Network


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

Hotspot


Dive into the research topics where Wayne Grant is active.

Publication


Featured researches published by Wayne Grant.


Nature | 2011

A stress response pathway regulates DNA damage through β2-adrenoreceptors and β-arrestin-1.

Makoto R. Hara; Jeffrey J. Kovacs; Erin J. Whalen; Sudarshan Rajagopal; Ryan T. Strachan; Wayne Grant; Aaron J. Towers; Barbara Williams; Christopher M. Lam; Kunhong Xiao; Sudha K. Shenoy; Simon G. Gregory; Seungkirl Ahn; Derek R. Duckett; Robert J. Lefkowitz

The human mind and body respond to stress, a state of perceived threat to homeostasis, by activating the sympathetic nervous system and secreting the catecholamines adrenaline and noradrenaline in the ‘fight-or-flight’ response. The stress response is generally transient because its accompanying effects (for example, immunosuppression, growth inhibition and enhanced catabolism) can be harmful in the long term. When chronic, the stress response can be associated with disease symptoms such as peptic ulcers or cardiovascular disorders, and epidemiological studies strongly indicate that chronic stress leads to DNA damage. This stress-induced DNA damage may promote ageing, tumorigenesis, neuropsychiatric conditions and miscarriages. However, the mechanisms by which these DNA-damage events occur in response to stress are unknown. The stress hormone adrenaline stimulates β2-adrenoreceptors that are expressed throughout the body, including in germline cells and zygotic embryos. Activated β2-adrenoreceptors promote Gs-protein-dependent activation of protein kinase A (PKA), followed by the recruitment of β-arrestins, which desensitize G-protein signalling and function as signal transducers in their own right. Here we elucidate a molecular mechanism by which β-adrenergic catecholamines, acting through both Gs–PKA and β-arrestin-mediated signalling pathways, trigger DNA damage and suppress p53 levels respectively, thus synergistically leading to the accumulation of DNA damage. In mice and in human cell lines, β-arrestin-1 (ARRB1), activated via β2-adrenoreceptors, facilitates AKT-mediated activation of MDM2 and also promotes MDM2 binding to, and degradation of, p53, by acting as a molecular scaffold. Catecholamine-induced DNA damage is abrogated in Arrb1-knockout (Arrb1−/−) mice, which show preserved p53 levels in both the thymus, an organ that responds prominently to acute or chronic stress, and in the testes, in which paternal stress may affect the offspring’s genome. Our results highlight the emerging role of ARRB1 as an E3-ligase adaptor in the nucleus, and reveal how DNA damage may accumulate in response to chronic stress.


Journal of Medicinal Chemistry | 2010

Tetrahydroisoquinoline derivatives as highly selective and potent rho kinase inhibitors

Xingang Fang; Yan Yin; Yen Ting Chen; Lei Yao; Bo Wang; Michael D. Cameron; Li Lin; Susan Khan; Claudia Ruiz; Thomas Schröter; Wayne Grant; Amiee Weiser; Jennifer Pocas; Alok Pachori; Stephans Schürer; Philip V. LoGrasso; Yangbo Feng

Rho kinase (ROCK) is a promising drug target for the treatment of many diseases including hypertension, multiple sclerosis, cancer, and glaucoma. The structure-activity relationships (SAR) around a series of tetrahydroisoquinolines were evaluated utilizing biochemical and cell-based assays to measure ROCK inhibition. These novel ROCK inhibitors possess high potency, high selectivity, and appropriate pharmacokinetic properties for glaucoma applications. The lead compound, 35, had subnanomolar potency in enzyme ROCK-II assays as well as excellent cell-based potency (IC(50) = 51 nM). In a kinase panel profiling, 35 had an off-target hit rate of only 1.6% against 442 kinases. Pharmacology studies showed that compound 35 was efficacious in reducing intraocular pressure (IOP) in rats with reasonably long duration of action. These results suggest that compound 35 may serve as a promising agent for further development in the treatment of glaucoma.


Bioorganic & Medicinal Chemistry Letters | 2009

Benzothiazoles as Rho-associated kinase (ROCK-II) inhibitors

Yan Yin; Li Lin; Claudia Ruiz; Michael D. Cameron; Jennifer Pocas; Wayne Grant; Thomas Schröter; Weimin Chen; Derek R. Duckett; Stephan C. Schürer; Philip V. LoGrasso; Yangbo Feng

A series of benzothiazole derivatives as ROCK inhibitors have been discovered. Compounds with good biochemical and cellular potency, and sufficient kinase selectivity have been identified.


ACS Medicinal Chemistry Letters | 2010

Discovery of Potent and Selective Urea-Based ROCK Inhibitors and Their Effects on Intraocular Pressure in Rats

Yan Yin; Michael D. Cameron; Li Lin; Susan Khan; Thomas Schröter; Wayne Grant; Jennifer Pocas; Yen Ting Chen; Stephan C. Schürer; Alok Pachori; Philip V. LoGrasso; Yangbo Feng

A series of urea-based Rho kinase (ROCK) inhibitors were designed and evaluated. The discovered compounds had excellent enzyme and cellular potency, high kinase selectivity, high aqueous solubility, good porcine corneal penetration, and appropriate DMPK profiles for topical applications as antiglaucoma therapeutics.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery and optimization of indoles and 7-azaindoles as Rho kinase (ROCK) inhibitors (part-I).

Sarwat Chowdhury; E. Hampton Sessions; Jennifer Pocas; Wayne Grant; Thomas Schröter; Li Lin; Claudia Ruiz; Michael D. Cameron; Stephan C. Schürer; Philip V. LoGrasso; Thomas D. Bannister; Yangbo Feng

Rho kinase (ROCK) inhibitors are potential therapeutic agents to treat disorders such as hypertension, multiple sclerosis, cancers, and glaucoma. Here, we disclose the synthesis, optimization, biological evaluation of potent indole and 7-azaindole based ROCK inhibitors that have high potency on ROCK (IC(50)=1 nM) with 740-fold selectivity over PKA (47). Moreover, 47 showed very good DMPK properties making it a good candidate for further development. Finally, docking studies with a homology model of ROCK-II were performed to rationalize the binding mode of these compounds and showed the compounds bound in both orientations to take advantage to H-bonds with Lys-121 of ROCK-II.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and biological evaluation of 4-quinazolinones as Rho kinase inhibitors

Xingang Fang; Yen Ting Chen; E. Hampton Sessions; Sarwat Chowdhury; Tomas Vojkovsky; Yan Yin; Jennifer Pocas; Wayne Grant; Thomas Schröter; Li Lin; Claudia Ruiz; Michael D. Cameron; Philip V. LoGrasso; Thomas D. Bannister; Yangbo Feng

Rho kinase (ROCK) is an attractive therapeutic target for various diseases including glaucoma, hypertension, and spinal cord injury. Herein, we report the development of a series of ROCK-II inhibitors based on 4-quinazolinone and quinazoline scaffolds. SAR studies at three positions of the quinazoline core led to the identification of analogs with high potency against ROCK-II and good selectivity over protein kinase A (PKA).


Journal of Medicinal Chemistry | 2013

Synthesis and biological evaluation of urea derivatives as highly potent and selective rho kinase inhibitors.

Yan Yin; Li Lin; Claudia Ruiz; Susan Khan; Michael D. Cameron; Wayne Grant; Jennifer Pocas; Nibal Eid; HaJeung Park; Thomas Schröter; Philip V. LoGrasso; Yangbo Feng

RhoA and its downstream effector ROCK mediate stress fiber formation and cell contraction through their effects on the phosphorylation of myosin light chain (MLC). Inhibition of the RhoA/ROCK pathway has proven to be a promising strategy for several indications such as cardiovascular disease, glaucoma, and inflammatory disease. In 2010, our group reported urea-based ROCK inhibitors as potential antiglaucoma agents. These compounds showed potent IC50 values in enzymatic and cell-based assays and significant intraocular pressure (IOP)-lowering effects in rats (∼7 mmHg). (22) To develop more advanced ROCK inhibitors targeting various potential applications (such as myocardial infarction, erectile dysfunction, multiple sclerosis, etc.) in addition to glaucoma, a thorough SAR for this urea-based scaffold was studied. The detailed optimization process, counter-screening, and in vitro and in vivo DMPK studies are discussed. Potent and selective ROCK inhibitors with various in vivo pharmacokinetic properties were discovered.


Bioorganic & Medicinal Chemistry Letters | 2013

Amino acid derived quinazolines as Rock/PKA inhibitors.

Sarwat Chowdhury; Yen Ting Chen; Xingang Fang; Wayne Grant; Jennifer Pocas; Michael D. Cameron; Claudia Ruiz; Li Lin; HaJeung Park; Thomas Schröter; Thomas D. Bannister; Philip V. LoGrasso; Yangbo Feng

SAR and lead optimization studies for Rock inhibitors based on amino acid-derived quinazolines are described. Studies demonstrated that these amino acid derived quinazolinones were mainly pan-Rock (I & II) inhibitors. While selectivity against other kinases could be achieved, selectivity for most of these compounds against PKA was not achieved. This is distinct from Rock inhibitors based on non-amino acid derived quinazolinones, where high selectivity against PKA could be obtained.(22) The inhibitors presented here in some cases possessed sub-nanomolar inhibition of Rock, nanomolar potency in ppMLC cell based assays, low to fair cytochrome P-450 inhibition, and good human microsomal stability.


MedChemComm | 2011

Asymmetric synthesis of potent chroman-based Rho kinase (ROCK-II) inhibitors

Yen Ting Chen; Tomas Vojkovsky; Xingang Fang; Jennifer Pocas; Wayne Grant; Amiee M. W. Handy; Thomas Schröter; Philip V. LoGrasso; Thomas D. Bannister; Yangbo Feng

Rho kinase (ROCK) is currently investigated as a target for various diseases such as glaucoma and spinal cord injury. Herein, we report the asymmetric synthesis of chroman 1, a highly potent ROCK inhibitor, and its analogs. The inhibitory properties of these compounds for ROCK-II and a selected set of highly homologous kinases are also discussed.


Science Translational Medicine | 2015

Therapeutic targeting of casein kinase 1δ in breast cancer

Laura H. Rosenberg; Marie Lafitte; Victor Quereda; Wayne Grant; Weimin Chen; Mathieu Bibian; Yoshihiko Noguchi; Mohammad Fallahi; Chunying Yang; Jenny C. Chang; William R. Roush; John L. Cleveland; Derek R. Duckett

Casein kinase 1δ promotes breast tumorigenesis by activation of β-catenin and can be targeted in some breast cancers. New way to target breast cancer Casein kinase 1δ (CK1δ) is a protein that helps regulate a variety of cellular processes and is overexpressed in a large percentage of breast and other cancers. Rosenberg et al. show that CK1δ contributes to the growth of breast tumors by activating the Wnt signaling pathway, which is carcinogenic, and that it can be inhibited by RNA interference or by a small-molecule drug. Treatment with a small-molecule inhibitor of CK1δ, called SR-3029, was effective at reducing the growth of breast cancer in multiple mouse models without any signs of major toxicity, suggesting that this therapeutic approach may be a good candidate for testing in human patients. Identification of specific drivers of human cancer is required to instruct the development of targeted therapeutics. We demonstrate that CSNK1D is amplified and/or overexpressed in human breast tumors and that casein kinase 1δ (CK1δ) is a vulnerability of human breast cancer subtypes overexpressing this kinase. Specifically, selective knockdown of CK1δ, or treatment with a highly selective and potent CK1δ inhibitor, triggers apoptosis of CK1δ-expressing breast tumor cells ex vivo, tumor regression in orthotopic models of triple-negative breast cancer, including patient-derived xenografts, and tumor growth inhibition in human epidermal growth factor receptor 2–positive (HER2+) breast cancer models. We also show that Wnt/β-catenin signaling is a hallmark of human tumors overexpressing CK1δ, that disabling CK1δ blocks nuclear accumulation of β-catenin and T cell factor transcriptional activity, and that constitutively active β-catenin overrides the effects of inhibition or silencing of CK1δ. Thus, CK1δ inhibition represents a promising strategy for targeted treatment in human breast cancer with Wnt/β-catenin involvement.

Collaboration


Dive into the Wayne Grant's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennifer Pocas

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Schröter

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Yangbo Feng

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Claudia Ruiz

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Derek R. Duckett

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Li Lin

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Yan Yin

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Yen Ting Chen

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge