yanke Liang.


Nature Chemical Biology | 2014

A unique inhibitor binding site in ERK1/2 is associated with slow binding kinetics.

A. Chaikuad; Eliana Mc Tacconi; Jutta Zimmer; Yanke Liang; Nathanael S. Gray; Madalena Tarsounas; Stefan Knapp

Activation of the ERK pathway is a hallmark of cancer and targeting of upstream signalling partners led to the development of approved drugs. Recently SCH772984 has been shown to be a selective and potent ERK1/2 inhibitor. Here we report the structural mechanism for its remarkable selectivity. In ERK1/2, SCH772984 induced a so far unknown binding pocket that accommodated the piperazine-phenyl-pyrimidine decoration. This novel binding pocket was created by an inactive conformation of the phosphate binding loop and an outward tilt of helix αC. In contrast, structure determination of SCH772984 with the off-target haspin and JNK1 revealed canonical but two distinct type-I binding modes. Intriguingly, the novel binding mode with ERK1/2 was associated with slow binding kinetics in vitro as well as in cell based assay systems. The described binding mode of SCH772984 with ERK1/2 enables the design of a new type of specific kinase inhibitors with prolonged on-target activity.


Nature Chemical Biology | 2016

Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors.

Tinghu Zhang; Nicholas Kwiatkowski; Calla M Olson; Sarah E Dixon-Clarke; Brian J. Abraham; Ann Katrin Greifenberg; Scott B. Ficarro; J.M. Elkins; Yanke Liang; Nancy M. Hannett; Theresa Manz; Mingfeng Hao; Bartlomiej Bartkowiak; Arno L. Greenleaf; Jarrod A. Marto; Matthias Geyer; Alex N. Bullock; Richard A. Young; Nathanael S. Gray

Cyclin-dependent kinases 12 and 13 (CDK12 and CDK13) play critical roles in the regulation of gene transcription. However, the absence of CDK12 and CDK13 inhibitors has hindered the ability to investigate the consequences of their inhibition in healthy cells and cancer cells. Here we describe the rational design of a first-in-class CDK12 and CDK13 covalent inhibitor, THZ531. Co-crystallization of THZ531 with CDK12-cyclin K indicates that THZ531 irreversibly targets a cysteine located outside the kinase domain. THZ531 causes a loss of gene expression with concurrent loss of elongating and hyperphosphorylated RNA polymerase II. In particular, THZ531 substantially decreases the expression of DNA damage response genes and key super-enhancer-associated transcription factor genes. Coincident with transcriptional perturbation, THZ531 dramatically induced apoptotic cell death. Small molecules capable of specifically targeting CDK12 and CDK13 may thus help identify cancer subtypes that are particularly dependent on their kinase activities.


ACS Chemical Biology | 2014

Discovery of a Potent, Covalent BTK Inhibitor for B-Cell Lymphoma

Hong Wu; Wenchao Wang; Feiyang Liu; Ellen Weisberg; Bei Tian; Yongfei Chen; Binhua Li; Aoli Wang; Beilei Wang; Zheng Zhao; Douglas W. McMillin; Chen Hu; Hong Li; Jinhua Wang; Yanke Liang; Sara J. Buhrlage; Junting Liang; Jing Liu; Guang Yang; Jennifer R. Brown; Steven P. Treon; Constantine S. Mitsiades; James D. Griffin; Qingsong Liu; Nathanael S. Gray

BTK is a member of the TEC family of non-receptor tyrosine kinases whose deregulation has been implicated in a variety of B-cell-related diseases. We have used structure-based drug design in conjunction with kinome profiling and cellular assays to develop a potent, selective, and irreversible BTK kinase inhibitor, QL47, which covalently modifies Cys481. QL47 inhibits BTK kinase activity with an IC50 of 7 nM, inhibits autophosphorylation of BTK on Tyr223 in cells with an EC50 of 475 nM, and inhibits phosphorylation of a downstream effector PLCγ2 (Tyr759) with an EC50 of 318 nM. In Ramos cells QL47 induces a G1 cell cycle arrest that is associated with pronounced degradation of BTK protein. QL47 inhibits the proliferation of B-cell lymphoma cancer cell lines at submicromolar concentrations.


Nature Communications | 2016

SIKs control osteocyte responses to parathyroid hormone

Marc N. Wein; Yanke Liang; Olga Göransson; Thomas B. Sundberg; Jinhua Wang; Elizabeth A. Williams; Maureen J. O'Meara; Nicolas Govea; Belinda Beqo; Shigeki Nishimori; Kenichi Nagano; Daniel J. Brooks; Janaina S. Martins; Braden Corbin; Anthony Anselmo; Ruslan I. Sadreyev; Joy Y. Wu; Kei Sakamoto; Marc Foretz; Ramnik J. Xavier; Roland Baron; Mary L. Bouxsein; Thomas J. Gardella; Paola Divieti-Pajevic; Nathanael S. Gray; Henry M. Kronenberg

Parathyroid hormone (PTH) activates receptors on osteocytes to orchestrate bone formation and resorption. Here we show that PTH inhibition of SOST (sclerostin), a WNT antagonist, requires HDAC4 and HDAC5, whereas PTH stimulation of RANKL, a stimulator of bone resorption, requires CRTC2. Salt inducible kinases (SIKs) control subcellular localization of HDAC4/5 and CRTC2. PTH regulates both HDAC4/5 and CRTC2 localization via phosphorylation and inhibition of SIK2. Like PTH, new small molecule SIK inhibitors cause decreased phosphorylation and increased nuclear translocation of HDAC4/5 and CRTC2. SIK inhibition mimics many of the effects of PTH in osteocytes as assessed by RNA-seq in cultured osteocytes and following in vivo administration. Once daily treatment with the small molecule SIK inhibitor YKL-05-099 increases bone formation and bone mass. Therefore, a major arm of PTH signalling in osteocytes involves SIK inhibition, and small molecule SIK inhibitors may be applied therapeutically to mimic skeletal effects of PTH.


Leukemia | 2016

Discovery of a BTK/MNK dual inhibitor for lymphoma and leukemia.

Hong Wu; Chen Hu; Aoli Wang; Ellen Weisberg; Yuching Chen; C-H Yun; Wenchao Wang; Yan Liu; Xiaochuan Liu; B Tian; Joshua L. Wang; Zheng Zhao; Yanke Liang; Binhua Li; Li Wang; Beilei Wang; Cheng Chen; Sara J. Buhrlage; Ziping Qi; Fengming Zou; Atsushi Nonami; Yunzhan Li; Stacey M. Fernandes; Sophia Adamia; Richard Stone; Ilene Galinsky; Xuefu Wang; Guang Yang; James D. Griffin; Jennifer R. Brown

Bruton’s tyrosine kinase (BTK) kinase is a member of the TEC kinase family and is a key regulator of the B-cell receptor (BCR)-mediated signaling pathway. It is important for B-cell maturation, proliferation, survival and metastasis. Pharmacological inhibition of BTK is clinically effective against a variety of B-cell malignances, such as mantle cell lymphoma, chronic lymphocytic leukemia (CLL), acute myeloid leukemia (AML) and activated B-cell–diffuse large B-cell lymphoma. MNK kinase is one of the key downstream regulators in the RAF–MEK–ERK signaling pathway and controls protein synthesis via regulating the activity of eIF4E. Inhibition of MNK activity has been observed to moderately inhibit the proliferation of AML cells. Through a structure-based drug-design approach, we have discovered a selective and potent BTK/MNK dual kinase inhibitor (QL-X-138), which exhibits covalent binding to BTK and noncovalent binding to MNK. Compared with the BTK kinase inhibitor (PCI-32765) and the MNK kinase inhibitor (cercosporamide), QL-X-138 enhanced the antiproliferative efficacies in vitro against a variety of B-cell cancer cell lines, as well as AML and CLL primary patient cells, which respond moderately to BTK inhibitor in vitro. The agent can effectively arrest the growth of lymphoma and leukemia cells at the G0–G1 stage and can induce strong apoptotic cell death. These primary results demonstrate that simultaneous inhibition of BTK and MNK kinase activity might be a new therapeutic strategy for B-cell malignances.


