23 results on '"Hamman BD"'
Search Results
2. PROTACs Targeting BRM (SMARCA2) Afford Selective In Vivo Degradation over BRG1 (SMARCA4) and Are Active in BRG1 Mutant Xenograft Tumor Models.
- Author
-
Berlin M, Cantley J, Bookbinder M, Bortolon E, Broccatelli F, Cadelina G, Chan EW, Chen H, Chen X, Cheng Y, Cheung TK, Davenport K, DiNicola D, Gordon D, Hamman BD, Harbin A, Haskell R, He M, Hole AJ, Januario T, Kerry PS, Koenig SG, Li L, Merchant M, Pérez-Dorado I, Pizzano J, Quinn C, Rose CM, Rousseau E, Soto L, Staben LR, Sun H, Tian Q, Wang J, Wang W, Ye CS, Ye X, Zhang P, Zhou Y, Yauch R, and Dragovich PS
- Subjects
- Humans, Proteolysis Targeting Chimera, Heterografts, Cell Line, Transcription Factors genetics, DNA Helicases genetics, Nuclear Proteins genetics, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms genetics
- Abstract
The identification of VHL-binding proteolysis targeting chimeras (PROTACs) that potently degrade the BRM protein (also known as SMARCA2) in SW1573 cell-based experiments is described. These molecules exhibit between 10- and 100-fold degradation selectivity for BRM over the closely related paralog protein BRG1 (SMARCA4). They also selectively impair the proliferation of the H1944 "BRG1-mutant" NSCLC cell line, which lacks functional BRG1 protein and is thus highly dependent on BRM for growth, relative to the wild-type Calu6 line. In vivo experiments performed with a subset of compounds identified PROTACs that potently and selectively degraded BRM in the Calu6 and/or the HCC2302 BRG1 mutant NSCLC xenograft models and also afforded antitumor efficacy in the latter system. Subsequent PK/PD analysis established a need to achieve strong BRM degradation (>95%) in order to trigger meaningful antitumor activity in vivo . Intratumor quantitation of mRNA associated with two genes whose transcription was controlled by BRM ( PLAU and KRT80 ) also supported this conclusion.
- Published
- 2024
- Full Text
- View/download PDF
3. Selective PROTAC-mediated degradation of SMARCA2 is efficacious in SMARCA4 mutant cancers.
- Author
-
Cantley J, Ye X, Rousseau E, Januario T, Hamman BD, Rose CM, Cheung TK, Hinkle T, Soto L, Quinn C, Harbin A, Bortolon E, Chen X, Haskell R, Lin E, Yu SF, Del Rosario G, Chan E, Dunlap D, Koeppen H, Martin S, Merchant M, Grimmer M, Broccatelli F, Wang J, Pizzano J, Dragovich PS, Berlin M, and Yauch RL
- Subjects
- Animals, Humans, Proteolysis, Mutation, Mammals, Transcription Factors genetics, DNA Helicases genetics, Nuclear Proteins genetics, Neoplasms genetics
- Abstract
The mammalian SWItch/Sucrose Non-Fermentable (SWI/SNF) helicase SMARCA4 is frequently mutated in cancer and inactivation results in a cellular dependence on its paralog, SMARCA2, thus making SMARCA2 an attractive synthetic lethal target. However, published data indicates that achieving a high degree of selective SMARCA2 inhibition is likely essential to afford an acceptable therapeutic index, and realizing this objective is challenging due to the homology with the SMARCA4 paralog. Herein we report the discovery of a potent and selective SMARCA2 proteolysis-targeting chimera molecule (PROTAC), A947. Selective SMARCA2 degradation is achieved in the absence of selective SMARCA2/4 PROTAC binding and translates to potent in vitro growth inhibition and in vivo efficacy in SMARCA4 mutant models, compared to wild type models. Global ubiquitin mapping and proteome profiling reveal no unexpected off-target degradation related to A947 treatment. Our study thus highlights the ability to transform a non-selective SMARCA2/4-binding ligand into a selective and efficacious in vivo SMARCA2-targeting PROTAC, and thereby provides a potential new therapeutic opportunity for patients whose tumors contain SMARCA4 mutations., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
4. Bicyclic Heterocyclic Replacement of an Aryl Amide Leading to Potent and Kinase-Selective Adaptor Protein 2-Associated Kinase 1 Inhibitors.
- Author
-
Hartz RA, Ahuja VT, Nara SJ, Kumar CMV, Manepalli RKVLP, Sarvasiddhi SK, Honkhambe S, Patankar V, Dasgupta B, Rajamani R, Muckelbauer JK, Camac DM, Ghosh K, Pokross M, Kiefer SE, Brown JM, Hunihan L, Gulianello M, Lewis M, Lippy JS, Surti N, Hamman BD, Allen J, Kostich WA, Bronson JJ, Macor JE, and Dzierba CD
- Subjects
- Amides pharmacology, Amides therapeutic use, Animals, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Quinazolines therapeutic use, Structure-Activity Relationship, Neuralgia drug therapy, Quinolines pharmacology, Quinolines therapeutic use
- Abstract
Adaptor protein 2-associated kinase 1 (AAK1) is a serine/threonine kinase that was identified as a therapeutic target for the potential treatment of neuropathic pain. Inhibition of AAK1 in the central nervous system, particularly within the spinal cord, was found to be the relevant site for achieving an antinociceptive effect. We previously reported that compound 7 is a brain-penetrant, AAK1 inhibitor that showed efficacy in animal models for neuropathic pain. One approach we took to improve upon the potency of 7 involved tying the amide back into the neighboring phenyl ring to form a bicyclic heterocycle. Investigation of the structure-activity relationships (SARs) of substituents on the resultant quinazoline and quinoline ring systems led to the identification of ( S )-31 , a brain-penetrant, AAK1-selective inhibitor with improved enzyme and cellular potency compared to 7 . The synthesis, SAR, and in vivo evaluation of a series of quinazoline and quinoline-based AAK1 inhibitors are described herein.
