320 results on '"S. Willard"'
Search Results
2. A Longitudinal Evaluation of a Multimodal POCUS Curriculum in Pediatric Residents
- Author
-
Reshma Sabnani, Celia S. Willard, Carolina Vega, and Zachary W. Binder
- Subjects
Pediatrics ,POCUS ,ultrasound curriculum ,paediatric POCUS ,Internal medicine ,RC31-1245 ,Medical technology ,R855-855.5 - Abstract
Introduction: Pediatric residency programs often do not include a point of care ultrasound (POCUS) curriculum. We analyzed a novel POCUS curriculum for pediatric residents that incorporated an online question bank (QB), in addition to a traditional teaching model of didactic instruction and hands-on learning experience. Methods: Four high-yield POCUS topics were chosen: Focused Assessment by Sonography for Trauma (FAST), soft tissue, lung, and cardiac. Residents completed online multiple-choice quizzes before and after each of four in-person learning sessions, taught by ultrasound faculty and fellows. At the end of the academic year participants completed a knowledge retention quiz. Confidence surveys were administered to participants throughout the course of the study. Differences in means were compared by Student’s t-test. Results: Learners demonstrated post-intervention score improvement for each of the four modules. Retention testing demonstrated retained improvement for the soft tissue and cardiac modules, but not for the FAST module. Self-reported confidence increased across all four topics. Conclusion: A multimodal POCUS curriculum utilizing a combination of an online QB and in-person teaching demonstrated lasting knowledge for pediatric trainees.
- Published
- 2023
- Full Text
- View/download PDF
3. Coronal Heating as Determined by the Solar Flare Frequency Distribution Obtained by Aggregating Case Studies
- Author
-
James Paul Mason, Alexandra Werth, Colin G. West, Allison Youngblood, Donald L. Woodraska, Courtney L. Peck, Arvind J. Aradhya, Yijian Cai, David Chaparro, James W. Erikson, Koushik Ganesan, T. R. Geerdts, Thi D Hoang, Thomas M. Horning, Yan Jin, Haixin Liu, Noah Lordi, Zheng Luo, Thanmay S. Menon, Josephine C. Meyer, Emma E Nelson, Kristin A. Oliver, Jorge L Ramirez Ortiz, Andrew Osborne, Alyx Patterson, Nick Pellatz, John Pitten, Nanako Shitara, Daniel Steckhahn, Aseem Visal, Hongda Wang, Chaoran Wang, Evan Wickenden, John Wilson, Mengyu Wu, Nikolay Yegovtsev, Ingrid H Zimmermann, James Holland Aaron, Jumana T. Abdullah, Jonathan M. Abrams, Riley Abrashoff, Andres B. Acevedo, Iker Acha, Daniela M. Meza Acosta, Megan M. Adam, Dante Q. Adams, Kalvyn N Adams, Elena R Adams, Zainab A. Akbar, Ushmi H. Akruwala, Adel Al-Ghazwi, Batool H. Alabbas, Areej A. Alawadhi, Yazeed A. Alharbi, Mohammed S. Alahmed, Mohammed A. Albakr, Yusef J. Albalushi, Jonathan Albaum, Ahmed Aldhamen, Nolan Ales, Mohammad Alesmail, Abdulelah Alhabeeb, Dania Alhamli, Isehaq Alhuseini, Suhail Alkaabi, Tameem Alkhezzi, Mohamed Alkubaisi, Nasser Allanqawi, Martin Allsbrook, Yousef A. Almohsen, Justin Thomas Almquist, Teeb Alnaji, Yousef A Alnasrallah, Nicholas Alonzi, Meshal Alosaimi, Emeen Alqabani, Mohammad Alrubaie, Reema A. Alsinan, Ava L. Altenbern, Abdullah Altokhais, Saleh A. Alyami, Federico Ameijenda, Hamzi Amer, Meggan Amos, Hunter J. Anderson, Carter Andrew, Jesse C Andringa, Abigail Angwin, Gabreece Van Anne, Andrew Aramians, Camila Villamil Arango, Jack. W. Archibald, Brian A. Arias-Robles, Maryam Aryan, Kevin Ash, Justin Astalos, N. S. Atchley-Rivers, Dakota N. Augenstein, Bryce W. Austin, Abhinav Avula, Matthew C. Aycock, Abdulrahman A. Baflah, Sahana Balaji, Brian Balajonda, Leo M Balcer, James O. Baldwin, David J Banda, Titus Bard, Abby Barmore, Grant M. Barnes, Logan D. W. Barnhart, Kevin M. Barone, Jessica L. Bartman, Claire Bassel, Catalina S Bastias, Batchimeg Bat-Ulzii, Jasleen Batra, Lexi Battist, Joshua Bay, Simone Beach, Sara Beard, Quinn I Beato, Ryan Beattie, Thomas Beatty, Tristan De La Beaujardiere, Jacob N. Beauprez, M. G. Beck, Lily Beck, Simone E. Becker, Braden Behr, Timothy A. Behrer, Joshua Beijer, Brennan J. Belei, Annelene L. Belknap, Aislyn Bell, Caden Bence, Evan Benke, Naomi Berhanu, Zachary D. Berriman-Rozen, Chrisanna Bertuccio, Owen A. Berv, Blaine B. Biediger, Samuel J Biehle, Brennen Billig, Jacob Billingsley, Jayce A. Billman, Connor J. Biron, Gabrielle E. Bisacca, Cassidy A. Blake, Guillermo Blandon, Olivia Blevins, Ethan Blouin, Michal Bodzianowski, Taylor A. Boeyink, Matthew Bondar, Lauren Bone, Alberto Espinosa De Los Monteros Bonilla, William T Borelli, Luke R. Borgerding, Troy Bowen, Christine Boyer, Aidan Boyer, Aidan P. Boyle, Tom Boyne, Donovan Branch, Ariana E. Brecl, David J. Brennan, Alexander J Brimhall, Jennifer L. Brockman, Sarah Brookins, Gabriel T. Brown, Cameron L. Brown, Ryan Brown, Jordi Brownlow, Grant Brumage-Heller, Preston J. Brumley, Samuel Bryan, A. Brzostowicz, Maryam Buhamad, Gigi Bullard-Connor, J. R. Ramirez Bunsow, Annemarie C. Burns, John J. Burritt, Nicholas David Burton, Taylor Burton, Celeste Busch, Dylan R. Butler, B. W. Buxton, Malena C. Toups, Carter C. Cabbage, Breonna Cage, Jackson R. Cahn, Andrew J Campbell, Braden P. Canales, Alejandro R. Cancio, Luke Carey, Emma L. Carillion, Michael Andrew Carpender, Emily Carpenter, Shivank Chadda, Paige Chambers, Jasey Chanders, Olivia M. Chandler, Ethan C. Chang, Mitchell G. Chapman, Logan T. Chapman, S. Chavali, Luis Chavez, Kevin Chen, Lily Chen, Sam Chen, Judy Chen, Jenisha Chhetri, Bradyn Chiles, Kayla M. Chizmar, Katherine E Christiansen, Nicholas A. Cisne, Alexis Cisneros, David B. Clark, Evelyn Clarke, Peter C Clarkson, Alexis R. Clausi, Brooke Cochran, Ryan W. Coe, Aislinn Coleman-Plante, Jake R. Colleran, Zachary Colleran, Curran Collier, Nathaniel A. Collins, Sarah Collins, Jack C. Collins, Michael Colozzi, Aurora Colter, Rebecca A. Cone, Thomas C. Conroy, Reese Conti, Charles J. Contizano, Destiny J. Cool, Nicholas M. Cooper, Jessica S Corbitt, Jonas Courtney, Olivia Courtney, Corben L. Cox, Wilmsen B. Craig, Joshua B. Creany, Anastasia Crews, K. A. Crocker, A. J. Croteau, Christian J. Crow, Zoe Cruse, Avril Cruz, Tyler L. Curnow, Hayden Current, Riley T. Curry, Libby Cutler, Aidan St. Cyr, Frederick M. Dabberdt, Johnston Daboub, Olivia Damgaard, Swagatam Das, Emma A. B. Davis, Elyse Debarros, Sean Deel, Megan E. Delasantos, Tianyue Deng, Zachary Derwin, Om Desai, Kai Dewey, John S. Dias, Kenzie A. Dice, R. Dick, Cyrus A. Dicken, Henry Dietrick, Alexis M. Dinser, Alyssa M. Dixon, Thomas J. Dixon, Helen C. Do, Chris H Doan, Connor Doane, Joshua Dodrill, Timothy Doermer, Lizbeth Montoya Dominguez, J. Dominguez, Emerson N. Domke, Caroline R. Doran, Jackson A. Dorr, Philip Dorricott, Danielle C. Dresdner, Michael Driscoll, Kailer H. Driscoll, Sheridan J. Duncan, Christian Dunlap, Gabrielle M. Dunn, Tien Q. Duong, Tomi Oshima Dupeyron, Peter Dvorak, Andrew East, Andrew N. East, Bree Edwards, Lauren Ehrlich, Sara I. Elbashir, Rasce Engelhardt, Jacob Engelstad, Colin England, Andrew Enrich, Abbey Erickson, Benjamin Erickson, Nathan Evans, Calvin A Ewing, Elizabeth A. Eyeson, Ian Faber, Avery M. Fails, John T Fauntleroy, Kevin Fell, Zitian Feng, Logan D. Fenwick, Nikita Feoktistov, Ryann Fife, John Alfred D. Figueirinhas, Jean-Paul Fisch, Emmalee Fischer, Jules Fischer-White, Aidan F. Fitton, Alexander Fix, Lydia Flackett, Fernando Flores, Aidan Floyd, Leonardo Del Foco, Adeduni Folarin, Aidan E. Forbes, Elise Fortino, Benjamin L. Fougere, Alexandra A. Fowler, Margaret Fox, James M. French, Katherine V. French, Florian G. Frick, Calvin R. Fuchs, Bethany E. S. Fuhrman, Sebastian Furney, Moutamen Gabir, Gabriela Galarraga, Skylar Gale, Keala C. Gapin, A. J. Garscadden, Rachel Gasser, Lily Gayou, Emily E. Gearhart, Jane Geisman, Julianne R. Geneser, Sl Genne, Julia G Gentile, Eleanor Gentry, Jacob D. George, Nathaniel James Georgiades, Phillip Gerhardstein, Clint Gersabeck, Bandar Abu Ghaith, Dorsa Ghiassi, B. C. Giebner, Dalton Gilmartin, Connor B. Gilpatrick, Michael Gjini, Olivia Golden, Nathan T. Golding, C. A. Goldsberry, Angel R. Gomez, Angel A. Gomez, Sean Gopalakrishnan, Mariam Gopalani, Nicholas Gotlib, Alaina S. Graham, Michael J Gray, Alannah H. Gregory, Joshua A. Gregory, Kristyn Grell, Justus Griego, Nicholas F. Griffin, Kyle J. Griffin, Matt Guerrero, Nicole Gunderson, Mutian Guo, E. R. Gustavsson, Grace K. Hach, L. N. Haile, Jessica Haines, Jack J. Mc Hale, Ryder Buchanan Hales, Mark S. Haley, Jacqueline L. Hall, Sean R. Hamilton, Soonhee Han, Tyler Hand, Luke C. Hanley, Connor M Hansen, Joshua A. Hansen, Jonathan Hansson, Tony Yunfei Hao, Nicholas Haratsaris, Isabelle Hardie, Dillon F. Hardwick, Cameron T. Hares, Logan Swous Harris, Coyle M. Harris, Omer Hart, Kyle Hashiro, Elsie Hattendorf, Calder Haubrich, Elijah Hawat, Griffin A. Hayrynen, Danielle A. Heintz, Tim Hellweg, Angel Hernandez, Emanuel Herrera, Robert N. Herrington, Tim Herwig, Troy M. Hesse, Quinn Hiatt, Lea Pearl Hibbard, Imari R. Hicks, Andrew J. Hicks, Nigel Highhouse, Annalise K. Hildebrand, Paula Hill, Hallie Hill, Evan Hintsa, Anna E. Hirschmann, Travis Hitt, Ella Ho, Isabelle J. Hoff, Alex Hoffman, Blake A. Hogen, Linda Horne, Timothy J Houck, Noah H. Howell, E. M. Hrudka, J. Hu, Jianyang Huang, Chenqi Huang, Shancheng Huang, Zachary A. Hudson, Nathan C. Hudson, Tyler J. Huebsch, Owen Hull, Samuel C Hunter, Troy Husted, Abigail P. Hutabarat, Leslie Huynh, Antonio E. Samour Ii, Yolande Idoine, Julia A. Ingram, Taro Iovan, Samuel A. Isert, Antonio Salcido-Alcontar Jr, Thomas Jacobsen, Alan A Jaimes, Connor Jameson, J. R. Jarriel, Sam Jarvis, Josh Jenkins, Alexander V. Jensen, Jacob Jeong, Luke A. Jeseritz, Trevor Jesse, Soo Yeun Ji, Yufan Jiang, Owen Johnson, Matthew Johnson, Sawyer Johnson, Julia Johnston, Braedon Y. Johnston, Olivia M. Jones, M. R. Jones, Tara Jourabchi, Tony A. House Jr., Parker Juels, Sabrina J. H. T. Kainz, Emily Kaiser, Nicolas Ian Kallemeyn, Madison H. Kalmus, Etash Kalra, Margaret Kamenetskiy, Jeerakit Kanokthippayakun, Shaun D. Kapla, Brennan J. Karsh, Caden J. Keating, Morgan A. Kelley, Michael P. Kelley, Nicholas Kelly, James Kelly, Teagan Kelly, Christopher M Kelly, Kellen Kennedy, Cayla J. Kennedy, Forrest Kennedy, Abigail Kennedy, Liana Kerr-Layton, Marilyn Ketterer, Ibraheem A. Khan, Usman Khan, Sapriya Khanal, Jack L. Kiechlin, Dominic Killian, Kevin Kim, Brian T. Kim, Matthew M. Kim, Jake Kim, Aspen Kimlicko, Isabel M Kipp, Hunter B. Kirkpatrick, Natalie Kissner, Emily R. Kite, Olivia R. Kleinhaus, Philip Whiting Knott, Will Koch, Greta Koenig, Emily Koke, Thomas Kokes, Yash S. Kothamdi, Zack Krajnak, Zoe M. Kresek, Dylan Kriegman, Jake E. Kritzberg, Davis J. Krueger, Bartlomiej Kubiak, Kirsten Kuehl, Chrisanne Kuester, Nicolas A. Kuiper, Aman Priyadarshi Kumar, Connor Kuybus, Daniel Kwiatkowski, Quintin Y. Lafemina, Kevin Lacjak, Kyle Lahmers, Antonia Lam, Kalin Landrey, Maxwell B. Lantz, Zachary Larter, Benjamin P. Lau, Megan Lauzon, Rian Lawlor, Tyler Learned, E. C. Lee, Junwon Lee, Adrianna J. Lee, Justin Lee, Alexis Ying-Shan Lee, Christian J Lee, Nathaniel F. Lee, Linzhi Leiker, Dylan Lengerich, Cecilia Leoni, Adrienne R. Lezak, David Y. Li, Isaac Li, Ryan Z. Liao, Bridget Linders, Morgan I Linger, Katherine