ACS Chemical Biology | 2016

Development of Chemical Probes for Investigation of Salt-Inducible Kinase Function in Vivo.

Thomas B. Sundberg; Yanke Liang; Huixian Wu; Hwan Geun Choi; Nam Doo Kim; Taebo Sim; Liv Johannessen; Adam Petrone; Bernard Khor; Daniel B. Graham; Isabel Latorre; Andrew J. Phillips; Stuart L. Schreiber; Jose R. Perez; Alykhan F. Shamji; Nathanael S. Gray; Ramnik J. Xavier

Salt-inducible kinases (SIKs) are promising therapeutic targets for modulating cytokine responses during innate immune activation. The study of SIK inhibition in animal models of disease has been limited by the lack of selective small-molecule probes suitable for modulating SIK function in vivo. We used the pan-SIK inhibitor HG-9-91-01 as a starting point to develop improved analogs, yielding a novel probe 5 (YKL-05-099) that displays increased selectivity for SIKs versus other kinases and enhanced pharmacokinetic properties. Well-tolerated doses of YKL-05-099 achieve free serum concentrations above its IC50 for SIK2 inhibition for >16 h and reduce phosphorylation of a known SIK substrate in vivo. While in vivo active doses of YKL-05-099 recapitulate the effects of SIK inhibition on inflammatory cytokine responses, they did not induce metabolic abnormalities observed in Sik2 knockout mice. These results identify YKL-05-099 as a useful probe to investigate SIK function in vivo and further support the development of SIK inhibitors for treatment of inflammatory disorders.


Nature Chemical Biology | 2017

Pharmacological perturbation of CDK9 using selective CDK9 inhibition or degradation

Calla M Olson; Baishan Jiang; Michael A. Erb; Yanke Liang; Zainab M. Doctor; Zinan Zhang; Tinghu Zhang; Nicholas Kwiatkowski; Myriam Boukhali; Jennifer L. Green; Wilhelm Haas; Tyzoon K. Nomanbhoy; Eric S. Fischer; Richard A. Young; James E. Bradner; Georg E. Winter; Nathanael S. Gray

Cyclin-dependent kinase 9 (CDK9), an important regulator of transcriptional elongation, is a promising target for cancer therapy, particularly for cancers driven by transcriptional dysregulation. We characterized NVP-2, a selective ATP-competitive CDK9 inhibitor, and THAL-SNS-032, a selective CDK9 degrader consisting of a CDK-binding SNS-032 ligand linked to a thalidomide derivative that binds the E3 ubiquitin ligase Cereblon (CRBN). To our surprise, THAL-SNS-032 induced rapid degradation of CDK9 without affecting the levels of other SNS-032 targets. Moreover, the transcriptional changes elicited by THAL-SNS-032 were more like those caused by NVP-2 than those induced by SNS-032. Notably, compound washout did not significantly reduce levels of THAL-SNS-032-induced apoptosis, suggesting that CDK9 degradation had prolonged cytotoxic effects compared with CDK9 inhibition. Thus, our findings suggest that thalidomide conjugation represents a promising strategy for converting multi-targeted inhibitors into selective degraders and reveal that kinase degradation can induce distinct pharmacological effects compared with inhibition.