- Published
- 2022
- Full Text
- View/download PDF
5. Discovery, Structure-Activity Relationships, and In Vivo Evaluation of Novel Aryl Amides as Brain Penetrant Adaptor Protein 2-Associated Kinase 1 (AAK1) Inhibitors for the Treatment of Neuropathic Pain.
- Author
-
Hartz RA, Ahuja VT, Nara SJ, Kumar CMV, Brown JM, Bristow LJ, Rajamani R, Muckelbauer JK, Camac D, Kiefer SE, Hunihan L, Gulianello M, Lewis M, Easton A, Lippy JS, Surti N, Pattipati SN, Dokania M, Elavazhagan S, Dandapani K, Hamman BD, Allen J, Kostich W, Bronson JJ, Macor JE, and Dzierba CD
- Subjects
- Amides chemical synthesis, Amides chemistry, Animals, Anti-Inflammatory Agents, Non-Steroidal chemical synthesis, Anti-Inflammatory Agents, Non-Steroidal chemistry, Caco-2 Cells, Dose-Response Relationship, Drug, Drug Discovery, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Microsomes, Liver chemistry, Microsomes, Liver metabolism, Molecular Structure, Neuralgia metabolism, Protein Kinases chemical synthesis, Protein Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Structure-Activity Relationship, Amides pharmacology, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Brain enzymology, Neuralgia drug therapy, Protein Kinases pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Effective treatment of chronic pain, in particular neuropathic pain, without the side effects that often accompany currently available treatment options is an area of significant unmet medical need. A phenotypic screen of mouse gene knockouts led to the discovery that adaptor protein 2-associated kinase 1 (AAK1) is a potential therapeutic target for neuropathic pain. The synthesis and optimization of structure-activity relationships of a series of aryl amide-based AAK1 inhibitors led to the identification of 59 , a brain penetrant, AAK1-selective inhibitor that proved to be a valuable tool compound. Compound 59 was evaluated in mice for the inhibition of μ2 phosphorylation. Studies conducted with 59 in pain models demonstrated that this compound was efficacious in the phase II formalin model for persistent pain and the chronic-constriction-injury-induced model for neuropathic pain in rats. These results suggest that AAK1 inhibition is a promising approach for the treatment of neuropathic pain.
- Published
- 2021
- Full Text
- View/download PDF
6. Differential PROTAC substrate specificity dictated by orientation of recruited E3 ligase.
- Author
-
Smith BE, Wang SL, Jaime-Figueroa S, Harbin A, Wang J, Hamman BD, and Crews CM
- Subjects
- Humans, Models, Molecular, Molecular Structure, Protein Domains, Proteolysis drug effects, Small Molecule Libraries chemistry, Substrate Specificity, Ubiquitin-Protein Ligases chemistry, Von Hippel-Lindau Tumor Suppressor Protein chemistry, p38 Mitogen-Activated Protein Kinases chemistry, p38 Mitogen-Activated Protein Kinases metabolism, Small Molecule Libraries pharmacology, Ubiquitin-Protein Ligases metabolism, Ubiquitination drug effects, Von Hippel-Lindau Tumor Suppressor Protein metabolism
- Abstract
PROteolysis-TArgeting Chimeras (PROTACs) are hetero-bifunctional molecules that recruit an E3 ubiquitin ligase to a given substrate protein resulting in its targeted degradation. Many potent PROTACs with specificity for dissimilar targets have been developed; however, the factors governing degradation selectivity within closely-related protein families remain elusive. Here, we generate isoform-selective PROTACs for the p38 MAPK family using a single warhead (foretinib) and recruited E3 ligase (von Hippel-Lindau). Based on their distinct linker attachments and lengths, these two PROTACs differentially recruit VHL, resulting in degradation of p38α or p38δ. We characterize the role of ternary complex formation in driving selectivity, showing that it is necessary, but insufficient, for PROTAC-induced substrate ubiquitination. Lastly, we explore the p38δ:PROTAC:VHL complex to explain the different selectivity profiles of these PROTACs. Our work attributes the selective degradation of two closely-related proteins using the same warhead and E3 ligase to heretofore underappreciated aspects of the ternary complex model.
- Published
- 2019
- Full Text
- View/download PDF
7. Identification and Characterization of Von Hippel-Lindau-Recruiting Proteolysis Targeting Chimeras (PROTACs) of TANK-Binding Kinase 1.
- Author
-
Crew AP, Raina K, Dong H, Qian Y, Wang J, Vigil D, Serebrenik YV, Hamman BD, Morgan A, Ferraro C, Siu K, Neklesa TK, Winkler JD, Coleman KG, and Crews CM
- Subjects
- Cell Line, Cell Proliferation drug effects, Chemistry Techniques, Synthetic, Fluorescence Polarization, Genes, ras, Humans, I-kappa B Kinase genetics, I-kappa B Kinase metabolism, Molecular Structure, Mutation, Protein Serine-Threonine Kinases genetics, RNA Interference, Structure-Activity Relationship, Von Hippel-Lindau Tumor Suppressor Protein chemistry, Von Hippel-Lindau Tumor Suppressor Protein genetics, Protein Serine-Threonine Kinases metabolism, Proteolysis drug effects, Von Hippel-Lindau Tumor Suppressor Protein metabolism
- Abstract
Proteolysis targeting chimeras (PROTACs) are bifunctional molecules that recruit an E3 ligase to a target protein to facilitate ubiquitination and subsequent degradation of that protein. While the field of targeted degraders is still relatively young, the potential for this modality to become a differentiated and therapeutic reality is strong, such that both academic and pharmaceutical institutions are now entering this interesting area of research. In this article, we describe a broadly applicable process for identifying degrader hits based on the serine/threonine kinase TANK-binding kinase 1 (TBK1) and have generalized the key structural elements associated with degradation activities. Compound 3i is a potent hit (TBK1 DC
50 = 12 nM, Dmax = 96%) with excellent selectivity against a related kinase IKKε, which was further used as a chemical tool to assess TBK1 as a target in mutant K-Ras cancer cells.- Published
- 2018
- Full Text
- View/download PDF
8. Lessons in PROTAC Design from Selective Degradation with a Promiscuous Warhead.
- Author
-
Bondeson DP, Smith BE, Burslem GM, Buhimschi AD, Hines J, Jaime-Figueroa S, Wang J, Hamman BD, Ishchenko A, and Crews CM
- Subjects
- Humans, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Proteolysis drug effects, Small Molecule Libraries chemical synthesis, Small Molecule Libraries chemistry, Ubiquitin-Protein Ligases metabolism, Ubiquitination drug effects, Drug Design, Proteasome Endopeptidase Complex metabolism, Protein Kinase Inhibitors pharmacology, Small Molecule Libraries pharmacology, Ubiquitin-Protein Ligases antagonists & inhibitors
- Abstract
Inhibiting protein function selectively is a major goal of modern drug discovery. Here, we report a previously understudied benefit of small molecule proteolysis-targeting chimeras (PROTACs) that recruit E3 ubiquitin ligases to target proteins for their ubiquitination and subsequent proteasome-mediated degradation. Using promiscuous CRBN- and VHL-recruiting PROTACs that bind >50 kinases, we show that only a subset of bound targets is degraded. The basis of this selectivity relies on protein-protein interactions between the E3 ubiquitin ligase and the target protein, as illustrated by engaged proteins that are not degraded as a result of unstable ternary complexes with PROTAC-recruited E3 ligases. In contrast, weak PROTAC:target protein affinity can be stabilized by high-affinity target:PROTAC:ligase trimer interactions, leading to efficient degradation. This study highlights design guidelines for generating potent PROTACs as well as possibilities for degrading undruggable proteins immune to traditional small-molecule inhibitors., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
9. Inhibition of AAK1 Kinase as a Novel Therapeutic Approach to Treat Neuropathic Pain.
- Author
-
Kostich W, Hamman BD, Li YW, Naidu S, Dandapani K, Feng J, Easton A, Bourin C, Baker K, Allen J, Savelieva K, Louis JV, Dokania M, Elavazhagan S, Vattikundala P, Sharma V, Das ML, Shankar G, Kumar A, Holenarsipur VK, Gulianello M, Molski T, Brown JM, Lewis M, Huang Y, Lu Y, Pieschl R, O'Malley K, Lippy J, Nouraldeen A, Lanthorn TH, Ye G, Wilson A, Balakrishnan A, Denton R, Grace JE, Lentz KA, Santone KS, Bi Y, Main A, Swaffield J, Carson K, Mandlekar S, Vikramadithyan RK, Nara SJ, Dzierba C, Bronson J, Macor JE, Zaczek R, Westphal R, Kiss L, Bristow L, Conway CM, Zambrowicz B, and Albright CF
- Subjects
- Animals, Electrophysiological Phenomena drug effects, Gene Knockout Techniques, HEK293 Cells, Humans, Male, Mice, Neuralgia metabolism, Neuralgia physiopathology, Nociception drug effects, Phenotype, Protein Kinase Inhibitors therapeutic use, Protein Serine-Threonine Kinases deficiency, Protein Serine-Threonine Kinases genetics, Rats, Spinal Cord drug effects, Spinal Cord enzymology, Spinal Cord physiopathology, Neuralgia drug therapy, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
To identify novel targets for neuropathic pain, 3097 mouse knockout lines were tested in acute and persistent pain behavior assays. One of the lines from this screen, which contained a null allele of the adapter protein-2 associated kinase 1 (AAK1) gene, had a normal response in acute pain assays (hot plate, phase I formalin), but a markedly reduced response to persistent pain in phase II formalin. AAK1 knockout mice also failed to develop tactile allodynia following the Chung procedure of spinal nerve ligation (SNL). Based on these findings, potent, small-molecule inhibitors of AAK1 were identified. Studies in mice showed that one such inhibitor, LP-935509, caused a reduced pain response in phase II formalin and reversed fully established pain behavior following the SNL procedure. Further studies showed that the inhibitor also reduced evoked pain responses in the rat chronic constriction injury (CCI) model and the rat streptozotocin model of diabetic peripheral neuropathy. Using a nonbrain-penetrant AAK1 inhibitor and local administration of an AAK1 inhibitor, the relevant pool of AAK1 for antineuropathic action was found to be in the spinal cord. Consistent with these results, AAK1 inhibitors dose-dependently reduced the increased spontaneous neural activity in the spinal cord caused by CCI and blocked the development of windup induced by repeated electrical stimulation of the paw. The mechanism of AAK1 antinociception was further investigated with inhibitors of α2 adrenergic and opioid receptors. These studies showed that α2 adrenergic receptor inhibitors, but not opioid receptor inhibitors, not only prevented AAK1 inhibitor antineuropathic action in behavioral assays, but also blocked the AAK1 inhibitor-induced reduction in spinal neural activity in the rat CCI model. Hence, AAK1 inhibitors are a novel therapeutic approach to neuropathic pain with activity in animal models that is mechanistically linked (behaviorally and electrophysiologically) to α2 adrenergic signaling, a pathway known to be antinociceptive in humans., (Copyright © 2016 The Author(s).)
- Published
- 2016
- Full Text
- View/download PDF
10. Discovery and Development of LX7101, a Dual LIM-Kinase and ROCK Inhibitor for the Treatment of Glaucoma.
- Author
-
Harrison BA, Almstead ZY, Burgoon H, Gardyan M, Goodwin NC, Healy J, Liu Y, Mabon R, Marinelli B, Samala L, Zhang Y, Stouch TR, Whitlock NA, Gopinathan S, McKnight B, Wang S, Patel N, Wilson AG, Hamman BD, Rice DS, and Rawlins DB
- Abstract
The structure of LX7101, a dual LIM-kinase and ROCK inhibitor for the treatment of ocular hypertension and associated glaucoma, is disclosed. Previously reported LIM kinase inhibitors suffered from poor aqueous stability due to solvolysis of the central urea. Replacement of the urea with a hindered amide resulted in aqueous stable compounds, and addition of solubilizing groups resulted in a set of compounds with good properties for topical dosing in the eye and good efficacy in a mouse model of ocular hypertension. LX7101 was selected as a clinical candidate from this group based on superior efficacy in lowering intraocular pressure and a good safety profile. LX7101 completed IND enabling studies and was tested in a Phase 1 clinical trial in glaucoma patients, where it showed efficacy in lowering intraocular pressure.
- Published
- 2014
- Full Text
- View/download PDF
11. Discovery of a Type III Inhibitor of LIM Kinase 2 That Binds in a DFG-Out Conformation.
- Author
-
Goodwin NC, Cianchetta G, Burgoon HA, Healy J, Mabon R, Strobel ED, Allen J, Wang S, Hamman BD, and Rawlins DB
- Abstract
The first allosteric, type III inhibitor of LIM-kinase 2 (LIMK2) is reported. A series of molecules that feature both an N-phenylsulfonamide and tertiary amide were not only very potent at LIMK2 but also were extremely selective against a panel of other kinases. Enzymatic kinetic studies showed these molecules to be noncompetitive with ATP, suggesting allosteric inhibition. X-ray crystallography confirmed that these sulfonamides are a rare example of a type III kinase inhibitor that binds away from the highly conserved hinge region and instead resides in the hydrophobic pocket formed in the DFG-out conformation of the kinase, thus accounting for the high level of selectivity observed.
- Published
- 2014
- Full Text
- View/download PDF
12. Genetic deletion of Mst1 alters T cell function and protects against autoimmunity.
- Author
-
Salojin KV, Hamman BD, Chang WC, Jhaver KG, Al-Shami A, Crisostomo J, Wilkins C, Digeorge-Foushee AM, Allen J, Patel N, Gopinathan S, Zhou J, Nouraldeen A, Jessop TC, Bagdanoff JT, Augeri DJ, Read R, Vogel P, Swaffield J, Wilson A, Platt KA, Carson KG, Main A, Zambrowicz BP, and Oravecz T
- Subjects
- Animals, Arthritis, Rheumatoid immunology, Base Sequence, DNA Primers, Lymphocyte Activation, Lymphocyte Culture Test, Mixed, Mice, Mice, Inbred BALB C, Reverse Transcriptase Polymerase Chain Reaction, Autoimmunity, Hepatocyte Growth Factor genetics, Proto-Oncogene Proteins genetics, T-Lymphocytes immunology
- Abstract
Mammalian sterile 20-like kinase 1 (Mst1) is a MAPK kinase kinase kinase which is involved in a wide range of cellular responses, including apoptosis, lymphocyte adhesion and trafficking. The contribution of Mst1 to Ag-specific immune responses and autoimmunity has not been well defined. In this study, we provide evidence for the essential role of Mst1 in T cell differentiation and autoimmunity, using both genetic and pharmacologic approaches. Absence of Mst1 in mice reduced T cell proliferation and IL-2 production in vitro, blocked cell cycle progression, and elevated activation-induced cell death in Th1 cells. Mst1 deficiency led to a CD4+ T cell development path that was biased toward Th2 and immunoregulatory cytokine production with suppressed Th1 responses. In addition, Mst1-/- B cells showed decreased stimulation to B cell mitogens in vitro and deficient Ag-specific Ig production in vivo. Consistent with altered lymphocyte function, deletion of Mst1 reduced the severity of experimental autoimmune encephalomyelitis (EAE) and protected against collagen-induced arthritis development. Mst1-/- CD4+ T cells displayed an intrinsic defect in their ability to respond to encephalitogenic antigens and deletion of Mst1 in the CD4+ T cell compartment was sufficient to alleviate CNS inflammation during EAE. These findings have prompted the discovery of novel compounds that are potent inhibitors of Mst1 and exhibit desirable pharmacokinetic properties. In conclusion, this report implicates Mst1 as a critical regulator of adaptive immune responses, Th1/Th2-dependent cytokine production, and as a potential therapeutic target for immune disorders.
- Published
- 2014
- Full Text
- View/download PDF
13. A high-throughput screen for receptor protein tyrosine phosphatase-gamma selective inhibitors.
- Author
-
Appiah KK, Kostich WA, Gerritz SW, Huang Y, Hamman BD, Allen J, Zhang W, Lanthorn TH, Albright CF, Westphal R, Banks MN, and O'Connell JC
- Subjects
- Dimethyl Sulfoxide pharmacology, Enzyme Inhibitors chemistry, Enzyme Stability drug effects, Reproducibility of Results, Research Design, Sensitivity and Specificity, Solvents pharmacology, Drug Discovery methods, Enzyme Inhibitors metabolism, High-Throughput Screening Assays, Receptor-Like Protein Tyrosine Phosphatases, Class 5 antagonists & inhibitors
- Abstract
Protein tyrosine phosphatase-γ (PTP-γ) is a receptor-like PTP whose biological function is poorly understood. A recent mouse PTP-γ genetic deletion model associated the loss of PTP-γ gene expression with a potential antidepressant phenotype. This led the authors to screen a subset of the Bristol-Myers Squibb (BMS) compound collection to identify selective small-molecule inhibitors of receptor-like PTP-γ (RPTP-γ) for use in evaluating enzyme function in vivo. Here, they report the design of a high-throughput fluorescence resonance energy transfer (FRET) assay based on the Z'-LYTE technology to screen for inhibitors of RPTP-γ. A subset of the BMS diverse compound collection was screened and several compounds identified as RPTP-γ inhibitors in the assay. After chemical triage and clustering, compounds were assessed for potency and selectivity by IC(50) determination with RPTP-γ and two other phosphatases, PTP-1B and CD45. One hundred twenty-nine RPTP-γ selective (defined as IC(50) value greater than 5- to 10-fold over PTP-1B and CD45) inhibitors were identified and prioritized for evaluation. One of these hits, 3-(3, 4-dichlorobenzylthio) thiophene-2-carboxylic acid, was the primary chemotype for the initiation of a medicinal chemistry program.
- Published
- 2011
- Full Text
- View/download PDF
14. Novel class of LIM-kinase 2 inhibitors for the treatment of ocular hypertension and associated glaucoma.
- Author
-
Harrison BA, Whitlock NA, Voronkov MV, Almstead ZY, Gu KJ, Mabon R, Gardyan M, Hamman BD, Allen J, Gopinathan S, McKnight B, Crist M, Zhang Y, Liu Y, Courtney LF, Key B, Zhou J, Patel N, Yates PW, Liu Q, Wilson AG, Kimball SD, Crosson CE, Rice DS, and Rawlins DB
- Subjects
- Administration, Topical, Animals, Antihypertensive Agents chemistry, Antihypertensive Agents pharmacology, Glaucoma drug therapy, Glaucoma physiopathology, Guanidines chemical synthesis, Guanidines chemistry, Guanidines pharmacology, In Vitro Techniques, Intraocular Pressure drug effects, Mice, Nitriles chemical synthesis, Nitriles chemistry, Nitriles pharmacology, Ocular Hypertension chemically induced, Ocular Hypertension physiopathology, Piperazines chemical synthesis, Piperazines chemistry, Piperazines pharmacology, Pyrimidines chemistry, Pyrimidines pharmacology, Pyrroles chemistry, Pyrroles pharmacology, Structure-Activity Relationship, Swine, Urea analogs & derivatives, Urea chemical synthesis, Urea chemistry, Urea pharmacology, Antihypertensive Agents chemical synthesis, Lim Kinases antagonists & inhibitors, Ocular Hypertension drug therapy, Pyrimidines chemical synthesis, Pyrroles chemical synthesis
- Abstract
The discovery of a pyrrolopyrimidine class of LIM-kinase 2 (LIMK2) inhibitors is reported. These LIMK2 inhibitors show good potency in enzymatic and cellular assays and good selectivity against ROCK. After topical dosing to the eye in a steroid induced mouse model of ocular hypertension, the compounds reduce intraocular pressure to baseline levels. The compounds also increase outflow facility in a pig eye perfusion assay. These results suggest LIMK2 may be an effective target for treating ocular hypertension and associated glaucoma.
- Published
- 2009
- Full Text
- View/download PDF
15. A homogeneous G protein-coupled receptor ligand binding assay based on time-resolved fluorescence resonance energy transfer.
- Author
-
Hu LA, Zhou T, Hamman BD, and Liu Q
- Subjects
- Cell Line, Cell Membrane drug effects, Cell Membrane metabolism, Chelating Agents chemistry, Complement C5a chemistry, Cyclic AMP metabolism, Humans, Ligands, Radioligand Assay, Receptor, Anaphylatoxin C5a antagonists & inhibitors, Terbium chemistry, Fluorescence Resonance Energy Transfer methods, Receptors, G-Protein-Coupled metabolism
- Abstract
Fluorescence resonance energy transfer (FRET) has emerged as a powerful tool to the study of protein-protein interactions, such as receptor-ligand binding. However, the application of FRET to the study of G protein-coupled receptors (GPCRs) has been limited by the method of labeling receptor with fluorescence probes. Here we described a novel time-resolved (TR)-FRET method to study GPCR-ligand binding by using human complement 5a (C5a) receptor (C5aR) as a model system. Human C5aR was expressed in human embryonic kidney 293 cells with a hemagglutinin (HA) epitope at the N-terminus. Purified human C5a was labeled with terbium chelate and used as the fluorescence donor. Monoclonal anti-HA antibody conjugated with Alexa Fluor 488 was used as the fluorescence acceptor. Robust FRET signal was observed when the labeled ligand and C5aR membrane were mixed in the presence of the conjugated anti-HA antibody. This FRET signal was specific and saturable. C5a binding affinity to C5aR measured by the FRET assay was consistent with the data as determined by competition binding analysis using radiolabeled C5a. The FRET assay was also used to determine affinity of C5aR antagonists by competition binding analysis, and the data are similar to those from radioligand binding studies. Compared to the commonly used radioligand binding assay, this TR-FRET-based assay provides a nonradioactive, faster, and sensitive homogeneous assay format that could be easily adapted to high-throughput screening. The principle of this assay should also be applicable to other GPCRs, especially to those receptors with peptide or protein ligands.
- Published
- 2008
- Full Text
- View/download PDF
16. A FRET-based assay platform for ultra-high density drug screening of protein kinases and phosphatases.
- Author
-
Rodems SM, Hamman BD, Lin C, Zhao J, Shah S, Heidary D, Makings L, Stack JH, and Pollok BA
- Subjects
- Adenosine Triphosphate physiology, Animals, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Fluorescence, Humans, Isoquinolines pharmacology, Marine Toxins, Microcystins, Peptides, Cyclic pharmacology, Phosphoprotein Phosphatases antagonists & inhibitors, Protein Kinase Inhibitors, Protein Kinases physiology, Signal Transduction physiology, Staurosporine pharmacology, Vanadates pharmacology, Biological Assay instrumentation, Drug Evaluation, Preclinical instrumentation, Peptide Library, Phosphoprotein Phosphatases chemistry, Protein Kinases chemistry, Sulfonamides
- Abstract
Protein phosphorylation is one of the major regulatory mechanisms involved in signal-induced cellular events, including cell proliferation, apoptosis, and metabolism. Because many facets of biology are regulated by protein phosphorylation, aberrant kinase and/or phosphatase activity forms the basis for many different types of pathology. The disease relevance of protein kinases and phosphatases has led many pharmaceutical and biotechnology companies to expend significant resources in lead discovery programs for these two target classes. The existence of >500 kinases and phosphatases encoded by the human genome necessitates development of methodologies for the rapid screening for novel and specific compound inhibitors. We describe here a fluorescence-based, molecular assay platform that is compatible with robotic, ultra-high throughput screening systems and can be applied to virtually all tyrosine and serine/threonine protein kinases and phosphatases. The assay has a coupled-enzyme format, utilizing the differential protease sensitivity of phosphorylated versus nonphosphorylated peptide substrates. In addition to screening individual kinases, the assay can be formatted such that kinase pathways are re-created in vitro to identify compounds that specifically interact with inactive kinases. Miniaturization of this assay format to the 1-microl scale allows for the rapid and accurate compound screening of a host of kinase and phosphatase targets, thereby facilitating the hunt for new leads for these target classes.
- Published
- 2002
- Full Text
- View/download PDF
17. Binding of a Pleckstrin homology domain protein to phosphoinositide in membranes: a miniaturized FRET-based assay for drug screening.
- Author
-
Hamman BD, Pollok BA, Bennett T, Allen J, and Heim R
- Subjects
- Agammaglobulinaemia Tyrosine Kinase, Animals, Binding, Competitive, Dose-Response Relationship, Drug, Glutathione Transferase metabolism, Green Fluorescent Proteins, Humans, Kinetics, Lipid Metabolism, Luminescent Proteins metabolism, Mice, Mutagenesis, Site-Directed, Protein Binding, Protein Structure, Tertiary, Protein-Tyrosine Kinases metabolism, Recombinant Fusion Proteins metabolism, Time Factors, Blood Proteins chemistry, Cell Membrane metabolism, Drug Evaluation, Preclinical, Phosphatidylinositols metabolism, Phosphoproteins chemistry, Spectrometry, Fluorescence methods
- Abstract
Pleckstrin homology (PH) domains are present in key proteins involved in many vital cell processes. For example, the PH domain of Bruton's tyrosine kinase (Btk) binds to phosphatidylinositol triphosphate (PIP(3)) in the plasma membrane after stimulation of the B-cell receptor in B cells. Mutations in the Btk PH domain result in changes in its affinity for PIP(3), with higher binding leading to cell transformation in vitro and lower binding leading to antibody deficiencies in both humans and mice. We describe here a fluorescence resonance energy transfer (FRET)-based biochemical assay that directly monitors the interaction of a PH domain with PIP(3) at a membrane surface. We overexpressed a fusion protein consisting of an enhanced green fluorescent protein (GFP) and the N-terminal 170 amino acids of a Tec family kinase that contains its PH domain (PH170). Homogeneous unilamellar vesicles were made that contained PIP(3) and octadecylrhodamine (OR), a lipophilic FRET acceptor for GFP. After optimization of both protein and vesicle components, we found that binding of the GFP-PH170 protein to PIP3 in vesicles that contain OR results in about a 90% reduction of GFP fluorescence. Using this assay to screen 1440 compounds, we identified three that efficiently inhibited binding of GFP-PH170 to PIP(3) in vesicles. This biochemical assay readily miniaturized to 1.8-microl reaction volumes and was validated in a 3456-well screening format.
- Published
- 2002
- Full Text
- View/download PDF
18. Identification of a membrane-spanning domain of the thiol-activated pore-forming toxin Clostridium perfringens perfringolysin O: an alpha-helical to beta-sheet transition identified by fluorescence spectroscopy.
- Author
-
Shepard LA, Heuck AP, Hamman BD, Rossjohn J, Parker MW, Ryan KR, Johnson AE, and Tweten RK
- Subjects
- Amino Acid Sequence, Clostridium perfringens, Fluorescence Polarization, Fluorescent Dyes, Hemolysin Proteins metabolism, Humans, Light, Membrane Proteins metabolism, Molecular Sequence Data, Mutagenesis, Site-Directed, Oxadiazoles metabolism, Peptide Fragments metabolism, Phospholipids metabolism, Scattering, Radiation, Spectrometry, Fluorescence, Spin Labels, Bacterial Toxins chemistry, Bacterial Toxins metabolism, Membrane Proteins chemistry, Peptide Fragments chemistry, Protein Structure, Secondary, Sulfhydryl Compounds pharmacology
- Abstract
Clostridium perfringens perfringolysin O (PFO or theta-toxin) is a cytolytic toxin that binds to cholesterol-containing membranes and then self-associates to spontaneously form aqueous pores of varying size in the bilayer. In this study, a membrane-spanning domain has been identified in PFO by a combination of fluorescence spectroscopic methods using the fluorescent dye N, N'-dimethyl-N-(iodoacetyl)-N'-(7-nitrobenz-2-oxa-1, 3-diazolyl)ethylenediamine (NBD) whose emission properties are sensitive to water. PFO was substituted with a single cysteine at most of the residues between amino acids K189 and N218, and then each cysteine was modified with NBD. Each purified NBD-labeled PFO was then bound to membranes, and the probe's environment was ascertained by measuring its fluorescence lifetime, emission intensity, and collisional quenching with either aqueous (iodide ions) or nonaqueous (nitroxide-labeled phospholipids) quenchers. Lifetime and intensity measurements revealed that the amino acid side chains in this region of the membrane-bound PFO polypeptide alternated between being in an aqueous or a nonaqueous environment. This pattern indicates that this portion of the membrane-bound PFO spans the membrane in an antiparallel beta-sheet conformation. The alternating exposure of these residues to the hydrophobic interior of the bilayer was demonstrated by their susceptibility to quenching by nitroxide moieties attached to phospholipid acyl chains. Residues K189-N218 therefore form a two-stranded, amphipathic beta-sheet in the membrane-bound PFO that creates a stable interface between the pore and the membrane. This same region packs as three short alpha-helices in the soluble, monomeric form of PFO, and therefore, the cholesterol-dependent conversion of PFO to a membrane-bound oligomer involves a major structural transition in which three alpha-helices unfold to form a membrane-spanning amphipathic beta-sheet.
- Published
- 1998
- Full Text
- View/download PDF
19. BiP maintains the permeability barrier of the ER membrane by sealing the lumenal end of the translocon pore before and early in translocation.
- Author
-
Hamman BD, Hendershot LM, and Johnson AE
- Subjects
- Adenosine Diphosphate pharmacology, Adenosine Triphosphate pharmacology, Animals, Biological Transport drug effects, Biological Transport physiology, Cell Fractionation, Fungal Proteins genetics, Fungal Proteins isolation & purification, HSP70 Heat-Shock Proteins genetics, Iodides pharmacokinetics, Ion Channel Gating drug effects, Ion Channel Gating physiology, Membrane Proteins chemistry, Microsomes chemistry, Microsomes metabolism, NAD pharmacokinetics, RNA, Messenger physiology, Rabbits, Ribosomes metabolism, Solubility, Water metabolism, Yeasts chemistry, Yeasts metabolism, Endoplasmic Reticulum chemistry, Endoplasmic Reticulum metabolism, Fungal Proteins metabolism, HSP70 Heat-Shock Proteins metabolism, Membrane Proteins metabolism
- Abstract
Secretory proteins are cotranslationally translocated across the mammalian ER membrane through an aqueous pore in the translocon while the permeability barrier is maintained by a tight ribosome-membrane junction. The lumenal end of the pore is also blocked early in translocation. Extraction of soluble lumenal proteins from microsomes and reconstitution with purified proteins demonstrate, by fluorescence collisional quenching, that BiP seals the lumenal end of this pore. BiP also seals translocons that are assembled but are not engaged in translocation. These ribosome-free translocons have smaller pores (9-15 A diameter versus 40-60 A in functioning translocons) and are generated when ribosomes dissociate from functioning translocons with large pores. BiP therefore maintains the permeability barrier by sealing both nontranslocating and newly targeted translocons.
- Published
- 1998
- Full Text
- View/download PDF
20. The aqueous pore through the translocon has a diameter of 40-60 A during cotranslational protein translocation at the ER membrane.
- Author
-
Hamman BD, Chen JC, Johnson EE, and Johnson AE
- Subjects
- 4-Chloro-7-nitrobenzofurazan, Animals, Biological Transport, Active, Cytosol metabolism, Fluorescent Dyes, Immunoglobulin Fab Fragments chemistry, Immunoglobulin Fab Fragments metabolism, Intracellular Membranes metabolism, Lipid Bilayers metabolism, Membrane Proteins genetics, Models, Molecular, NAD metabolism, Permeability, Protein Biosynthesis, Protein Conformation, RNA, Transfer, Lys metabolism, Rabbits, Saccharomyces cerevisiae metabolism, Water metabolism, Endoplasmic Reticulum metabolism, Membrane Proteins metabolism
- Abstract
Eukaryotic secretory proteins are cotranslationally translocated through the endoplasmic reticulum (ER) membrane via aqueous pores that span the lipid bilayer. Fluorescent probes were incorporated into nascent secretory proteins using modified Lys-tRNAs, and the resulting nascent chains were sealed off from the cytosol in fully assembled translocation intermediates. Fluorescence quenching agents of different sizes were then introduced into the ER lumen in order to determine which were small enough to enter the pore and to quench the fluorescence of probes inside the ribosome and/or the pore. These accessibility studies showed that the aqueous pore in a functioning translocon is 40-60 A in diameter, making it the largest hole observed to date in a membrane that must maintain a permeability barrier.
- Published
- 1997
- Full Text
- View/download PDF
21. Dimer/monomer equilibrium and domain separations of Escherichia coli ribosomal protein L7/L12.
- Author
-
Hamman BD, Oleinikov AV, Jokhadze GG, Traut RR, and Jameson DM
- Subjects
- Bacterial Proteins genetics, Bacterial Proteins isolation & purification, Cysteine chemistry, Dimerization, Escherichia coli genetics, Fluorescein, Fluoresceins, Fluorescence Polarization, Genetic Variation, Protein Conformation, Ribosomal Proteins genetics, Ribosomal Proteins isolation & purification, Bacterial Proteins chemistry, Escherichia coli chemistry, Ribosomal Proteins chemistry
- Abstract
The dimer to monomer equilibrium and interdomain separations of cysteine variants of L7/L12 have been investigated using fluorescence spectroscopy. Steady-state polarization measurements on cysteine containing variants of L7/L12, labeled with 5-(iodoacetamido)fluorescein, demonstrated dimer to monomer dissociation constants near 30 nM for variants labeled at position 33, in the N-terminal domain, and positions 63 and 89, in the C-terminal domain. A dissociation constant near 300 nM was determined for a variant labeled at position 12, in the N-terminal domain. The polarization of a labeled C-terminal fragment did not change over the range of 200 microM to 1 nM, indicating that this construct remains monomeric at these concentrations, whereas a dimer to monomer dissociation constant near 300 nM was observed for an FITC labeled N-terminal fragment. Intersubunit fluorescence resonance energy self-transfer was observed when appropriate probes were attached to cysteines at residues 12 or 33, located in the N-terminal domain. Probes attached to cysteines at positions 63 or 89 in the C-terminal domain, however, did not exhibit intersubunit self-transfer. These results indicate that these residues in the C-terminal domains are, on average, separated by greater than 85 A. Intersubunit self-transfer does occur in a C-89 double mutation variant lacking 11 residues in the putative hinge region, indicating that the loss of the hinge region brings the two C-terminal domains closer together. Rapid subunit exchange between unlabeled wild-type L7/L12 and L7/L12 variants labeled in the N-terminal domain was also demonstrated by the loss of self-transfer upon mixing of the two proteins.
- Published
- 1996
- Full Text
- View/download PDF
22. Rotational and conformational dynamics of Escherichia coli ribosomal protein L7/L12.
- Author
-
Hamman BD, Oleinikov AV, Jokhadze GG, Traut RR, and Jameson DM
- Subjects
- Bacterial Proteins genetics, Dimerization, Escherichia coli chemistry, Escherichia coli genetics, Fluorescence Polarization, Fluorescent Dyes, Mutagenesis, Site-Directed, Protein Conformation, Recombinant Proteins chemistry, Recombinant Proteins genetics, Ribosomal Proteins genetics, Thermodynamics, Bacterial Proteins chemistry, Ribosomal Proteins chemistry
- Abstract
Fluorescence methods were utilized to study dynamic aspects of the 24 kDa dimeric Escherichia coli ribosomal protein L7/L12. Oligonucleotide site-directed mutagenesis was used to introduce cysteine residues at specific locations along the peptide chain, in both the C-terminal and N-terminal domains, and various sulfhydryl reactive fluorescence probes (iodoacetamido) fluorescein, IAEDANS, pyrenemethyl iodoacetate) were attached to these residues. In addition to the full-length proteins, a hinge-deleted variant and variants corresponding to the C-terminal fragment and the N-terminal fragment were also studied. Both steady-state and time-resolved fluorescence measurements were carried out, and the results demonstrated that L7/L12 is not a rigid molecule. Specifically, the two C-terminal domains move freely with respect to one another and with respect to the dimeric N-terminal domain. Removal of the hinge region, however, significantly reduces the mobility of the C-terminal domains. The data also show that the rotational relaxation time monitored by the fluorescent probe-depends upon the probe's excited state lifetime. This observation is interpreted to indicate that a hierarchy of motions exists in the L7/L12 molecule including facile motions of the C-terminal domains and dimeric N-terminal domain, in addition to the overall tumbling of the protein. Probes attached to the N-terminal domain exhibit global rotational relaxation times consistent with the molecular mass of the dimeric N-terminal fragment. Upon reconstitution of labeled L7/L12 with ribosomal cores, however, the motion associated with the dimeric N-terminal domain is greatly diminished while the facile motion of the C-terminal domains is almost unchanged.
- Published
- 1996
- Full Text
- View/download PDF
23. Tetramethylrhodamine dimer formation as a spectroscopic probe of the conformation of Escherichia coli ribosomal protein L7/L12 dimers.
- Author
-
Hamman BD, Oleinikov AV, Jokhadze GG, Bochkariov DE, Traut RR, and Jameson DM
- Subjects
- Binding Sites, Cysteine, Fluorescent Dyes, Genetic Variation, Macromolecular Substances, Models, Structural, Ribosomal Protein L10, Ribosomes metabolism, Sequence Deletion, Escherichia coli metabolism, Protein Conformation, Rhodamines, Ribosomal Proteins chemistry
- Abstract
The fluorescent probe tetramethylrhodamine iodoacetamide was attached to cysteine residues substituted at various specific locations in full-length and deletion variants of the homodimeric Escherichia coli ribosomal protein L7/L12. Ground-state tetramethylrhodamine dimers form between the two subunits of L7/L12 depending upon the location of the probe. The formation of tetramethylrhodamine dimers caused the appearance of a new absorption band at 518 nm that was used to estimate the extent of interaction of the probes in the different protein variants. Intersubunit tetramethylrhodamine dimers form when tetramethylrhodamine acetamide is attached to two different sites in the N-terminal domain of the L7/L12 dimer (residues 12 or 33), but not when attached to sites in the C-terminal domain (residues 63, 89, or 99). The tetramethylrhodamine dimers do form at sites in the C-terminal domain in L7/L12 variants that contain deletions of 11 or 18 residues within the putative flexible hinge that separates the N- and C-terminal domains. The tetramethylrhodamine dimers disappear rapidly (within 5 s) upon addition of excess unlabeled wild-type L7/L12. It appears that singly labeled L7/L12 dimers are formed by exchange with wild-type dimers. Binding of L7/L12:tetramethylrhodamine cysteine 33 or cysteine 12 dimers either to L7/L12-depleted ribosomal core particles, or to ribosomal protein L10 alone, results in disappearance of the 518-nm absorption band. This result implies a conformational change in the N-terminal domain of L7/L12 upon its binding to the ribosome, or to L10.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.