B. Linnane, Sam Lippincott, Barrett Lister, Shelby D Litton, Nianzi Liu, Steven Y. Liu, Timothy W. Logan, Nathan Londres, Mia C. Lonergan, Emily Lookhoff, N. E. Loomis, Christian Lopez, Justin Loring, Jeffrey Lucca, Dax Lukianow, Nathan M. Cheang, William Macdonald, Claire A. Madonna, Kasey O. Madsen, Tiffany E. Maksimuk, Macguire Mallory, Ryan A. Malone, Blake Maly, Xander R. Manzanares, Aimee S. Maravi, Serafima M. Marcus, Nasreen Marikar, Josie A. Marquez, Mathew J. Marquez, Lauren Marsh, Toni Marsh, Logan S. Martin, Alexa M. Martinez, Jose R. Martinez, Hazelia K. Martinez, Cara Martyr, Mirna Masri, Giorgio Matessi, Adam Izz Khan Mohd Reduan Mathavan, Randi M. Mathieson, Kabir P. Mathur, Graham Mauer, Victoria A. Mayer, Liam Mazzotta, Glen S. Mccammon, Rowan Mcconvey, Tyler Mccormick, Andrew Mccoy, Kelleen Mcentee, Meaghan V. Mcgarvey, Riley M. Mcgill, James K. Mcintyre, Finbar K. Mckemey, Zane Mcmorris, Jesse J. Mcmullan, Ella Mcquaid, Caden Mcvey, Kyle Mccurry, Mateo M. Medellin, Melissa Medialdea, Amar Mehidic, Stella Meillon, Jonah B. Meiselman-Ashen, Sarah Mellett, Dominic Menassa, Citlali Mendez, Patricia Mendoza-Anselmi, Riley Menke, Sarah Mesgina, William J. Mewhirter, Ethan Meyer, Aya M. Miften, Ethan J. Miles, Andrew Miller, Joshua B. Miller, Emily B. Millican, Sarah J. Millican, Dylan P. Mills, Josh Minimo, Jay H. Misener, Alexander J. Mitchell, Alexander Z. Mizzi, Luis Molina-Saenz, Tyler S Moll, Hayden Moll, Maximus Montano, Michael Montoya, Eli Monyek, Jacqueline Rodriguez Mora, Gavin Morales, Genaro Morales, Annalise M. Morelock, Cora Morency, Angel J. Moreno, Remy Morgan, Alexander P. Moss, Brandon A. Muckenthaler, Alexander Mueller, Owen T. Mulcahy, Aria T. Mundy, Alexis A. Muniz, Maxwell J. Murphy, Madalyn C. Murphy, Ryan C. Murphy, Tyler Murrel, Andrew J. Musgrave, Michael S. Myer, Kshmya Nandu, Elena R. Napoletano, Abdulaziz Naqi, Anoothi Narayan, Liebe Nasser, Brenna K Neeland, Molly Nehring, Maya Li Nelson, Lena P. Nguyen, Lena Nguyen, Leonardo Nguyen, Valerie A. Nguyen, Khoa D Nguyen, Kelso Norden, Cooper Norris, Dario Nuñez, Rosemary O. Nussbaum, Cian O’Sullivan, Ian O’Neill, S. H. Oakes, Anand Odbayar, Caleb Ogle, Sean Oishi-Holder, Nicholas Olguin, Nathaniel P. Olson, Jason Ong, Elena N. Opp, Dan Orbidan, Ryan Oros, Althea E. Ort, Matthew Osborn, Austin Osogwin, Grant Otto, Jessica Oudakker, Igor Overchuk, Hannah M. Padgette, Jacqueline Padilla, Mallory Palizzi, Madeleine L. Palmgren, Adler Palos, Luke J. Pan, Nathan L. Parker, Sasha R. Parker, Evan J. Parkinson, Anish Parulekar, Paige J. Pastor, Kajal Patel, Akhil Patel, Neil S. Patel, Samuel Patti, Catherine Patton, Genevieve K. Payne, Matthew P. Payne, Harrison M. Pearl, Charles B. Beck Von Peccoz, Alexander J. Pedersen, Lily M. Pelster, Munisettha E. Peou, J. S. Perez, Freddy Perez, Anneliese Pesce, Audrey J. Petersen, B. Peterson, Romeo S. L. Petric, Joshua Pettine, Ethan J. Phalen, Alexander V. Pham, Denise M. Phan, Callie C Pherigo, Lance Phillips, Justin Phillips, Krista Phommatha, Alex Pietras, Tawanchai P. Pine, Sedique Pitsuean-Meier, Andrew M. Pixley, Will Plantz, William C. Plummer, Kaitlyn E. Plutt, Audrey E. Plzak, Kyle Pohle, Hyden Polikoff, Matthew Pollard, Madelyn Polly, Trevor J. Porter, David Price, Nicholas K. Price, Gale H. Prinster, Henry Austin Propper, Josh Quarderer, Megan S. Quinn, Oliver Quinonez, Devon Quispe, Cameron Ragsdale, Anna L. Rahn, M. Rakhmonova, Anoush K Ralapanawe, Nidhi Ramachandra, Nathaniel Ramirez, Ariana C. Ramirez, Sacha Ramirez, Parker Randolph, Anurag Ranjan, Frederick C Rankin, Sarah Grace Rapaport, Nicholas O Ratajczyk, Mia G. V. Ray, Brian D. Reagan, John C. Recchia, Brooklyn J. Reddy, Joseph Reed, Charlie Reed, Justin Reeves, Eileen N. Reh, Ferin J. Von Reich, Andrea B. Reyna, Alexander Reynolds, Hope Reynolds, Matthew Rippel, Guillermo A. Rivas, Anna Linnea Rives, Amanda M. Robert, Samuel M. Robertson, Maeve Rodgers, Stewart Rojec, Andres C. Romero, Ryan Rosasco, Beth Rossman, Michael Rotter, Tyndall Rounsefell, Charlotte Rouse, Allie C. Routledge, Marc G. Roy, Zoe A. Roy, Ryan Ruger, Kendall Ruggles-Delgado, Ian C. Rule, Madigan Rumley, Brenton M. Runyon, Collin Ruprecht, Bowman Russell, Sloan Russell, Diana Ryder, David Saeb, J. Salazar, Violeta Salazar, Maxwell Saldi, Jose A. Salgado, Adam D. Salindeho, Ethan S. Sanchez, Gustavo Sanchez-Sanchez, Darian Sarfaraz, Sucheta Sarkar, Ginn A. Sato, Carl Savage, Marcus T. Schaller, Benjamin T. Scheck, Jared A. W. Schlenker, Matthew J Schofer, Stephanie H. Schubert, Courtney Schultze, Grace K Schumacher, Kasper Seglem, Lauren Serio, Octave Seux, Hannan Shahba, Callie D. Shannahan, Shajesh Sharma, Nathan Shaver, Timothy Shaw, Arlee K. Shelby, Emma Shelby, Grace Shelchuk, Tucker Sheldrake, Daniel P. Sherry, Kyle Z. Shi, Amanda M. Shields, Kyungeun Shin, Michael C. Shockley, Dominick Shoha, Jadon Shortman, Mitchell Shuttleworth, Lisa Sibrell, Molly G. Sickler, Nathan Siles, H. K. Silvester, Conor Simmons, Dylan M. Simone, Anna Simone, Savi Singh, Maya A. Singh, Madeline Sinkovic, Leo Sipowicz, Chris Sjoroos, Ryan Slocum, Colin Slyne, Korben Smart, Alexandra N. Smith, Kelly Smith, Corey Smith, Elena K. Smith, Samantha M. Smith, Percy Smith, Trevor J Smith, G. L. Snyder, Daniel A. Soby, Arman S. Sohail, William J. Solorio, Lincoln Solt, Caitlin Soon, Ava A Spangler, Benjamin C. Spicer, Ashish Srivastava, Emily Stamos, Peter Starbuck, Ethan K. Stark, Travis Starling, Caitlyn Staudenmier, Sheen L. Steinbarth, Christopher H. Steinsberger, Tyler Stepaniak, Ellie N. Steward, Trey Stewart, T. C. Stewart, Cooper N. Stratmeyer, Grant L. Stratton, Jordin L. Stribling, S. A Sulaiman, Brandon J Sullivan, M. E. Sundell, Sohan N. Sur, Rohan Suri, Jason R. Swartz, Joshua D. Sweeney, Konner Syed, Emi Szabo, Philip Szeremeta, Michael-Tan D. Ta, Nolan C. Tanguma, Kyle Taulman, Nicole Taylor, Eleanor Taylor, Liam C. Taylor, K. E. Tayman, Yesica Tellez, Richard Terrile, Corey D Tesdahl, Quinn N. Thielmann, Gerig Thoman, Daniel Thomas, Jeffrey J. Thomas, William N. Thompson, Noah R. Thornally, Darien P. Tobin, Kelly Ton, Nathaniel J. Toon, Kevin Tran, Bryn Tran, Maedee Trank-Greene, Emily D. Trautwein, Robert B. Traxler, Judah Tressler, Tyson R. Trofino, Thomas Troisi, Benjamin L. Trunko, Joshua K. Truong, Julia Tucker, Thomas D Umbricht, C. H. Uphoff, Zachary T. Upthegrove, Shreenija Vadayar, Whitney Valencia, Mia M. Vallery, Eleanor Vanetten, John D. Vann, Ilian Varela, Alexandr Vassilyev, Nicholas J. Vaver, Anjali A. Velamala, Evan Vendetti, Nancy Ortiz Venegas, Aditya V. Vepa, Marcus T. Vess, Jenna S. Veta, Andrew Victory, Jessica Vinson, Connor Maklain Vogel, Michaela Wagoner, Steven P. Wallace, Logan Wallace, Caroline Waller, Jiawei Wang, Keenan Warble, N. R. D. Ward-Chene, James Adam Watson, Robert J. Weber, Aidan B. Wegner, Anthony A Weigand, Amanda M. Weiner, Ayana West, Ethan Benjamin Wexler, Nicola H. Wheeler, Jamison R. White, Zachary White, Oliver S. White, Lloyd C. Whittall, Isaac Wilcove, Blake C. Wilkinson, John S. Willard, Abigail K. Williams, Sajan Williams, Orion K. Wilson, Evan M. Wilson, Timothy R. Wilson, Connor B. Wilson, Briahn Witkoff, Aubrey M. Wolfe, Jackson R. Wolle, Travis M. Wood, Aiden L. Woodard, Katelynn Wootten, Catherine Xiao, Jianing Yang, Zhanchao Yang, Trenton J. Young, Isabel Young, Thomas Zenner, Jiaqi Zhang, Tianwei Zhao, Tiannie Zhao, Noah Y. Zhao, Chongrui Zhou, Josh J Ziebold, Lucas J. Ziegler, James C. Zygmunt, Jinhua Zhang, and H. J. Lewandowski
- Subjects
Solar physics ,Solar flares ,Astrostatistics distributions ,Astrophysics ,QB460-466 - Abstract
Flare frequency distributions represent a key approach to addressing one of the largest problems in solar and stellar physics: determining the mechanism that counterintuitively heats coronae to temperatures that are orders of magnitude hotter than the corresponding photospheres. It is widely accepted that the magnetic field is responsible for the heating, but there are two competing mechanisms that could explain it: nanoflares or Alfvén waves. To date, neither can be directly observed. Nanoflares are, by definition, extremely small, but their aggregate energy release could represent a substantial heating mechanism, presuming they are sufficiently abundant. One way to test this presumption is via the flare frequency distribution, which describes how often flares of various energies occur. If the slope of the power law fitting the flare frequency distribution is above a critical threshold, α = 2 as established in prior literature, then there should be a sufficient abundance of nanoflares to explain coronal heating. We performed >600 case studies of solar flares, made possible by an unprecedented number of data analysts via three semesters of an undergraduate physics laboratory course. This allowed us to include two crucial, but nontrivial, analysis methods: preflare baseline subtraction and computation of the flare energy, which requires determining flare start and stop times. We aggregated the results of these analyses into a statistical study to determine that α = 1.63 ± 0.03. This is below the critical threshold, suggesting that Alfvén waves are an important driver of coronal heating.
- Published
- 2023
- Full Text
- View/download PDF
4. Tirzepatide is an imbalanced and biased dual GIP and GLP-1 receptor agonist
- Author
-
Francis S. Willard, Jonathan D. Douros, Maria B.N. Gabe, Aaron D. Showalter, David B. Wainscott, Todd M. Suter, Megan E. Capozzi, Wijnand J.C. van der Velden, Cynthia Stutsman, Guemalli R. Cardona, Shweta Urva, Paul J. Emmerson, Jens J. Holst, David A. D’Alessio, Matthew P. Coghlan, Mette M. Rosenkilde, Jonathan E. Campbell, and Kyle W. Sloop
- Subjects
Therapeutics ,Medicine - Abstract
Tirzepatide (LY3298176) is a dual GIP and GLP-1 receptor agonist under development for the treatment of type 2 diabetes mellitus (T2DM), obesity, and nonalcoholic steatohepatitis. Early phase trials in T2DM indicate that tirzepatide improves clinical outcomes beyond those achieved by a selective GLP-1 receptor agonist. Therefore, we hypothesized that the integrated potency and signaling properties of tirzepatide provide a unique pharmacological profile tailored for improving broad metabolic control. Here, we establish methodology for calculating occupancy of each receptor for clinically efficacious doses of the drug. This analysis reveals a greater degree of engagement of tirzepatide for the GIP receptor than the GLP-1 receptor, corroborating an imbalanced mechanism of action. Pharmacologically, signaling studies demonstrate that tirzepatide mimics the actions of native GIP at the GIP receptor but shows bias at the GLP-1 receptor to favor cAMP generation over β-arrestin recruitment, coincident with a weaker ability to drive GLP-1 receptor internalization compared with GLP-1. Experiments in primary islets reveal β-arrestin1 limits the insulin response to GLP-1, but not GIP or tirzepatide, suggesting that the biased agonism of tirzepatide enhances insulin secretion. Imbalance toward GIP receptor, combined with distinct signaling properties at the GLP-1 receptor, together may account for the promising efficacy of this investigational agent.
- Published
- 2020
- Full Text
- View/download PDF
5. Mutual Cooperativity of Three Allosteric Sites on the Dopamine D1 Receptor
- Author
-
Xushan Wang, Erik J. Hembre, Paul J. Goldsmith, James P. Beck, Kjell A. Svensson, Francis S. Willard, and Robert F. Bruns
- Subjects
Pharmacology ,Molecular Medicine - Published
- 2022
- Full Text
- View/download PDF
6. Supplementary Figure 4 from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
PDF file - 34K, Effect of BAY36-7620 on prostate cancer cells proliferation
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 2 from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
PDF file - 61K, Immunohistochemical staining of metabotropic glutamate receptor 1 (GRM1) in selected non-malignant prostatic tissues
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure Legend from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
PDF file - 83K
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
Purpose: During glutaminolysis, glutamine is catabolized to glutamate and incorporated into citric acid cycle and lipogenesis. Serum glutamate levels were measured in patients with primary prostate cancer or metastatic castrate-resistant prostate cancer (mCRPCa) to establish clinical relevance. The effect of glutamate deprivation or blockade by metabotropic glutamate receptor 1 (GRM1) antagonists was investigated on prostate cancer cells' growth, migration, and invasion to establish biologic relevance.Experimental Design: Serum glutamate levels were measured in normal men (n = 60) and patients with primary prostate cancer (n = 197) or mCRPCa (n = 109). GRM1 expression in prostatic tissues was examined using immunohistochemistry (IHC). Cell growth, migration, and invasion were determined using cell cytotoxicity and modified Boyden chamber assays, respectively. Apoptosis was detected using immunoblotting against cleaved caspases, PARP, and γ-H2AX.Results: Univariate and multivariate analyses showed significantly higher serum glutamate levels in Gleason score ≥ 8 than in the Gleason score ≤ 7 and in African Americans than in the Caucasian Americans. African Americans with mCRPCa had significantly higher serum glutamate levels than those with primary prostate cancer or benign prostate. However, in Caucasian Americans, serum glutamate levels were similar in normal research subjects and patients with mCRPC. IHC showed weak or no expression of GRM1 in luminal acinar epithelial cells of normal or hyperplastic glands but high expression in primary or metastatic prostate cancer tissues. Glutamate deprivation or blockade decreased prostate cancer cells' proliferation, migration, and invasion and led to apoptotic cell death.Conclusions: Glutamate expression is mechanistically associated with and may provide a biomarker of prostate cancer aggressiveness. Clin Cancer Res; 18(21); 5888–901. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table 1 from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
PDF file - 66K, Descriptives of the study cohort used for serum glutamate quantification
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 3 from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
PDF file - 80K, Immunohistochemical staining of metabotropic glutamate receptor 1 (GRM1) in selected prostate cancers
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 1 from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
PDF file - 156K, Comparison of serum glutamate levels in African Americans and Caucasian Americans with metastatic-castrate resistant prostate cancer
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table 2 from Serum Glutamate Levels Correlate with Gleason Score and Glutamate Blockade Decreases Proliferation, Migration, and Invasion and Induces Apoptosis in Prostate Cancer Cells
- Author
-
Robert L. Vessella, James L. Mohler, Oliver Sartor, Katayoon Rezaei, Mojgan Shourideh, Hillary Thompson, Stacey S. Willard, Giovanni D. Lorusso, Charles Manhardt, Dhatchayini Subramani, Ray Scioneaux, Kristopher Attwood, Gissou Azabdaftari, Sunipa Majumdar, and Shahriar Koochekpour
- Abstract
PDF file - 75K, Multiple covariate adjusted analysis of serum glutamate levels in patients with primary and metastatic castrate-resistant prostate cancer
- Published
- 2023
- Full Text
- View/download PDF
14. Discovery of an Orally Efficacious Positive Allosteric Modulator of the Glucagon-like Peptide-1 Receptor
- Author
-
Daniel A. Briere, David B. Wainscott, José Miguel Minguez, Cynthia Stutsman, Qi Chen, Ana B. Bueno, Alma Jiménez, Kyle W. Sloop, Ana I. Mateo, Guemalli R. Cardona, Wenzhen Ma, Aaron D. Showalter, Graham R Cumming, Ana M. Castaño, Francis S. Willard, Richard W. Zink, Javier Agejas, Nathan Yumibe, and Christopher M Corkins
- Subjects
Blood Glucose ,Models, Molecular ,Agonist ,endocrine system ,Allosteric modulator ,medicine.drug_class ,Metabolite ,Administration, Oral ,Pharmacology ,01 natural sciences ,Glucagon-Like Peptide-1 Receptor ,Rats, Sprague-Dawley ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Allosteric Regulation ,Drug Discovery ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Receptor ,030304 developmental biology ,0303 health sciences ,Chemistry ,digestive, oral, and skin physiology ,Long-term potentiation ,Ligand (biochemistry) ,Glucagon-like peptide-1 ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Molecular Medicine ,hormones, hormone substitutes, and hormone antagonists ,Ex vivo - Abstract
The identification of LSN3318839, a positive allosteric modulator of the glucagon-like peptide-1 receptor (GLP-1R), is described. LSN3318839 increases the potency and efficacy of the weak metabolite GLP-1(9-36)NH2 to become a full agonist at the GLP-1R and modestly potentiates the activity of the highly potent full-length ligand, GLP-1(7-36)NH2. LSN3318839 preferentially enhances G protein-coupled signaling by the GLP-1R over β-arrestin recruitment. Ex vivo experiments show that the combination of GLP-1(9-36)NH2 and LSN3318839 produces glucose-dependent insulin secretion similar to that of GLP-1(7-36)NH2. Under nutrient-stimulated conditions that release GLP-1, LSN3318839 demonstrates robust glucose lowering in animal models alone or in treatment combination with sitagliptin. From a therapeutic perspective, the biological properties of LSN3318839 support the concept that GLP-1R potentiation is sufficient for reducing hyperglycemia.
- Published
- 2021
- Full Text
- View/download PDF
15. Nuisance compounds in cellular assays
- Author
-
J. Willem M. Nissink, Jonathan B. Baell, Francis S. Willard, Ina Rothenaigner, Jonathan Z. Sexton, Douglas S. Auld, Jayme L. Dahlin, John Strelow, Michael A. Walters, Bruce K. Hua, Steve Haney, Lori Ferrins, Jarrod Walsh, Bridget K. Wagner, Jonathan A. Lee, and Kamyar Hadian
- Subjects
High-throughput screening ,Phenotypic screening ,Clinical Biochemistry ,Chemical biology ,Computational biology ,Biology ,01 natural sciences ,Biochemistry ,Article ,Assay interference ,Artifacts ,Chemical Biology ,Drug Discovery ,High-content Screening ,High-throughput Screening ,Interference ,Nuisance Compounds ,Phenotypic Drug Discovery ,Phenotypic Screening ,Artificial Intelligence ,Humans ,Molecular Biology ,Pharmacology ,Biological Products ,010405 organic chemistry ,Drug discovery ,Cheminformatics ,0104 chemical sciences ,Pharmaceutical Preparations ,High-content screening ,Molecular Medicine ,Classical pharmacology ,Nuisance - Abstract
Compounds that exhibit assay interference or undesirable mechanisms of bioactivity (“nuisance compounds”) are routinely encountered in cellular assays, including phenotypic and high-content screening assays. Much is known regarding compound-dependent assay interferences in cell-free assays. However, despite the essential role of cellular assays in chemical biology and drug discovery, there is considerably less known about nuisance compounds in more complex cell-based assays. In our view, a major obstacle to realizing the full potential of chemical biology will not just be difficult-to-drug targets or even the sheer number of targets, but rather nuisance compounds, due to their ability to waste significant resources and erode scientific trust. In this review, we summarize our collective academic, government, and industry experiences regarding cellular nuisance compounds. We describe assay design strategies to mitigate the impact of nuisance compounds and suggest best practices to efficiently address these compounds in complex biological settings. Nuisance compounds can waste significant resources by producing promising bioactivities that are attributable to undesirable mechanisms of action. Addressing nuisance compounds is particularly challenging in cellular assays. Dahlin et al. summarize academic, government, and industry experiences with assay design and hit triage to specifically address cellular nuisance compounds.
- Published
- 2021
- Full Text
- View/download PDF
16. A synthetic method to assay adhesion-family G-protein coupled receptors. Determination of the G-protein coupling profile of ADGRG6(GPR126)
- Author
-
Francis S. Willard, Jozie L. Hayes, and Enric Lizano
- Subjects
Models, Molecular ,0301 basic medicine ,Gs alpha subunit ,G protein ,Recombinant Fusion Proteins ,Biophysics ,Ligands ,Models, Biological ,Second Messenger Systems ,Biochemistry ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,0302 clinical medicine ,GTP-Binding Proteins ,Heterotrimeric G protein ,Cyclic AMP ,Humans ,Amino Acid Sequence ,Receptor ,Molecular Biology ,G protein-coupled receptor ,biology ,Chemistry ,Cell Biology ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Gq alpha subunit ,030220 oncology & carcinogenesis ,Second messenger system ,biology.protein ,Synthetic Biology ,Signal transduction ,Cell Adhesion Molecules ,Protein Binding ,Signal Transduction - Abstract
G-protein coupled receptors (GPCRs) are the largest family of membrane-spanning receptors in metazoans and mediate diverse biological processes such as chemotaxis, vision, and neurotransmission. Adhesion GPCRs represent an understudied class of GPCRs. Adhesion GPCRs (ADGRs) are activated by an intrinsic proteolytic mechanism executed by the G-protein autoproteolysis inducing domain that defines this class of GPCRs. It is hypothesized that agonist ligands modulate the proteolyzed receptor to regulate the activity of a tethered agonist peptide that is an intramolecular activator of ADGRs. The mechanism of activation of ADGRs in physiological settings is unclear and the toolbox for interrogating ADGR physiology in cellular models is limited. Therefore, we generated a novel enterokinase-activated tethered ligand system for ADGRG6(GPR126). Enterokinase addition to cells expressing a synthetic ADGRG6 protein induced potent and efficacious signal transduction through heterotrimeric G-protein coupled second messenger pathways including cyclic nucleotide production, intracellular calcium mobilization, and GPCR-pathway linked reporter gene induction. These studies support the hypothesis that ADGRG6(GPR126) is coupled to multiple heterotrimeric G-proteins: including Gαs, Gαq, and Gα12. This novel assay method is robust, specific, and compatible with numerous cell pharmacology approaches. We present a new tool for determination of the biological function of ADGRs and the identification of ligands that engage these receptors.
- Published
- 2021
- Full Text
- View/download PDF
17. A High-Throughput Assay for the Pancreatic Islet Beta-Cell Potassium Channel: Use in the Pharmacological Characterization of Insulin Secretagogues Identified from Phenotypic Screening
- Author
-
Samreen K. Syed, David Gene Barrett, Francis S. Willard, Karen Leigh Cox, Dirk Tomandl, Alexander M. Efanov, and Luyan Song
- Subjects
medicine.medical_treatment ,Phenotypic screening ,Drug Evaluation, Preclinical ,Sulfonylurea Receptors ,Insulin-Secreting Cells ,Insulin Secretion ,Drug Discovery ,medicine ,Diazoxide ,Humans ,Potassium Channels, Inwardly Rectifying ,Cells, Cultured ,Drug discovery ,Chemistry ,Secretagogues ,Insulin ,Optical Imaging ,Potassium channel ,High-Throughput Screening Assays ,Potassium channel activity ,Phenotype ,Sulfonylurea Compounds ,Biochemistry ,Molecular Medicine ,Classical pharmacology ,Beta cell ,medicine.drug - Abstract
Phenotypic screening is a neoclassical approach for drug discovery. We conducted phenotypic screening for insulin secretion enhancing agents using INS-1E insulinoma cells as a model system for pancreatic beta-cells. A principal regulator of insulin secretion in beta-cells is the metabolically regulated potassium channel Kir6.2/SUR1 complex. To characterize hit compounds, we developed an assay to quantify endogenous potassium channel activity in INS-1E cells. We quantified ligand-regulated potassium channel activity in INS-1E cells using fluorescence imaging and thallium flux. Potassium channel activity was metabolically regulated and coupled to insulin secretion. The pharmacology of channel opening agents (diazoxide) and closing agents (sulfonylureas) was used to validate the applicability of the assay. A precise high-throughput assay was enabled, and phenotypic screening hits were triaged to enable a higher likelihood of discovering chemical matter with novel and useful mechanisms of action.
- Published
- 2021
- Full Text
- View/download PDF
18. Structural basis for GLP-1 receptor activation by LY3502970, an orally active nonpeptide agonist
- Author
-
Hitoshi Yoshino, Takahiro Kawai, Dan Feng, Shunsuke Nagao, Francis S. Willard, Kyle W. Sloop, Tong Sun Kobilka, Aaron D. Showalter, Yoshiki Kawabe, David B. Wainscott, Brian A. Droz, Matthew P. Coghlan, Brian K. Kobilka, Masanori Fukazawa, Yoshiyuki Suzuki, and Bingfa Sun
- Subjects
Male ,Models, Molecular ,0301 basic medicine ,Agonist ,Swine ,type 2 diabetes mellitus ,G protein ,medicine.drug_class ,cryoelectron microscopy ,Administration, Oral ,Aminopyridines ,Mice, Transgenic ,030209 endocrinology & metabolism ,Pharmacology ,Incretins ,Partial agonist ,Glucagon-Like Peptide-1 Receptor ,LY3502970 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Species Specificity ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Receptor ,Glucagon-like peptide 1 receptor ,G protein-coupled receptor ,Multidisciplinary ,Chemistry ,Tryptophan ,Biological Sciences ,OWL833 ,Rats ,Macaca fascicularis ,HEK293 Cells ,030104 developmental biology ,Mechanism of action ,Benzamides ,Mutagenesis, Site-Directed ,Anti-Obesity Agents ,medicine.symptom ,Exenatide ,medicine.drug - Abstract
Significance Glucagon-like peptide-1 receptor agonists have become established as a leading class of diabetes medications. However, these peptide-based drugs are administered by subcutaneous injection or, in one case, by a complex oral dosing regimen. We now report the discovery of LY3502970, a potent and selective small-molecule GLP-1R agonist. LY3502970 exhibits preclinical pharmacology equivalent to a marketed injectable GLP-1R agonist and possesses pharmacokinetic properties compatible with oral dosing in humans. Cryoelectron microscopy (cryo-EM) studies reveal an ECD-driven receptor binding mode for LY3502970 that provides a favorable pharmacological profile., Glucagon-like peptide-1 receptor (GLP-1R) agonists are efficacious antidiabetic medications that work by enhancing glucose-dependent insulin secretion and improving energy balance. Currently approved GLP-1R agonists are peptide based, and it has proven difficult to obtain small-molecule activators possessing optimal pharmaceutical properties. We report the discovery and mechanism of action of LY3502970 (OWL833), a nonpeptide GLP-1R agonist. LY3502970 is a partial agonist, biased toward G protein activation over β-arrestin recruitment at the GLP-1R. The molecule is highly potent and selective against other class B G protein–coupled receptors (GPCRs) with a pharmacokinetic profile favorable for oral administration. A high-resolution structure of LY3502970 in complex with active-state GLP-1R revealed a unique binding pocket in the upper helical bundle where the compound is bound by the extracellular domain (ECD), extracellular loop 2, and transmembrane helices 1, 2, 3, and 7. This mechanism creates a distinct receptor conformation that may explain the partial agonism and biased signaling of the compound. Further, interaction between LY3502970 and the primate-specific Trp33 of the ECD informs species selective activity for the molecule. In efficacy studies, oral administration of LY3502970 resulted in glucose lowering in humanized GLP-1R transgenic mice and insulinotropic and hypophagic effects in nonhuman primates, demonstrating an effect size in both models comparable to injectable exenatide. Together, this work determined the molecular basis for the activity of an oral agent being developed for the treatment of type 2 diabetes mellitus, offering insights into the activation of class B GPCRs by nonpeptide ligands.
- Published
- 2020
- Full Text
- View/download PDF
19. Structural insights into probe-dependent positive allosterism of the GLP-1 receptor
- Author
-
Betty Chau, Joseph D. Ho, Francisco J Agejas, Francis S. Willard, Ana B. Bueno, Tong Sun Kobilka, Brian K. Kobilka, Isabel Rojo, James Ficorilli, Bingfa Sun, Aaron D. Showalter, Qi Chen, Cynthia Stutsman, Dan Feng, Kyle W. Sloop, Michal Vieth, Alma Jiménez, David B. Wainscott, and Graham R Cumming
- Subjects
0303 health sciences ,Allosteric modulator ,Chemistry ,Drug discovery ,030302 biochemistry & molecular biology ,Allosteric regulation ,Cell Biology ,Ligand (biochemistry) ,03 medical and health sciences ,Heterotrimeric G protein ,Biophysics ,Receptor ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,Glucagon-like peptide 1 receptor ,030304 developmental biology ,G protein-coupled receptor - Abstract
Drugs that promote the association of protein complexes are an emerging therapeutic strategy. We report discovery of a G protein-coupled receptor (GPCR) ligand that stabilizes an active state conformation by cooperatively binding both the receptor and orthosteric ligand, thereby acting as a 'molecular glue'. LSN3160440 is a positive allosteric modulator of the GLP-1R optimized to increase the affinity and efficacy of GLP-1(9-36), a proteolytic product of GLP-1(7-36). The compound enhances insulin secretion in a glucose-, ligand- and GLP-1R-dependent manner. Cryo-electron microscopy determined the structure of the GLP-1R bound to LSN3160440 in complex with GLP-1 and heterotrimeric Gs. The modulator binds high in the helical bundle at an interface between TM1 and TM2, allowing access to the peptide ligand. Pharmacological characterization showed strong probe dependence of LSN3160440 for GLP-1(9-36) versus oxyntomodulin that is driven by a single residue. Our findings expand protein-protein modulation drug discovery to uncompetitive, active state stabilizers for peptide hormone receptors.
- Published
- 2020
- Full Text
- View/download PDF
20. Structural determinants of dual incretin receptor agonism by tirzepatide
- Author
-
Bingfa Sun, Francis S. Willard, Dan Feng, Jorge Alsina-Fernandez, Qi Chen, Michal Vieth, Joseph D. Ho, Aaron D. Showalter, Cynthia Stutsman, Liyun Ding, Todd M. Suter, James D. Dunbar, John W. Carpenter, Faiz Ahmad Mohammed, Eitaro Aihara, Robert A. Brown, Ana B. Bueno, Paul J. Emmerson, Julie S. Moyers, Tong Sun Kobilka, Matthew P. Coghlan, Brian K. Kobilka, and Kyle W. Sloop
- Subjects
endocrine system ,Multidisciplinary ,Diabetes Mellitus, Type 2 ,digestive, oral, and skin physiology ,Humans ,Gastric Inhibitory Polypeptide ,Incretins ,hormones, hormone substitutes, and hormone antagonists ,Glucagon-Like Peptide-1 Receptor ,Receptors, Gastrointestinal Hormone - Abstract
Significance Tirzepatide is a dual agonist of the glucose-dependent insulinotropic polypeptide receptor (GIPR) and the glucagon-like peptide-1 receptor (GLP-1R), which are incretin receptors that regulate carbohydrate metabolism. This investigational agent has proven superior to selective GLP-1R agonists in clinical trials in subjects with type 2 diabetes mellitus. Intriguingly, although tirzepatide closely resembles native GIP in how it activates the GIPR, it differs markedly from GLP-1 in its activation of the GLP-1R, resulting in less agonist-induced receptor desensitization. We report how cryogenic electron microscopy and molecular dynamics simulations inform the structural basis for the unique pharmacology of tirzepatide. These studies reveal the extent to which fatty acid modification, combined with amino acid sequence, determines the mode of action of a multireceptor agonist.
- Published
- 2022
21. A fast coarse filtering method for peptide identification by mass spectrometry.
- Author
-
Smriti R. Ramakrishnan, Rui Mao 0001, Aleksey A. Nakorchevskiy, John T. Prince, Willard S. Willard, Weijia Xu, Edward M. Marcotte, and Daniel P. Miranker
- Published
- 2006
- Full Text
- View/download PDF
22. LY3437943, a novel triple glucagon, GIP, and GLP-1 receptor agonist for glycemic control and weight loss: From discovery to clinical proof of concept
- Author
-
Tamer Coskun, Shweta Urva, William C. Roell, Hongchang Qu, Corina Loghin, Julie S. Moyers, Libbey S. O’Farrell, Daniel A. Briere, Kyle W. Sloop, Melissa K. Thomas, Valentina Pirro, David B. Wainscott, Francis S. Willard, Matthew Abernathy, LaRonda Morford, Yu Du, Charles Benson, Ruth E. Gimeno, Axel Haupt, and Zvonko Milicevic
- Subjects
Physiology ,Body Weight ,Mice, Obese ,Cell Biology ,Gastric Inhibitory Polypeptide ,Glycemic Control ,Glucagon ,Glucagon-Like Peptide-1 Receptor ,Receptors, Gastrointestinal Hormone ,Mice ,Weight Loss ,Receptors, Glucagon ,Animals ,Molecular Biology - Abstract
With an increasing prevalence of obesity, there is a need for new therapies to improve body weight management and metabolic health. Multireceptor agonists in development may provide approaches to fulfill this unmet medical need. LY3437943 is a novel triple agonist peptide at the glucagon receptor (GCGR), glucose-dependent insulinotropic polypeptide receptor (GIPR), and glucagon-like peptide-1 receptor (GLP-1R). In vitro, LY3437943 shows balanced GCGR and GLP-1R activity but more GIPR activity. In obese mice, administration of LY3437943 decreased body weight and improved glycemic control. Body weight loss was augmented by the addition of GCGR-mediated increases in energy expenditure to GIPR- and GLP-1R-driven calorie intake reduction. In a phase 1 single ascending dose study, LY3437943 showed a safety and tolerability profile similar to other incretins. Its pharmacokinetic profile supported once-weekly dosing, and a reduction in body weight persisted up to day 43 after a single dose. These findings warrant further clinical assessment of LY3437943.
- Published
- 2022
23. 679-P: The Novel GIP, GLP-1, and Glucagon Triple Receptor Agonist LY3437943 Exhibits Robust Efficacy in Preclinical Models of Obesity and Diabetes
- Author
-
David B. Wainscott, Francis S. Willard, Fariba Norouziyan Cooper, James Ficorilli, Kyle W. Sloop, Jorge Alsina-Fernandez, William C. Roell, Tamer Coskun, Over Cabrera, Shweta Urva, Aaron D. Showalter, Libbey S. O’Farrell, Xiaoping Ruan, Hongchang Qu, Julie S. Moyers, Ajit Regmi, and Lili Guo
- Subjects
Agonist ,medicine.medical_specialty ,Gastric emptying ,business.industry ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Incretin ,medicine.disease ,Glucagon ,Obesity ,law.invention ,Endocrinology ,law ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Recombinant DNA ,business ,Diet-induced obese - Abstract
The ever-growing prevalence of obesity and its associated comorbidities (T2D, NASH/NAFLD) is driving the need to discover new therapies for improving metabolic health. Recently, multi-receptor agonists have offered promise for meeting this need. Here, we characterize LY3437943, a novel single agent tri-agonist at the GIP, GLP-1, and glucagon (Gcg) receptors (R). Pharmacologic analysis of LY3437943 in cAMP assays using recombinant cell lines expressing the individual receptors indicated a potency balance favoring GIPR agonism (1.7- and 2.5-fold less potent at the GLP-1R and GcgR, respectively, but 7-fold more potent at the GIPR; all potencies in relation to the native ligands). In endogenous cells, LY3437943 regulated adipocyte lipolysis and hepatocyte glucose output. In vivo studies demonstrated regulation of multiple metabolic endpoints. Acute treatment with LY3437943 dose-dependently inhibited semi-liquid gastric emptying in mice and enhanced glucose dependent insulin secretion in rat IVGTT experiments. Chronic studies in diet induced obese mice reduced food intake and body weight (45% weight loss primarily via reduced fat mass) superior to other GIPR and GLP-1R agonists. In these experiments, LY3437943 lowered blood glucose and plasma insulin, indicating improved insulin sensitivity. Additionally, chronic administration improved biomarkers of liver health, decreasing both plasma alanine aminotransferase and liver triglycerides. Rodent and cynomolgus monkey PK modeling also suggested the potential for weekly dosing in humans. Taken these findings together, LY3437943 is a novel tri-agonist at the GIPR, GLP-1R, and GcgR, producing superior weight loss and glycemic control compared with other incretin receptor-targeting molecules and offers additional benefit for liver health. These findings prompt evaluation of the potential clinical benefit of LY3437943 in patients with obesity and metabolic diseases. Disclosure T. Coskun: Employee; Self; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company. F. S. Willard: Employee; Self; Eli Lilly and Company. J. V. Ficorilli: None. O. Cabrera: None. S. Urva: Employee; Self; Eli Lilly and Company. F. Norouziyan cooper: None. L. Guo: Employee; Self; Eli Lilly and Company. J. Alsina-fernandez: None. H. Qu: Employee; Self; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company. J. S. Moyers: Employee; Self; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company. W. C. Roell: Employee; Self; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company. L. O’farrell: None. A. Regmi: Employee; Self; Eli Lilly and Company. X. Ruan: None. A. D. Showalter: None. K. Sloop: Employee; Self; Eli Lilly and Company. D. B. Wainscott: Employee; Self; Eli Lilly and Company, Employee; Spouse/Partner; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company, Stock/Shareholder; Spouse/Partner; Eli Lilly and Company. Funding Eli Lilly and Company
- Published
- 2021
- Full Text
- View/download PDF
24. 639-P: GIP Receptor Agonism Enhances Weight Loss from Either a Biased or an Unbiased GLP-1 Receptor Agonist in DIO Mice
- Author
-
Jorge Alsina-Fernandez, Francis S. Willard, Cynthia Stutsman, Kyle W. Sloop, Matthew P. Coghlan, Guemalli R. Cardona, Tamer Coskun, Aaron D. Showalter, Libbey S. O’Farrell, Over Cabrera, and David B. Wainscott
- Subjects
medicine.medical_specialty ,Endocrinology ,GIP receptor ,Chemistry ,Weight loss ,Endocrinology, Diabetes and Metabolism ,Internal medicine ,Internal Medicine ,medicine ,Agonism ,medicine.symptom ,Glucagon-like peptide 1 receptor - Abstract
GIP receptor (GIPR) agonism enhances the reduction of food intake and weight loss induced by GLP-1 receptor (GLP-1R) agonism. Recently, GLP-1R agonists have been described that exhibit biased agonism as determined using cells engineered to facilitate measuring the two canonical signaling pathways engaged upon binding the GLP-1R. Such “biased agonists” retain the ability to activate the G alpha S/cyclic AMP (cAMP) pathway to a similar magnitude as native GLP-1 but exhibit markedly weaker ability to induce receptor recruitment of beta-arrestin. The prototype biased GLP-1R agonist Exendin-Phe1 (Ex-Phe1) is reported to exhibit greater weight and glucose control in diet-induced obese (DIO) mice than its unbiased parent Exendin-4 (Ex4). Herein, we investigated whether the enhanced weight loss of Ex-Phe1 erodes the ability of GIPR agonism to further enhance the efficacy of GLP-1R agonism. The peptides were first characterized in vitro to validate the biased nature of Ex-Ph1. In cells expressing either the human or murine GLP-1R, Ex-Phe1 activated cAMP signaling to a similar magnitude as GLP-1 and Ex4 but with approximately 5-fold lower potency than Ex4. The maximum effect of Ex-Phe1 upon human or mouse GLP-1R recruitment of beta-arrestin was 20% compared with full efficacy for Ex4 relative to GLP-1. In this assay Ex-Phe1 was marginally less potent than Ex4. In a 14-day osmotic minipump DIO mice study, Ex4 and Ex-Phe1 each dose dependently reduced food intake and lowered body weight. Ex-Phe1 was 10-fold more potent than Ex4, and at the maximum dose, delivered superior weight loss. Combination treatment with d-Ala-GIP resulted in similar additional body weight loss to that achieved by either Ex-Phe1 or Ex4 alone by enhancing food intake reduction and increasing energy expenditure. D-Ala-GIP alone had no effect on body weight. These findings indicate that GIPR agonism enhances GLP-1R agonism mediated weight loss irrespective of the pathway bias nature of the latter. Disclosure M. P. Coghlan: Employee; Self; Eli Lilly and Company. K. Sloop: Employee; Self; Eli Lilly and Company. T. Coskun: Employee; Self; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company. L. O’farrell: None. A. D. Showalter: None. D. B. Wainscott: Employee; Self; Eli Lilly and Company, Employee; Spouse/Partner; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company, Stock/Shareholder; Spouse/Partner; Eli Lilly and Company. C. Stutsman: None. G. Cardona: Employee; Self; Eli Lilly and Company, Stock/Shareholder; Self; Eli Lilly and Company. O. Cabrera: None. J. Alsina-fernandez: None. F. S. Willard: Employee; Self; Eli Lilly and Company. Funding Eli Lilly and Company
- Published
- 2021
- Full Text
- View/download PDF
25. Glucagon-like peptide-1 receptor ligand interactions: structural cross talk between ligands and the extracellular domain.
- Author
-
Graham M West, Francis S Willard, Kyle W Sloop, Aaron D Showalter, Bruce D Pascal, and Patrick R Griffin
- Subjects
Medicine ,Science - Abstract
Activation of the glucagon-like peptide-1 receptor (GLP-1R) in pancreatic β-cells potentiates insulin production and is a current therapeutic target for the treatment of type 2 diabetes mellitus (T2DM). Like other class B G protein-coupled receptors (GPCRs), the GLP-1R contains an N-terminal extracellular ligand binding domain. N-terminal truncations on the peptide agonist generate antagonists capable of binding to the extracellular domain, but not capable of activating full length receptor. The main objective of this study was to use Hydrogen/deuterium exchange (HDX) to identify how the amide hydrogen bonding network of peptide ligands and the extracellular domain of GLP-1R (nGLP-1R) were altered by binding interactions and to then use this platform to validate direct binding events for putative GLP-1R small molecule ligands. The HDX studies presented here for two glucagon-like peptide-1 receptor (GLP-1R) peptide ligands indicates that the antagonist exendin-4[9-39] is significantly destabilized in the presence of nonionic detergents as compared to the agonist exendin-4. Furthermore, HDX can detect stabilization of exendin-4 and exendin-4[9-39] hydrogen bonding networks at the N-terminal helix [Val19 to Lys27] upon binding to the N-terminal extracellular domain of GLP-1R (nGLP-1R). In addition we show hydrogen bonding network stabilization on nGLP-1R in response to ligand binding, and validate direct binding events with the extracellular domain of the receptor for putative GLP-1R small molecule ligands.
- Published
- 2014
- Full Text
- View/download PDF
26. Selective Phosphodiesterase 1 Inhibitor BTTQ Reduces Blood Pressure in Spontaneously Hypertensive and Dahl Salt Sensitive Rats: Role of Peripheral Vasodilation
- Author
-
Sherif Khedr, Alexander Staruschenko, Tamika D. Meredith, Xin Zhou, Kevin Michael Ruley, Mark C. Kowala, Mark David Rekhter, Simon J. Atkinson, Cynthia Darshini Jesudason, James S. Bean, Asim Bikash Dey, Maia Terashvili, Leah L. Porras, Michael Robert Wiley, Joseph V Hass, and Francis S. Willard
- Subjects
0301 basic medicine ,arterial hypertension ,spontaneously hypertensive rat ,Physiology ,Dahl salt sensitive rat ,phosphodiesterase 1 ,Vasodilation ,030204 cardiovascular system & hematology ,Pharmacology ,lcsh:Physiology ,03 medical and health sciences ,0302 clinical medicine ,Spontaneously hypertensive rat ,Physiology (medical) ,medicine ,vasodilation ,Mesenteric arteries ,lcsh:QP1-981 ,Chemistry ,Lisinopril ,Phosphodiesterase ,030104 developmental biology ,Blood pressure ,medicine.anatomical_structure ,ACE inhibitor ,Vascular resistance ,medicine.drug - Abstract
Regulation of the peripheral vascular resistance via modulating the vessel diameter has been considered as a main determinant of the arterial blood pressure. Phosphodiesterase enzymes (PDE1-11) hydrolyse cyclic nucleotides, which are key players controlling the vessel diameter and, thus, peripheral resistance. Here, we have tested and reported the effects of a novel selective PDE1 inhibitor (BTTQ) on the cardiovascular system. Normal Sprague Dawley, spontaneously hypertensive (SHR), and Dahl salt-sensitive rats were used to test in vivo the efficacy of the compound. Phosphodiesterase radiometric enzyme assay revealed that BTTQ inhibited all three isoforms of PDE1 in nanomolar concentration, while micromolar concentrations were needed to induce effective inhibition for other PDEs. The myography study conducted on mesenteric arteries revealed a potent vasodilatory effect of the drug, which was confirmed in vivo by an increase in the blood flow in the rat ear arteriols reflected by the rise in the temperature. Furthermore, BTTQ proved a high efficacy in lowering the blood pressure about 9, 36, and 24 mmHg in normal Sprague Dawley, SHR and, Dahl salt-sensitive rats, respectively, compared to the vehicle-treated group. Moreover, additional blood pressure lowering of about 22 mmHg could be achieved when BTTQ was administered on top of ACE inhibitor lisinopril, a current standard of care in the treatment of hypertension. Therefore, PDE1 inhibition induced efficient vasodilation that was accompanied by a significant reduction of blood pressure in different hypertensive rat models. Administration of BTTQ was also associated with increased heart rate in both models of hypertension as well as in the normotensive rats. Thus, PDE1 appears to be an attractive therapeutic target for the treatment of resistant hypertension, while tachycardia needs to be addressed by further compound structural optimization.
- Published
- 2020
- Full Text
- View/download PDF
27. Synthetic protease-activated class B GPCRs
- Author
-
Tamika D. Meredith, Kyle W. Sloop, Francis S. Willard, Wenzhen Ma, Joseph D. Ho, J. Michael Sauder, and Aaron D. Showalter
- Subjects
0301 basic medicine ,Proteolysis ,medicine.medical_treatment ,Recombinant Fusion Proteins ,Biophysics ,Protein Engineering ,Transfection ,Biochemistry ,Glucagon-Like Peptide-1 Receptor ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Glucagon-Like Peptide 1 ,Insulin Secretion ,medicine ,Animals ,Humans ,Receptor ,Molecular Biology ,Peptide sequence ,G protein-coupled receptor ,Protease ,medicine.diagnostic_test ,Chemistry ,Cell Biology ,Ligand (biochemistry) ,Fusion protein ,Cell biology ,Rats ,030104 developmental biology ,HEK293 Cells ,030220 oncology & carcinogenesis ,Exenatide ,Linker ,hormones, hormone substitutes, and hormone antagonists ,Peptide Hydrolases - Abstract
G-protein coupled receptors (GPCRs) are the ligand detection machinery of a majority of extracellular signaling systems in metazoans. Novel chemical and biological tools to probe the structure-function relationships of GPCRs have impacted both basic and applied GPCR research. To better understand the structure-function of class B GPCRs, we generated receptor-ligand fusion chimeric proteins that can be activated by exogenous enzyme application. As a prototype, fusion proteins of the glucagon-like peptide-1 receptor (GLP-1R) with GLP-1(7-36) and exendin-4(1-39) peptides incorporating enterokinase-cleavable N-termini were generated. These receptors are predicted to generate fusion protein neo-epitopes upon proteolysis with enterokinase that are identical to the N-termini of GLP-1 agonists. This system was validated by measuring enterokinase-dependent GLP-1R mediated cAMP accumulation, and a structure-activity relationship for both linker length and peptide sequence was observed. Moreover, our results show this approach can be used in physiologically relevant cell systems, as GLP-1R-ligand chimeras were shown to induce glucose-dependent insulin secretion in insulinoma cells upon exposure to enterokinase. This approach suggests new strategies for understanding the structure-function of peptide-binding GPCRs.
- Published
- 2020
28. 324-OR: The Incretin Action of GIP Is Mediated through Both Alpha and Beta Cells
- Author
-
Kimberley El, Paul Emmerson, David A. D'Alessio, Kyle W. Sloop, Jennifer L. Brown, Megan Capozzi, Sarah M. Gray, Francis S. Willard, and Jonathan Campbell
- Subjects
0301 basic medicine ,Amino acid activation ,endocrine system ,medicine.medical_specialty ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Glucagon secretion ,Incretin ,030209 endocrinology & metabolism ,Proglucagon ,Glucagon ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Postprandial ,Internal medicine ,Internal Medicine ,medicine ,Secretion ,Receptor ,hormones, hormone substitutes, and hormone antagonists - Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) are the principal incretins connecting nutrient intake to postprandial insulin secretion. Both GIP and GLP-1 directly stimulate insulin secretion in β-cells through cognate receptors (GIPR; GLP-1R) in a glucose-dependent manner. The GIPR is also expressed on α-cells and GIP stimulates glucagon secretion. α- to β-cell communication, manifest as activation of β-cell GLP-1R by proglucagon peptides (PGDP), dictates the magnitude of the insulin response. We hypothesized that GIPR-stimulation of PGDP enhances insulin secretion through α- to β-cell communication, and that GIPR activity in α-cells is nutrient dependent, similar to β-cells. Mouse islets perifused with GIP or alanine alone doubled their secretion of glucagon, but the combination produced ∼10x glucagon secretion, a synergistic effect that was present at both 2.8 and 10 mM glucose. At 10 mM glucose, GIP plus alanine also had a synergistic effect on insulin secretion. This synergy was abolished by the GLP-1R antagonist exendin-(9-39) indicating that amino acid enhancement of GIP-stimulated insulin secretion is dependent on α- to β-cell communication. In vivo, mice with α-cell deletion of GIPR had normal intraperitoneal and oral glucose tolerance, but impaired glycemic control and reduced insulin secretion in response to a mixed nutrient stimulus, a condition that enhances α-cell activity. Together, these results demonstrate that GIPR activity in α-cells is dependent upon amino acid activation and contributes significantly to insulin secretion through α- to β-cell communication. Disclosure K.M. El: None. M. Capozzi: None. S.M. Gray: None. J.L. Brown: None. F.S. Willard: None. P. Emmerson: None. K. Sloop: None. D. D’Alessio: Advisory Panel; Self; Eli Lilly and Company. Consultant; Self; Intarcia Therapeutics. Research Support; Self; Ansh Labs, Eli Lilly and Company, Merck Sharp & Dohme Corp. Other Relationship; Self; Novo Nordisk A/S. J. Campbell: Research Support; Self; Eli Lilly and Company, Novo Nordisk Inc. Speaker’s Bureau; Self; Merck Sharp & Dohme Corp. Funding American Diabetes Association (1-17-JDF-074 to J.C.)
- Published
- 2020
- Full Text
- View/download PDF
29. Discovery and pharmacology of the covalent GLP-1 receptor (GLP-1R) allosteric modulator BETP: A novel tool to probe GLP-1R pharmacology
- Author
-
Francis S, Willard, Joseph D, Ho, and Kyle W, Sloop
- Subjects
Small Molecule Libraries ,Pyrimidines ,Allosteric Regulation ,Drug Discovery ,Cyclic AMP ,Animals ,Humans ,Amino Acid Sequence ,Glucagon-Like Peptide-1 Receptor - Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a significant therapeutic target for small molecule drug discovery given the therapeutic impact of peptide agonists in the diabetes sphere. We review the discovery and subsequent characterization of the small molecule GLP-1R allosteric modulator 4-(3-(Benzyloxy)phenyl)-2-(ethylsulfinyl)-6-(trifluoromethyl)pyrimidine (BETP). BETP is a covalent modulator of the GLP-1R, and we discuss the pharmacological implications and possible structural basis of this novel mode of action. We highlight the insights into class B G-protein coupled receptor pharmacology and biology provided by studies conducted with BETP. These include the descriptions of exquisite allosteric modulator probe dependence and biased signaling in vitro and in vivo. We conclude with an analysis of the utility of BETP as a chemical probe for the GLP-1R.
- Published
- 2020
30. A P-loop mutation in Gα subunits prevents transition to the active state: implications for G-protein signaling in fungal pathogenesis.
- Author
-
Dustin E Bosch, Francis S Willard, Ravikrishna Ramanujam, Adam J Kimple, Melinda D Willard, Naweed I Naqvi, and David P Siderovski
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Heterotrimeric G-proteins are molecular switches integral to a panoply of different physiological responses that many organisms make to environmental cues. The switch from inactive to active Gαβγ heterotrimer relies on nucleotide cycling by the Gα subunit: exchange of GTP for GDP activates Gα, whereas its intrinsic enzymatic activity catalyzes GTP hydrolysis to GDP and inorganic phosphate, thereby reverting Gα to its inactive state. In several genetic studies of filamentous fungi, such as the rice blast fungus Magnaporthe oryzae, a G42R mutation in the phosphate-binding loop of Gα subunits is assumed to be GTPase-deficient and thus constitutively active. Here, we demonstrate that Gα(G42R) mutants are not GTPase deficient, but rather incapable of achieving the activated conformation. Two crystal structure models suggest that Arg-42 prevents a typical switch region conformational change upon Gα(i1)(G42R) binding to GDP·AlF(4)(-) or GTP, but rotameric flexibility at this locus allows for unperturbed GTP hydrolysis. Gα(G42R) mutants do not engage the active state-selective peptide KB-1753 nor RGS domains with high affinity, but instead favor interaction with Gβγ and GoLoco motifs in any nucleotide state. The corresponding Gα(q)(G48R) mutant is not constitutively active in cells and responds poorly to aluminum tetrafluoride activation. Comparative analyses of M. oryzae strains harboring either G42R or GTPase-deficient Q/L mutations in the Gα subunits MagA or MagB illustrate functional differences in environmental cue processing and intracellular signaling outcomes between these two Gα mutants, thus demonstrating the in vivo functional divergence of G42R and activating G-protein mutants.
- Published
- 2012
- Full Text
- View/download PDF
31. Beyond Glucagon-like Peptide-1: Is G-Protein Coupled Receptor Polypharmacology the Path Forward to Treating Metabolic Diseases?
- Author
-
Daniel A. Briere, Paul J. Emmerson, Francis S. Willard, and Kyle W. Sloop
- Subjects
0301 basic medicine ,Pharmacology ,business.industry ,Drug discovery ,Insulin ,medicine.medical_treatment ,Type 2 Diabetes Mellitus ,030209 endocrinology & metabolism ,Context (language use) ,Glucagon ,Glucagon-like peptide-1 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Medicine ,Pharmacology (medical) ,Receptor ,business ,G protein-coupled receptor - Abstract
[Image: see text] The glucagon-like peptide-1 receptor (GLP-1R) is a class B G-protein coupled receptor (GPCR) that has proven to be an effective target for developing medicines that treat type 2 diabetes mellitus (T2DM). GLP-1R agonists improve T2DM by enhancing glucose-stimulated insulin secretion, delaying gastric transit, decreasing glucagon levels, and reducing body weight due to anorexigenic actions. The therapeutic successes of these agents helped inspire the design of new multifunctional molecules that are GLP-1R agonists but also activate receptors linked to pathways that enhance insulin sensitization and/or energy expenditure. Herein, these agents are discussed in the context of polypharmacological approaches that may enable even further improvement in treatment outcomes. Moreover, we revisit classical polypharmaceutical GPCR approaches and how they may be utilized for treatment of T2DM. To determine optimal combination regimens, changes in drug discovery practices are likely needed because compensatory mechanisms appear to underlie progression of T2DM and limit the ability of current therapies to induce disease regression or remission.
- Published
- 2018
- Full Text
- View/download PDF
32. The current state of GPCR-based drug discovery to treat metabolic disease
- Author
-
Kyle W. Sloop, Michael A. Statnick, Francis S. Willard, and Paul J. Emmerson
- Subjects
0301 basic medicine ,Pharmacology ,Drug discovery ,business.industry ,Human immunodeficiency virus (HIV) ,Type 2 Diabetes Mellitus ,Disease ,Molecular Pharmacology ,Bioinformatics ,medicine.disease_cause ,03 medical and health sciences ,030104 developmental biology ,medicine ,Metabolic disease ,business - Abstract
One approach of modern drug discovery is to identify agents that enhance or diminish signal transduction cascades in various cell types and tissues by modulating the activity of GPCRs. This strategy has resulted in the development of new medicines to treat many conditions, including cardiovascular disease, psychiatric disorders, HIV/AIDS, certain forms of cancer and Type 2 diabetes mellitus (T2DM). These successes justify further pursuit of GPCRs as disease targets and provide key learning that should help guide identifying future therapeutic agents. This report reviews the current landscape of GPCR drug discovery with emphasis on efforts aimed at developing new molecules for treating T2DM and obesity. We analyse historical efforts to generate GPCR-based drugs to treat metabolic disease in terms of causal factors leading to success and failure in this endeavour. Linked articles This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
- Published
- 2018
- Full Text
- View/download PDF
33. Regulation of Endogenous (Male) Rodent GLP-1 Secretion and Human Islet Insulin Secretion by Antagonism of Somatostatin Receptor 5
- Author
-
Marta Adeva, Samreen K. Syed, Thomas B. Farb, Xin Zhou, Thomas James Beauchamp, David Andrew Coates, Francis S. Willard, Tamika D. Meredith, Susan L. Gackenheimer, Michael A. Statnick, Maria Angeles Martinez-Grau, Miguel A. Toledo, Brian A. Droz, James Ficorilli, Alexander M. Efanov, Gema Ruano, Over Cabrera, Todd M. Suter, Victoriano Molero, Krister Bokvist, and David Gene Barrett
- Subjects
Male ,0301 basic medicine ,endocrine system ,medicine.medical_specialty ,medicine.medical_treatment ,Incretin ,CHO Cells ,Benzoates ,Rats, Sprague-Dawley ,Islets of Langerhans ,Mice ,03 medical and health sciences ,Cricetulus ,Endocrinology ,Glucagon-Like Peptide 1 ,Cricetinae ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Insulin ,Somatostatin receptor 2 ,Spiro Compounds ,Somatostatin receptor 1 ,Receptors, Somatostatin ,education ,Cells, Cultured ,Glycemic ,Mice, Knockout ,education.field_of_study ,Secretory Pathway ,Somatostatin receptor-5 ,business.industry ,Rats ,Rats, Zucker ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Somatostatin ,Antagonism ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
Incretin and insulin responses to nutrient loads are suppressed in persons with diabetes, resulting in decreased glycemic control. Agents including sulfonylureas and dipeptidyl peptidase-4 inhibitors (DPP4i) partially reverse these effects and provide therapeutic benefit; however, their modes of action limit efficacy. Because somatostatin (SST) has been shown to suppress insulin and glucagonlike peptide-1 (GLP-1) secretion through the Gi-coupled SST receptor 5 (SSTR5) isoform in vitro, antagonism of SSTR5 may improve glycemic control via intervention in both pathways. Here, we show that a potent and selective SSTR5 antagonist reverses the blunting effects of SST on insulin secretion from isolated human islets, and demonstrate that SSTR5 antagonism affords increased levels of systemic GLP-1 in vivo. Knocking out Sstr5 in mice provided a similar increase in systemic GLP-1 levels, which were not increased further by treatment with the antagonist. Treatment of mice with the SSTR5 antagonist in combination with a DPP4i resulted in increases in systemic GLP-1 levels that were more than additive and resulted in greater glycemic control compared with either agent alone. In isolated human islets, the SSTR5 antagonist completely reversed the inhibitory effect of exogenous SST-14 on insulin secretion. Taken together, these data suggest that SSTR5 antagonism should increase circulating GLP-1 levels and stimulate insulin secretion (directly and via GLP-1) in humans, improving glycemic control in patients with diabetes.
- Published
- 2017
- Full Text
- View/download PDF
34. G protein-coupled receptor allosteric modulators
- Author
-
David B. Wainscott and Francis S. Willard
- Subjects
Agonist ,Allosteric modulator ,medicine.drug_class ,Chemistry ,Allosteric regulation ,medicine ,Biophysics ,Cooperativity ,Receptor ,G protein-coupled receptor - Abstract
Allosteric modulators represent a key modality to drug G protein-coupled receptors. In this chapter, we highlight the role of allosteric modulator affinity (KB) in defining the pharmacological profile necessary for in vivo efficacy. Using simulation approaches with the Stockton-Ehlert and the allosteric operational models, we investigate the role of KB in allosteric modulator action. The relationship between orthosteric agonist concentration, affinity, and allosteric parameters defines the pharmaceutical potential of positive and negative allosteric modulators. Simulations demonstrate that positive allosteric modulators with weak affinities or low exposures may be compensated by having high allosteric cooperativity. To understand the pharmacology of G protein-coupled receptor allosteric modulators, we need to obtain information about the in vivo concentrations of orthosteric agonists and coupling efficiencies of endogenous tissues. Quantification and optimization of the KB parameter within allosteric modulator structure-activity relationships is an underappreciated aspect of allosteric modulator drug design. The development of radioligands and binding assays to determine allosteric modulator affinities will greatly facilitate these efforts. Modulator affinity is a key determinant of pharmacological dose and manipulation of this will allow the design of allosteric modulators with superior pharmacological properties, and thus, the generation of effective allosteric GPCR-directed medicines.
- Published
- 2020
- Full Text
- View/download PDF
35. Contributors
- Author
-
Waseem Imtiaz Ahmad, Demet Araç, Patricio Atanes, Bellinda Benhamú, Apurba Bhattarai, Célia A. Bouazza, Robert Cerchio, Steven J. Charlton, Amitabha Chattopadhyay, Suzie Chen, Arnau Cordomí, George R. Dubyak, Frederick J. Ehlert, Treyton S. Farnan, Sławomir Filipek, Rafael Franco, Karolina Gherbi, Javier González-Maeso, Nickolaj J. Groenewoud, Thais Rafael Guimarães, Xu Han, Terence E. Hébert, Nicholas D. Holliday, Jakub Jakowiecki, Beata Jastrzebska, Karen Priyadarshini John, Sadashiva S. Karnik, Peter Keov, Amanuel Kibrom, Elizabeth A. Knauss, Irina Kufareva, G. Aditya Kumar, Steven La Plante, Sacha Thierry Larda, Katherine Leon, Jing Li, María L. López-Rodríguez, Ryan D. Martin, Yinglong Miao, Przemysław Miszta, Madhura Mohole, Gemma Navarro, Tony Ngo, Marvin T. Nieman, Szymon Niewieczerzał, Joseph T. Ortega, Aditya Pandey, Leonardo Pardo, Paul S.-H. Park, Shanta J. Persaud, Robert Scott Prosser, Xiangli Qu, Sudarshan Rajagopal, Kathy Sengmany, Durba Sengupta, Salvador Sierra, Khuraijam Dhanachandra Singh, Nicola J. Smith, Sean S. So, Amantha Thathiah, Rudy Toneatti, Henar Vázquez-Villa, Gayathri Viswanathan, David B. Wainscott, Jinan Wang, Dejian Wang, Francis S. Willard, Donna S. Woulfe, Beili Wu, and Kevin Zheng
- Published
- 2020
- Full Text
- View/download PDF
36. Discovery and pharmacology of the covalent GLP-1 receptor (GLP-1R) allosteric modulator BETP: A novel tool to probe GLP-1R pharmacology
- Author
-
Kyle W. Sloop, Francis S. Willard, and Joseph D. Ho
- Subjects
chemistry.chemical_classification ,Agonist ,Allosteric modulator ,chemistry ,medicine.drug_class ,Drug discovery ,medicine ,Biophysics ,Peptide ,Receptor ,Small molecule ,Glucagon-like peptide 1 receptor ,G protein-coupled receptor - Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a significant therapeutic target for small molecule drug discovery given the therapeutic impact of peptide agonists in the diabetes sphere. We review the discovery and subsequent characterization of the small molecule GLP-1R allosteric modulator 4-(3-(Benzyloxy)phenyl)-2-(ethylsulfinyl)-6-(trifluoromethyl)pyrimidine (BETP). BETP is a covalent modulator of the GLP-1R, and we discuss the pharmacological implications and possible structural basis of this novel mode of action. We highlight the insights into class B G-protein coupled receptor pharmacology and biology provided by studies conducted with BETP. These include the descriptions of exquisite allosteric modulator probe dependence and biased signaling in vitro and in vivo. We conclude with an analysis of the utility of BETP as a chemical probe for the GLP-1R.
- Published
- 2020
- Full Text
- View/download PDF
37. Structural insights into probe-dependent positive allosterism of the GLP-1 receptor
- Author
-
Ana B, Bueno, Bingfa, Sun, Francis S, Willard, Dan, Feng, Joseph D, Ho, David B, Wainscott, Aaron D, Showalter, Michal, Vieth, Qi, Chen, Cynthia, Stutsman, Betty, Chau, James, Ficorilli, Francisco J, Agejas, Graham R, Cumming, Alma, Jiménez, Isabel, Rojo, Tong Sun, Kobilka, Brian K, Kobilka, and Kyle W, Sloop
- Subjects
Models, Molecular ,Allosteric Regulation ,Molecular Structure ,Glucagon-Like Peptide 1 ,Protein Conformation ,Allosteric Site ,Glucagon-Like Peptide-1 Receptor - Abstract
Drugs that promote the association of protein complexes are an emerging therapeutic strategy. We report discovery of a G protein-coupled receptor (GPCR) ligand that stabilizes an active state conformation by cooperatively binding both the receptor and orthosteric ligand, thereby acting as a 'molecular glue'. LSN3160440 is a positive allosteric modulator of the GLP-1R optimized to increase the affinity and efficacy of GLP-1(9-36), a proteolytic product of GLP-1(7-36). The compound enhances insulin secretion in a glucose-, ligand- and GLP-1R-dependent manner. Cryo-electron microscopy determined the structure of the GLP-1R bound to LSN3160440 in complex with GLP-1 and heterotrimeric G
- Published
- 2019
38. Activation of the GLP-1 receptor by a non-peptidic agonist
- Author
-
Arthur Christopoulos, Patrick M. Sexton, Lachlan Clydesdale, Asuka Inoue, Yi Lynn Liang, Giuseppe Deganutti, Tin T. Truong, Matthew J. Belousoff, Michael E. Christe, Sebastian G.B. Furness, Michael Gregory Bell, Christopher A. Reynolds, Denise Wootten, Kyle W. Sloop, Ming-Wei Wang, Francis S. Willard, Peishen Zhao, Laurence J. Miller, Madeleine M. Fletcher, and Radostin Danev
- Subjects
0301 basic medicine ,Agonist ,Models, Molecular ,medicine.drug_class ,Pyridines ,Phenylalanine ,CHO Cells ,Glucagon-Like Peptide-1 Receptor ,03 medical and health sciences ,0302 clinical medicine ,Protein structure ,Cricetulus ,Cricetinae ,medicine ,Functional selectivity ,Extracellular ,Animals ,Humans ,Receptor ,Protein Structure, Quaternary ,Glucagon-like peptide 1 receptor ,Multidisciplinary ,Chemistry ,Ligand (biochemistry) ,Isoquinolines ,Protein Structure, Tertiary ,Transmembrane domain ,Kinetics ,030104 developmental biology ,Structural Homology, Protein ,Biophysics ,030217 neurology & neurosurgery - Abstract
Class B G-protein-coupled receptors are major targets for the treatment of chronic diseases, including diabetes and obesity1. Structures of active receptors reveal peptide agonists engage deep within the receptor core, leading to an outward movement of extracellular loop 3 and the tops of transmembrane helices 6 and 7, an inward movement of transmembrane helix 1, reorganization of extracellular loop 2 and outward movement of the intracellular side of transmembrane helix 6, resulting in G-protein interaction and activation2,3,4,5,6. Here we solved the structure of a non-peptide agonist, TT-OAD2, bound to the glucagon-like peptide-1 (GLP-1) receptor. Our structure identified an unpredicted non-peptide agonist-binding pocket in which reorganization of extracellular loop 3 and transmembrane helices 6 and 7 manifests independently of direct ligand interaction within the deep transmembrane domain pocket. TT-OAD2 exhibits biased agonism, and kinetics of G-protein activation and signalling that are distinct from peptide agonists. Within the structure, TT-OAD2 protrudes beyond the receptor core to interact with the lipid or detergent, providing an explanation for the distinct activation kinetics that may contribute to the clinical efficacy of this compound series. This work alters our understanding of the events that drive the activation of class B receptors.
- Published
- 2019
39. Selective Phosphodiesterase 1 Inhibitor LY1 Reduces Blood Pressure in Spontaneously Hypertensive and Dahl Salt Sensitive Rats
- Author
-
Tamika D. Meredith, Sherif Khedr, Mark David Rekhter, Simon Atkinson, Francis S. Willard, Leah L. Porras, Asim Bikash Dey, Michael Robert Wiley, Maia Terashvili, Mark C. Kowala, and Alexander Staruschenko
- Subjects
Dahl Salt-Sensitive Rats ,medicine.medical_specialty ,Endocrinology ,Blood pressure ,Chemistry ,Internal medicine ,Genetics ,medicine ,Phosphodiesterase ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2020
- Full Text
- View/download PDF
40. The 2nd National COPD Readmissions Summit and Beyond: From Theory to Implementation
- Author
-
Byron Thomashow, Rhonda Rotert, Kristen S. Willard, Leonard Fromer, Barbara P. Yawn, Jamie B Sullivan, Catherine 'Casey' S. Jones, Alpesh Amin, and Jean M. Rommes
- Subjects
Pulmonary and Respiratory Medicine ,Spirometry ,COPD ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Best practice ,medicine.medical_treatment ,Attendance ,medicine.disease ,respiratory tract diseases ,03 medical and health sciences ,National COPD Readmissions Summit ,0302 clinical medicine ,Quality of life (healthcare) ,030228 respiratory system ,medicine ,Pulmonary rehabilitation ,030212 general & internal medicine ,Intensive care medicine ,business ,Medicaid ,Simulation ,Patient education - Abstract
Chronic obstructive pulmonary disease (COPD) hospitalizations and readmissions adversely impact the health and quality of life of COPD patients. Under the Hospital Readmissions Reduction Program, the Centers for Medicare & Medicaid Services reduce payments to those hospitals exceeding expected rates of COPD readmissions within 30 days of hospital discharge. It was within this climate that the COPD Foundation held its 2nd COPD Readmissions Summit in March 2015. Experts in attendance: (1) categorized challenges to optimal COPD care, ( 2) analyzed the state of care delivery and readmissions reduction strategies and (3) identified the best available evidence-based approaches to improving care delivery across the continuum, including early diagnosis via spirometry, ongoing device, oxygen and medication reconciliation, treatment that addresses comorbidities and preventive care, robust patient education, prompt post-acute follow up, home health services and pulmonary rehabilitation. Results of this collaborative event formed the basis for PRAXIS, the COPD Foundation’s initiative to improve COPD care across the health continuum and to reduce readmissions.
- Published
- 2016
- Full Text
- View/download PDF
41. The insulin secretory action of novel polycyclic guanidines: Discovery through open innovation phenotypic screening, and exploration of structure–activity relationships
- Author
-
Michael B. Shaghafi, Francis S. Willard, Larry E. Overman, and David Gene Barrett
- Subjects
medicine.medical_treatment ,Phenotypic screening ,Clinical Biochemistry ,Pharmaceutical Science ,Guanidines ,Biochemistry ,Islets of Langerhans ,Structure-Activity Relationship ,chemistry.chemical_compound ,Diabetes mellitus ,Drug Discovery ,medicine ,Animals ,Insulin ,Polycyclic Compounds ,Guanidine ,Insulin secretion ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Drug discovery ,Pancreatic islets ,Diabetes ,Organic Chemistry ,medicine.disease ,Rats ,Glucose ,Phenotype ,medicine.anatomical_structure ,chemistry ,Molecular Medicine ,Insulin secretogogue - Abstract
We report the discovery of the glucose-dependent insulin secretogogue activity of a novel class of polycyclic guanidines through phenotypic screening as part of the Lilly Open Innovation Drug Discovery platform. Three compounds from the University of California, Irvine, 1–3, having the 3-arylhexahydropyrrolo[1,2-c]pyrimidin-1-amine scaffold acted as insulin secretagogues under high, but not low, glucose conditions. Exploration of the structure–activity relationship around the scaffold demonstrated the key role of the guanidine moiety, as well as the importance of two lipophilic regions, and led to the identification of 9h, which stimulated insulin secretion in isolated rat pancreatic islets in a glucose-dependent manner.
- Published
- 2014
- Full Text
- View/download PDF
42. Establishment and Characterization of a Highly Tumorigenic African American Prostate Cancer Cell Line, E006AA-hT
- Author
-
Gissou Azabdaftari, Stacey S. Willard, Mohammad Saleem, Shahriar Koochekpour, Kristopher Attwood, Chunhong Liu, Shafat Ali, and Mojgan Shourideh
- Subjects
Male ,Cell ,Biology ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Prostate cancer ,Cell Line, Tumor ,medicine ,Humans ,Epigenetics ,African American ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Genetics ,Cell growth ,Prostate Cancer ,tumorigenic ,Prostatic Neoplasms ,Karyotype ,E006AA ,Cell Biology ,medicine.disease ,3. Good health ,Black or African American ,medicine.anatomical_structure ,Receptors, Androgen ,Cell culture ,Androgen Receptor ,Cancer research ,Ploidy ,Carcinogenesis ,Research Paper ,Developmental Biology - Abstract
Genuine racial differences in prostate cancer (PCa) biology have been considered among the potential reasons to explain PCa disparities. There is no animal model to represent all aspects of human PCa and, more specifically, to be used for PCa disparity research. The lack of a spontaneously transformed in vitro cell-based model system has been a significant impediment to investigating and understanding potential molecular mechanisms, and the hormonal, genetic, and epigenetic factors underlying the biological and clinical aggressiveness of PCa in African American (AA) men. In this study, we established and characterized the E006AA-hT cell line as a highly tumorigenic subline of the previously characterized primary AA-PCa cell line, E006AA. Extensive characterization of the E006AA-hT cell line was accomplished using cytodifferentiation and prostate-specific markers, spectral karyotyping, cell line authentication assays, cell proliferation and migration assays, and in vitro tumorigenesis assays. Spectral karyotyping of E006AA-hT showed a hypertriploid chromosome complement and shared cytogenetic changes similar to its parental cells such as diploid X, absence of Y-chromosomes, numerical gains in chromosomes 5,6,8,10,17,20,21, and marker chromosomes of unknown origin. In addition, E006AA-hT also presented numerous clonal and structural aberrations such as insertion, deletion, duplication, and translocations in chromosomes 1-5, 8, 9, 11, 13, 14, 17, and 18. The E006AA-hT cell line was shown to be highly tumorigenic and produced tumors at an accelerated growth rate in both athymic nude and triple-deficient SCID mice. Silencing the mutated androgen receptor (AR-599 Ser>Gly) did not affect proliferation (loss-of-function), but decreased migration (gain-of-function) in E006AA-hT and its parental cell type. These data support that AR-point mutations may lead simultaneously to different “loss-of-function” and “gain-of-function” phenotypes in PCa cells. E006AA-Par and its subline as the only available spontaneously transformed low- and highly-tumorigenic primary AA-PCa cell lines could be used for basic and translational research aimed in supporting prostate cancer disparity research.
- Published
- 2014
- Full Text
- View/download PDF
43. Structural Determinants of RGS-RhoGEF Signaling Critical to Entamoeba histolytica Pathogenesis
- Author
-
Adam J. Kimple, Dustin E. Bosch, David P. Siderovski, Robin E. Muller, Francis S. Willard, Stephen L. Rogers, Alyssa J. Manning, and Mischa Machius
- Subjects
Models, Molecular ,Cell Survival ,G protein ,Protozoan Proteins ,Small G Protein ,Biology ,Crystallography, X-Ray ,Protein Structure, Secondary ,Article ,Cell Line ,Host-Parasite Interactions ,Entamoeba histolytica ,Regulator of G protein signaling ,Structural Biology ,Heterotrimeric G protein ,Cell Adhesion ,Animals ,Guanine Nucleotide Exchange Factors ,Protein Interaction Domains and Motifs ,Trophozoites ,Cell Shape ,Molecular Biology ,Binding Sites ,Effector ,Chemotaxis ,Hydrolysis ,biology.organism_classification ,Molecular biology ,GTP-Binding Protein alpha Subunits ,Cell biology ,Pleckstrin homology domain ,Drosophila melanogaster ,Amino Acid Substitution ,Mutagenesis, Site-Directed ,Guanosine Triphosphate ,Guanine nucleotide exchange factor ,Rho Guanine Nucleotide Exchange Factors ,Protein Binding ,Signal Transduction - Abstract
Summary G protein signaling pathways, as key components of physiologic responsiveness and timing, are frequent targets for pharmacologic intervention. Here, we identify an effector for heterotrimeric G protein α subunit (EhGα1) signaling from Entamoeba histolytica , the causative agent of amoebic colitis. EhGα1 interacts with this effector and guanosine triphosphatase-accelerating protein, EhRGS-RhoGEF, in a nucleotide state-selective fashion. Coexpression of EhRGS-RhoGEF with constitutively active EhGα1 and EhRacC leads to Rac-dependent spreading in Drosophila S2 cells. EhRGS-RhoGEF overexpression in E. histolytica trophozoites leads to reduced migration toward serum and lower cysteine protease activity, as well as reduced attachment to, and killing of, host cells. A 2.3 A crystal structure of the full-length EhRGS-RhoGEF reveals a putative inhibitory helix engaging the Dbl homology domain Rho-binding surface and the pleckstrin homology domain. Mutational analysis of the EhGα1/EhRGS-RhoGEF interface confirms a canonical "regulator of G protein signaling" domain rather than a RhoGEF-RGS ("rgRGS") domain, suggesting a convergent evolution toward heterotrimeric and small G protein cross-talk.
- Published
- 2013
- Full Text
- View/download PDF
44. Structural determinants of G-protein alpha subunit selectivity by regulator of G-protein signaling 2 (RGS2)
- Author
-
Adam J. Kimple, Francis S. Willard, David P. Siderovski, Meera Soundararajan, Stephanie Q. Hutsell, A.K. Roos, Vincent Setola, Brenda Temple, Bryan L. Roth, Daniel J. Urban, and Stefan Knapp
- Subjects
Models, Molecular ,Gs alpha subunit ,G protein ,GTP-Binding Protein alpha Subunits ,Protein subunit ,Recombinant Fusion Proteins ,Biology ,Transfection ,Biochemistry ,Cell Line ,Evolution, Molecular ,Regulator of G protein signaling ,Fluorescence Resonance Energy Transfer ,Humans ,Point Mutation ,Protein Interaction Domains and Motifs ,Molecular Biology ,RGS2 ,G protein-coupled receptor ,Binding Sites ,Hydrolysis ,Mechanisms of Signal Transduction ,Cell Biology ,Surface Plasmon Resonance ,Cell biology ,Protein Structure, Tertiary ,Luminescent Proteins ,sense organs ,Guanosine Triphosphate ,biological phenomena, cell phenomena, and immunity ,RGS Proteins ,hormones, hormone substitutes, and hormone antagonists ,Protein Binding - Abstract
"Regulator of G-protein signaling" (RGS) proteins facilitate the termination of G protein-coupled receptor (GPCR) signaling via their ability to increase the intrinsic GTP hydrolysis rate of Galpha subunits (known as GTPase-accelerating protein or "GAP" activity). RGS2 is unique in its in vitro potency and selectivity as a GAP for Galpha(q) subunits. As many vasoconstrictive hormones signal via G(q) heterotrimer-coupled receptors, it is perhaps not surprising that RGS2-deficient mice exhibit constitutive hypertension. However, to date the particular structural features within RGS2 determining its selectivity for Galpha(q) over Galpha(i/o) substrates have not been completely characterized. Here, we examine a trio of point mutations to RGS2 that elicits Galpha(i)-directed binding and GAP activities without perturbing its association with Galpha(q). Using x-ray crystallography, we determined a model of the triple mutant RGS2 in complex with a transition state mimetic form of Galpha(i) at 2.8-A resolution. Structural comparison with unliganded, wild type RGS2 and of other RGS domain/Galpha complexes highlighted the roles of these residues in wild type RGS2 that weaken Galpha(i) subunit association. Moreover, these three amino acids are seen to be evolutionarily conserved among organisms with modern cardiovascular systems, suggesting that RGS2 arose from the R4-subfamily of RGS proteins to have specialized activity as a potent and selective Galpha(q) GAP that modulates cardiovascular function.
- Published
- 2016
- Full Text
- View/download PDF
45. A measurement of the proton structure function F-2(x, Q(2)) at low x and low Q(2) at HERA
- Author
-
C. Adloff, S. Aid, M. Anderson, V. Andreev, B. Andrieu, V. Arkadov, C. Arndt, I. Ayyaz, A. Babaev, J. Bähr, J. Bán, Y. Ban, P. Baranov, E. Barrelet, R. Barschke, W. Bartel, U. Bassler, H.P. Beck, M. Beck, H.-J. Behrend, A. Belousov, Ch. Berger, G. Bernardi, G. Bertrand-Coremans, R. Beyer, P. Biddulph, P. Bispham, J.C. Bizot, K. Borras, F. Botterweck, V. Boudry, S. Bourov, A. Braemer, W. Braunschweig, V. Brisson, W. Brückner, P. Bruel, D. Bruncko, C. Brune, R. Buchholz, L. Büngener, J. Bürger, F.W. Büsser, A. Buniatian, S. Burke, M.J. Burton, G. Buschhorn, D. Calvet, A.J. Campbell, T. Carli, M. Charlet, D. Clarke, B. Clerbaux, S. Cocks, J.G. Contreras, C. Cormack, J.A. Coughlan, A. Courau, M.-C. Cousinou, B.E. Cox, G. Cozzika, D.G. Cussans, J. Cvach, S. Dagoret, J.B. Dainton, W.D. Dau, K. Daum, M. David, C.L. Davis, A. De Roeck, E.A. De Wolf, B. Delcourt, M. Dirkmann, P. Dixon, W. Dlugosz, C. Dollfus, K.T. Donovan, J.D. Dowell, H.B. Dreis, A. Droutskoi, J. Ebert, T.R. Ebert, G. Eckerlin, V. Efremenko, S. Egli, R. Eichler, F. Eisele, E. Eisenhandler, E. Elsen, M. Erdmann, A.B. Fahr, L. Favart, A. Fedotov, R. Felst, J. Feltesse, J. Ferencei, F. Ferrarotto, K. Flamm, M. Fleischer, M. Flieser, G. Flügge, A. Fomenko, J. Formánek, J.M. Foster, G. Franke, E. Gabathuler, K. Gabathuler, F. Gaede, J. Garvey, J. Gayler, M. Gebauer, H. Genzel, R. Gerhards, A. Glazov, L. Goerlich, N. Gogitidze, M. Goldberg, D. Goldner, K. Golec-Biernat, B. Gonzalez-Pineiro, I. Gorelov, C. Grab, H. Grässler, T. Greenshaw, R.K. Griffiths, G. Grindhammer, A. Gruber, C. Gruber, T. Hadig, D. Haidt, L. Hajduk, T. Haller, M. Hampel, W.J. Haynes, B. Heinemann, G. Heinzelmann, R.C.W. Henderson, H. Henschel, I. Herynek, M.F. Hess, K. Hewitt, K.H. Hiller, C.D. Hilton, J. Hladký, M. Höppner, D. Hoffmann, T. Holtom, R. Horisberger, V.L. Hudgson, M. Hütte, M. Ibbotson, Ç. İşsever, H. Itterbeck, A. Jacholkowska, C. Jacobsson, M. Jacquet, M. Jaffre, J. Janoth, D.M. Jansen, L. Jönsson, D.P. Johnson, H. Jung, P.I.P. Kalmus, M. Kander, D. Kant, U. Kathage, J. Katzy, H.H. Kaufmann, O. Kaufmann, M. Kausch, S. Kazarian, I.R. Kenyon, S. Kermiche, C. Keuker, C. Kiesling, M. Klein, C. Kleinwort, G. Knies, T. Köhler, J.H. Köhne, H. Kolanoski, S.D. Kolya, V. Korbel, P. Kostka, S.K. Kotelnikov, T. Krämerkämper, M.W. Krasny, H. Krehbiel, D. Krücker, A. Küpper, H. Küster, M. Kuhlen, T. Kurča, J. Kurzhöfer, B. Laforge, M.P.J. Landon, W. Lange, U. Langenegger, A. Lebedev, F. Lehner, V. Lemaitre, S. Levonian, M. Lindstroem, F. Linsel, J. Lipinski, B. List, G. Lobo, J.W. Lomas, G.C. Lopez, V. Lubimov, D. Lüke, L. Lytkin, N. Magnussen, H. Mahlke-Krüger, E. Malinovski, R. Maraček, P. Marage, J. Marks, R. Marshall, J. Martens, G. Martin, R. Martin, H.-U. Martyn, J. Martyniak, T. Mavroidis, S.J. Maxfield, S.J. McMahon, A. Mehta, K. Meier, P. Merkel, F. Metlica, A. Meyer, H. Meyer, J. Meyer, P.-O. Meyer, A. Migliori, S. Mikocki, D. Milstead, J. Moeck, F. Moreau, J.V. Morris, E. Mroczko, D. Müller, T. Walter, K. Müller, P. Murín, V. Nagovizin, R. Nahnhauer, B. Naroska, Th. Naumann, I. Négri, P.R. Newman, D. Newton, H.K. Nguyen, T.C. Nicholls, F. Niebergall, C. Niebuhr, Ch. Niedzballa, H. Niggli, G. Nowak, T. Nunnemann, M. Nyberg-Werther, H. Oberlack, J.E. Olsson, D. Ozerov, P. Palmen, E. Panaro, A. Panitch, C. Pascaud, S. Passaggio, G.D. Patel, H. Pawletta, E. Peppel, E. Perez, J.P. Phillips, A. Pieuchot, D. Pitzl, R. Pöschl, G. Pope, B. Povh, S. Prell, K. Rabbertz, G. Rädel, P. Reimer, H. Rick, S. Riess, E. Rizvi, P. Robmann, R. Roosen, K. Rosenbauer, A. Rostovtsev, F. Rouse, C. Royon, K. Rüter, S. Rusakov, K. Rybicki, D.P.C. Sankey, P. Schacht, S. Schiek, S. Schleif, P. Schleper, W. von Schlippe, D. Schmidt, G. Schmidt, L. Schoeffel, A. Schöning, V. Schröder, E. Schuhmann, B. Schwab, F. Sefkow, A. Semenov, V. Shekelyan, I. Sheviakov, L.N. Shtarkov, G. Siegmon, U. Siewert, Y. Sirois, I.O. Skillicorn, T. Sloan, P. Smirnov, M. Smith, V. Solochenko, Y. Soloviev, A. Specka, J. Spiekermann, S. Spielman, H. Spitzer, F. Squinabol, P. Steffen, R. Steinberg, J. Steinhart, B. Stella, A. Stellberger, J. Stier, J. Stiewe, U. Stöβlein, K. Stolze, U. Straumann, W. Struczinski, J.P. Sutton, S. Tapprogge, M. Taševský, V. Tchernyshov, S. Tchetchelnitski, J. Theissen, G. Thompson, P.D. Thompson, N. Tobien, R. Todenhagen, P. Truöl, G. Tsipolitis, J. Turnau, E. Tzamariudaki, P. Uelkes, A. Usik, S. Valkár, A. Valkárová, C. Valĺee, P. Van Esch, P. Van Mechelen, D. Vandenplas, Y. Vazdik, P. Verrecchia, G. Villet, K. Wacker, A. Wagener, M. Wagener, R. Wallny, B. Waugh, G. Weber, M. Weber, D. Wegener, A. Wegner, T. Wengler, M. Werner, L.R. West, S. Wiesand, T. Wilksen, S. Willard, M. Winde, G.-G. Winter, C. Wittek, M. Wobisch, H. Wollatz, E. Wünsch, J. Žáček, D. Zarbock, Z. Zhang, A. Zhokin, P. Zini, F. Zomer, J. Zsembery, M. zurNedden, Laboratoire Leprince-Ringuet (LLR), Centre National de la Recherche Scientifique (CNRS)-École polytechnique (X)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3), Laboratoire de Physique Nucléaire et de Hautes Énergies (LPNHE), Centre National de la Recherche Scientifique (CNRS)-Université Paris Diderot - Paris 7 (UPD7)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Université Pierre et Marie Curie - Paris 6 (UPMC), Laboratoire de l'Accélérateur Linéaire (LAL), Centre National de la Recherche Scientifique (CNRS)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Université Paris-Sud - Paris 11 (UP11), Centre de Physique des Particules de Marseille (CPPM), Aix Marseille Université (AMU)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Centre National de la Recherche Scientifique (CNRS), H1, Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-École polytechnique (X)-Centre National de la Recherche Scientifique (CNRS), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud - Paris 11 (UP11)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Centre National de la Recherche Scientifique (CNRS), Levy, Jean-Michel, and H1 Collaboration
- Subjects
Physics ,Nuclear and High Energy Physics ,Particle physics ,[PHYS.HEXP] Physics [physics]/High Energy Physics - Experiment [hep-ex] ,Proton ,010308 nuclear & particles physics ,Structure function ,FOS: Physical sciences ,HERA ,01 natural sciences ,High Energy Physics - Experiment ,Nuclear physics ,Momentum ,High Energy Physics - Experiment (hep-ex) ,Cross section (physics) ,0103 physical sciences ,[PHYS.HEXP]Physics [physics]/High Energy Physics - Experiment [hep-ex] ,010306 general physics - Abstract
The results of a measurement of the proton structure function F_2(x,Q^2)and the virtual photon-proton cross section are reported for momentum transfers squared Q^2 between 0.35 GeV^2 and 3.5 GeV^2 and for Bjorken-x values down to 6 10^{-6} using data collected by the HERA experiment H1 in 1995. The data represent an increase in kinematic reach to lower x and Q^2 values of about a factor of 5 compared to previous H1 measurements. Including measurements from fixed target experiments the rise of F_2 with decreasing x is found to be less steep for the lowest Q^2 values measured. Phenomenological models at low Q^2 are compared with the data., Comment: 27 pages, 10 Figures
- Published
- 2016
46. Crystal structure of the human PRMT5:MEP50 complex
- Author
-
Zahid Quyoom Bonday, Bomie Han, Louis Nickolaus Jungheim, Aiping Zhang, Marijane Russell, Kenneth Weichert, Brandon Doyle, Tarun Gheyi, Francis S. Willard, Stephen Antonysamy, J. Michael Sauder, S. Emtage, Zhanna Druzina, Charles Rauch, Robert M. Campbell, Yuewei Qian, and S.R. Wasserman
- Subjects
Models, Molecular ,Regulation of gene expression ,Protein-Arginine N-Methyltransferases ,Multidisciplinary ,Methyltransferase ,Protein Conformation ,Protein arginine methyltransferase 5 ,Signal transducing adaptor protein ,Biological Sciences ,Biology ,Crystallography, X-Ray ,Cell biology ,Histone H4 ,Protein structure ,Biochemistry ,Catalytic Domain ,Humans ,Methylosome protein 50 ,Dimerization ,Adaptor Proteins, Signal Transducing - Abstract
Protein arginine methyltransferases (PRMTs) play important roles in several cellular processes, including signaling, gene regulation, and transport of proteins and nucleic acids, to impact growth, differentiation, proliferation, and development. PRMT5 symmetrically di-methylates the two-terminal ω-guanidino nitrogens of arginine residues on substrate proteins. PRMT5 acts as part of a multimeric complex in concert with a variety of partner proteins that regulate its function and specificity. A core component of these complexes is the WD40 protein MEP50/WDR77/p44, which mediates interactions with binding partners and substrates. We have determined the crystal structure of human PRMT5 in complex with MEP50 (methylosome protein 50), bound to an S-adenosylmethionine analog and a peptide substrate derived from histone H4. The structure of the surprising hetero-octameric complex reveals the close interaction between the seven-bladed β-propeller MEP50 and the N-terminal domain of PRMT5, and delineates the structural elements of substrate recognition.
- Published
- 2012
- Full Text
- View/download PDF
47. Small Molecule Allosteric Modulation of the Glucagon-Like Peptide-1 Receptor Enhances the Insulinotropic Effect of Oxyntomodulin
- Author
-
Francis S. Willard, Denise Wootten, Kyle W. Sloop, Krister Bokvist, James Ficorilli, Patrick M. Sexton, Jorge Alsina-Fernandez, Thomas B. Farb, Arthur Christopoulos, Emilia Elizabeth Savage, Aaron D. Showalter, and Sebastian Gb Furness
- Subjects
Agonist ,medicine.drug_class ,Allosteric regulation ,CHO Cells ,Biology ,Glucagon-Like Peptide-1 Receptor ,Cell Line ,chemistry.chemical_compound ,GTP-Binding Proteins ,Glucagon-Like Peptide 1 ,Cricetinae ,Heterotrimeric G protein ,Cyclic AMP ,Receptors, Glucagon ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Insulin ,Receptor ,Receptor modulator ,Pharmacology ,Drug Synergism ,Small molecule ,Cell biology ,Oxyntomodulin ,HEK293 Cells ,Diabetes Mellitus, Type 2 ,chemistry ,Biochemistry ,Molecular Medicine ,Signal transduction ,Signal Transduction - Abstract
Identifying novel mechanisms to enhance glucagon-like peptide-1 (GLP-1) receptor signaling may enable nascent medicinal chemistry strategies with the aim of developing new orally available therapeutic agents for the treatment of type 2 diabetes mellitus. Therefore, we tested the hypothesis that selectively modulating the low-affinity GLP-1 receptor agonist, oxyntomodulin, would improve the insulin secretory properties of this naturally occurring hormone to provide a rationale for pursuing an unexplored therapeutic approach. Signal transduction and competition binding studies were used to investigate oxyntomodulin activity on the GLP-1 receptor in the presence of the small molecule GLP-1 receptor modulator, 4-(3-benzyloxyphenyl)-2-ethylsulfinyl-6-(trifluoromethyl)pyrimidine (BETP). In vivo, the intravenous glucose tolerance test characterized oxyntomodulin-induced insulin secretion in animals administered the small molecule. BETP increased oxyntomodulin binding affinity for the GLP-1 receptor and enhanced oxyntomodulin-mediated GLP-1 receptor signaling as measured by activation of the α subunit of heterotrimeric G protein and cAMP accumulation. In addition, oxyntomodulin-induced insulin secretion was enhanced in the presence of the compound. BETP was pharmacologically characterized to induce biased signaling by oxyntomodulin. These studies demonstrate that small molecules targeting the GLP-1 receptor can increase binding and receptor activation of the endogenous peptide oxyntomodulin. The biased signaling engendered by BETP suggests that GLP-1 receptor mobilization of cAMP is the critical insulinotropic signaling event. Because of the unique metabolic properties of oxyntomodulin, identifying molecules that enhance its activity should be pursued to assess the efficacy and safety of this novel mechanism.
- Published
- 2012
- Full Text
- View/download PDF
48. Physiology and Emerging Biochemistry of the Glucagon-Like Peptide-1 Receptor
- Author
-
Kyle W. Sloop and Francis S. Willard
- Subjects
lcsh:Internal medicine ,endocrine system ,lcsh:Specialties of internal medicine ,Endocrinology, Diabetes and Metabolism ,Allosteric regulation ,lcsh:Medicine ,Physiology ,Review Article ,Biology ,Crystallography, X-Ray ,Ligands ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,Biochemistry ,Models, Biological ,Glucagon-Like Peptide-1 Receptor ,lcsh:RC581-951 ,Glucagon-Like Peptide 1 ,Cyclic AMP ,Receptors, Glucagon ,medicine ,Homeostasis ,Humans ,Glucose homeostasis ,lcsh:RC31-1245 ,Receptor ,lcsh:RC648-665 ,Venoms ,Liraglutide ,lcsh:R ,digestive, oral, and skin physiology ,General Medicine ,Ligand (biochemistry) ,Small molecule ,Glucose ,Diabetes Mellitus, Type 2 ,Structural biology ,Exenatide ,Signal transduction ,Peptides ,Allosteric Site ,hormones, hormone substitutes, and hormone antagonists ,Protein Binding ,Signal Transduction ,medicine.drug - Abstract
The glucagon-like peptide-1 (GLP-1) receptor is one of the best validated therapeutic targets for the treatment of type 2 diabetes mellitus (T2DM). Over several years, the accumulation of basic, translational, and clinical research helped define the physiologic roles of GLP-1 and its receptor in regulating glucose homeostasis and energy metabolism. These efforts provided much of the foundation for pharmaceutical development of the GLP-1 receptor peptide agonists, exenatide and liraglutide, as novel medicines for patients suffering from T2DM. Now, much attention is focused on better understanding the molecular mechanisms involved in ligand induced signaling of the GLP-1 receptor. For example, advancements in biophysical and structural biology techniques are being applied in attempts to more precisely determine ligand binding and receptor occupancy characteristics at the atomic level. These efforts should better inform three-dimensional modeling of the GLP-1 receptor that will help inspire more rational approaches to identify and optimize small molecule agonists or allosteric modulators targeting the GLP-1 receptor. This article reviews GLP-1 receptor physiology with an emphasis on GLP-1 induced signaling mechanisms in order to highlight new molecular strategies that help determine desired pharmacologic characteristics for guiding development of future nonpeptide GLP-1 receptor activators.
- Published
- 2012
- Full Text
- View/download PDF
49. Open Innovation for Phenotypic Drug Discovery: The PD2 Assay Panel
- Author
-
Francis S. Willard, Jonathan A. Lee, Christopher M. Moxham, Tamika D. Meredith, Robert B. Peery, Karen Leigh Cox, Sarah E. Oliver, Jennifer Oler, Steven A. Heidler, Rachelle J. Sells Galvin, Wendy H. Gough, Alan David Palkowitz, Shaoyou Chu, and Saba Husain
- Subjects
Research groups ,Drug Evaluation, Preclinical ,Neovascularization, Physiologic ,Computational biology ,Biology ,Bioinformatics ,Biochemistry ,Cell Line ,Analytical Chemistry ,Mice ,Apolipoproteins E ,Drug Discovery ,Insulin Secretion ,Animals ,Humans ,Insulin ,Protein Kinase Inhibitors ,Open innovation ,Osteoblasts ,Drug discovery ,Nocodazole ,Statistical validation ,Cell Cycle ,Reproducibility of Results ,Cell Differentiation ,Tubulin Modulators ,Rats ,Wnt Proteins ,Phenotype ,Open source ,Chemical diversity ,Molecular targets ,Molecular Medicine ,Classical pharmacology ,HeLa Cells ,Signal Transduction ,Biotechnology - Abstract
Phenotypic lead generation strategies seek to identify compounds that modulate complex, physiologically relevant systems, an approach that is complementary to traditional, target-directed strategies. Unlike gene-specific assays, phenotypic assays interrogate multiple molecular targets and signaling pathways in a target "agnostic" fashion, which may reveal novel functions for well-studied proteins and discover new pathways of therapeutic value. Significantly, existing compound libraries may not have sufficient chemical diversity to fully leverage a phenotypic strategy. To address this issue, Eli Lilly and Company launched the Phenotypic Drug Discovery Initiative (PD(2)), a model of open innovation whereby external research groups can submit compounds for testing in a panel of Lilly phenotypic assays. This communication describes the statistical validation, operations, and initial screening results from the first PD(2) assay panel. Analysis of PD(2) submissions indicates that chemical diversity from open source collaborations complements internal sources. Screening results for the first 4691 compounds submitted to PD(2) have confirmed hit rates from 1.6% to 10%, with the majority of active compounds exhibiting acceptable potency and selectivity. Phenotypic lead generation strategies, in conjunction with novel chemical diversity obtained via open-source initiatives such as PD(2), may provide a means to identify compounds that modulate biology by novel mechanisms and expand the innovation potential of drug discovery.
- Published
- 2011
- Full Text
- View/download PDF
50. Abstract No. 594 Nusinersen for spinal muscular atrophy: the era of interventional radiology molecular therapy is here
- Author
-
S. Willard, David Aria, Richard B. Towbin, Robin D. Kaye, and C. Schaefer
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine ,Radiology, Nuclear Medicine and imaging ,Nusinersen ,Interventional radiology ,Spinal muscular atrophy ,Radiology ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business ,Molecular therapy - Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.