Cell Reports | 2017

A UV-Independent Topical Small-Molecule Approach for Melanin Production in Human Skin

Nisma Mujahid; Yanke Liang; Ryo Murakami; Hwan Geun Choi; Allison S. Dobry; Jinhua Wang; Yusuke Suita; Qing Yu Weng; Jennifer Allouche; Lajos Kemény; Andrea L. Hermann; Elisabeth Roider; Nathanael S. Gray; David E. Fisher

SUMMARY The presence of dark melanin (eumelanin) within human epidermis represents one of the strongest predictors of low skin cancer risk. Topical rescue of eumelanin synthesis, previously achieved in “redhaired” Mc1r-deficient mice, demonstrated significant protection against UV damage. However, application of a topical strategy for human skin pigmentation has not been achieved, largely due to the greater barrier function of human epidermis. Salt-inducible kinase (SIK) has been demonstrated to regulate MITF, the master regulator of pigment gene expression, through its effects on CRTC and CREB activity. Here, we describe the development of small-molecule SIK inhibitors that were optimized for human skin penetration, resulting in MITF upregulation and induction of melanogenesis. When topically applied, pigment production was induced in Mc1r-deficient mice and normal human skin. These findings demonstrate a realistic pathway toward UV-independent topical modulation of human skin pigmentation, potentially impacting UV protection and skin cancer risk.


Antiviral Research | 2017

Discovery of host-targeted covalent inhibitors of dengue virus.

Mélissanne de Wispelaere; Margot Carocci; Yanke Liang; Qingsong Liu; Eileen Sun; Michael L. Vetter; Jinhua Wang; Nathanael S. Gray; Priscilla L. Yang

&NA; We report here on an approach targeting the host reactive cysteinome to identify inhibitors of host factors required for the infectious cycle of Flaviviruses and other viruses. We used two parallel cellular phenotypic screens to identify a series of covalent inhibitors, exemplified by QL‐XII‐47, that are active against dengue virus. We show that the compounds effectively block viral protein expression and that this inhibition is associated with repression of downstream processes of the infectious cycle, and thus significantly contributes to the potent antiviral activity of these compounds. We demonstrate that QL‐XII‐47s antiviral activity requires selective, covalent modification of a host target by showing that the compounds antiviral activity is recapitulated when cells are preincubated with QL‐XII‐47 and then washed prior to viral infection and by showing that QL‐XII‐47R, a non‐reactive analog, lacks antiviral activity at concentrations more than 20‐fold higher than QL‐XII‐47s IC90. QL‐XII‐47s inhibition of Zika virus, West Nile virus, hepatitis C virus, and poliovirus further suggests that it acts via a target mediating inhibition of these other medically relevant viruses. These results demonstrate the utility of screens targeting the host reactive cysteinome for rapid identification of compounds with potent antiviral activity. HighlightsTargeting the host cysteinome leads to the discovery of broad‐spectrum antivirals.The covalent inhibitor QL‐XII‐47 potently inhibits dengue virus.QL‐XII‐47 and related compounds block viral protein expression.QL‐XII‐47 exhibits broad‐spectrum antiviral activity.


ACS Medicinal Chemistry Letters | 2017

Structure–Activity Relationship Study of QL47: A Broad-Spectrum Antiviral Agent

Yanke Liang; Mélissanne de Wispelaere; Margot Carocci; Qingsong Liu; Jinhua Wang; Priscilla L. Yang; Nathanael S. Gray

Here we report the structure-activity relationship (SAR) investigations of QL-XII-47 (QL47), a compound that possesses broad-spectrum antiviral activity against dengue virus and other RNA viruses. A medicinal chemistry campaign initiated from QL47, a previously reported covalent BTK inhibitor, to derive YKL-04-085, which is devoid of any kinase activity when screened against a panel of 468 kinases and with improved pharmacokinetic properties. Both QL47 and YKL-04-085 are potent inhibitors of viral translation and exhibit cellular antiviral activity at 35-fold lower concentrations relative to inhibition of host-cell proliferation.

Collaboration


Dive into the Yanke Liang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew J. Phillips

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard A. Young

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge