48 results on '"Rosenberg, Oren S"'
Search Results
2. Structure and dynamics of the essential endogenous mycobacterial polyketide synthase Pks13
- Author
-
Kim, Sun Kyung, Dickinson, Miles Sasha, Finer-Moore, Janet, Guan, Ziqiang, Kaake, Robyn M., Echeverria, Ignacia, Chen, Jen, Pulido, Ernst H., Sali, Andrej, Krogan, Nevan J., Rosenberg, Oren S., and Stroud, Robert M.
- Published
- 2023
- Full Text
- View/download PDF
3. Imaging joint infections using D-methyl-11C-methionine PET/MRI: initial experience in humans
- Author
-
Polvoy, Ilona, Seo, Youngho, Parker, Matthew, Stewart, Megan, Siddiqua, Khadija, Manacsa, Harrison S., Ravanfar, Vahid, Blecha, Joseph, Hope, Thomas A., Vanbrocklin, Henry, Flavell, Robert R., Barry, Jeffrey, Hansen, Erik, Villanueva-Meyer, Javier E., Engel, Joanne, Rosenberg, Oren S., Wilson, David M., and Ohliger, Michael A.
- Published
- 2022
- Full Text
- View/download PDF
4. Engineered ACE2 receptor traps potently neutralize SARS-CoV-2
- Author
-
Glasgow, Anum, Glasgow, Jeff, Limonta, Daniel, Solomon, Paige, Lui, Irene, Zhang, Yang, Nix, Matthew A., Rettko, Nicholas J., Zha, Shoshana, Yamin, Rachel, Kao, Kevin, Rosenberg, Oren S., Ravetch, Jeffrey V., Wiita, Arun P., Leung, Kevin K., Lim, Shion A., Zhou, Xin X., Hobman, Tom C., Kortemme, Tanja, and Wells, James A.
- Published
- 2020
5. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing
- Author
-
Gordon, David E., Jang, Gwendolyn M., Bouhaddou, Mehdi, Xu, Jiewei, Obernier, Kirsten, White, Kris M., O’Meara, Matthew J., Rezelj, Veronica V., Guo, Jeffrey Z., Swaney, Danielle L., Tummino, Tia A., Hüttenhain, Ruth, Kaake, Robyn M., Richards, Alicia L., Tutuncuoglu, Beril, Foussard, Helene, Batra, Jyoti, Haas, Kelsey, Modak, Maya, Kim, Minkyu, Haas, Paige, Polacco, Benjamin J., Braberg, Hannes, Fabius, Jacqueline M., Eckhardt, Manon, Soucheray, Margaret, Bennett, Melanie J., Cakir, Merve, McGregor, Michael J., Li, Qiongyu, Meyer, Bjoern, Roesch, Ferdinand, Vallet, Thomas, Mac Kain, Alice, Miorin, Lisa, Moreno, Elena, Naing, Zun Zar Chi, Zhou, Yuan, Peng, Shiming, Shi, Ying, Zhang, Ziyang, Shen, Wenqi, Kirby, Ilsa T., Melnyk, James E., Chorba, John S., Lou, Kevin, Dai, Shizhong A., Barrio-Hernandez, Inigo, Memon, Danish, Hernandez-Armenta, Claudia, Lyu, Jiankun, Mathy, Christopher J. P., Perica, Tina, Pilla, Kala Bharath, Ganesan, Sai J., Saltzberg, Daniel J., Rakesh, Ramachandran, Liu, Xi, Rosenthal, Sara B., Calviello, Lorenzo, Venkataramanan, Srivats, Liboy-Lugo, Jose, Lin, Yizhu, Huang, Xi-Ping, Liu, YongFeng, Wankowicz, Stephanie A., Bohn, Markus, Safari, Maliheh, Ugur, Fatima S., Koh, Cassandra, Savar, Nastaran Sadat, Tran, Quang Dinh, Shengjuler, Djoshkun, Fletcher, Sabrina J., O’Neal, Michael C., Cai, Yiming, Chang, Jason C. J., Broadhurst, David J., Klippsten, Saker, Sharp, Phillip P., Wenzell, Nicole A., Kuzuoglu-Ozturk, Duygu, Wang, Hao-Yuan, Trenker, Raphael, Young, Janet M., Cavero, Devin A., Hiatt, Joseph, Roth, Theodore L., Rathore, Ujjwal, Subramanian, Advait, Noack, Julia, Hubert, Mathieu, Stroud, Robert M., Frankel, Alan D., Rosenberg, Oren S., Verba, Kliment A., Agard, David A., Ott, Melanie, Emerman, Michael, Jura, Natalia, von Zastrow, Mark, Verdin, Eric, Ashworth, Alan, Schwartz, Olivier, d’Enfert, Christophe, Mukherjee, Shaeri, Jacobson, Matt, Malik, Harmit S., Fujimori, Danica G., Ideker, Trey, Craik, Charles S., Floor, Stephen N., Fraser, James S., Gross, John D., Sali, Andrej, Roth, Bryan L., Ruggero, Davide, Taunton, Jack, Kortemme, Tanja, Beltrao, Pedro, Vignuzzi, Marco, García-Sastre, Adolfo, Shokat, Kevan M., Shoichet, Brian K., and Krogan, Nevan J.
- Published
- 2020
- Full Text
- View/download PDF
6. Bioorthogonal Radiolabeling of Azide-Modified Bacteria Using [18F]FB-sulfo-DBCO.
- Author
-
Alanizi, Aryn A., Sorlin, Alexandre M., Parker, Matthew F. L., López-Álvarez, Marina, Qin, Hecong, Lee, Sang Hee, Blecha, Joseph, Rosenberg, Oren S., Engel, Joanne, Ohliger, Michael A., Flavell, Robert R., and Wilson, David M.
- Published
- 2024
- Full Text
- View/download PDF
7. Enabling genetic analysis of diverse bacteria with Mobile-CRISPRi
- Author
-
Peters, Jason M., Koo, Byoung-Mo, Patino, Ramiro, Heussler, Gary E., Hearne, Cameron C., Qu, Jiuxin, Inclan, Yuki F., Hawkins, John S., Lu, Candy H. S., Silvis, Melanie R., Harden, M. Michael, Osadnik, Hendrik, Peters, Joseph E., Engel, Joanne N., Dutton, Rachel J., Grossman, Alan D., Gross, Carol A., and Rosenberg, Oren S.
- Published
- 2019
- Full Text
- View/download PDF
8. Controlling CRISPR-Cas9 with ligand-activated and ligand-deactivated sgRNAs
- Author
-
Kundert, Kale, Lucas, James E., Watters, Kyle E., Fellmann, Christof, Ng, Andrew H., Heineike, Benjamin M., Fitzsimmons, Christina M., Oakes, Benjamin L., Qu, Jiuxin, Prasad, Neha, Rosenberg, Oren S., Savage, David F., El-Samad, Hana, Doudna, Jennifer A., and Kortemme, Tanja
- Published
- 2019
- Full Text
- View/download PDF
9. Evaluating the Performance of Pathogen-Targeted Positron Emission Tomography Radiotracers in a Rat Model of Vertebral Discitis-Osteomyelitis.
- Author
-
Parker, Matthew F L, López-Álvarez, Marina, Alanizi, Aryn A, Luu, Justin M, Polvoy, Ilona, Sorlin, Alexandre M, Qin, Hecong, Lee, Sanghee, Rabbitt, Sarah J, Pichardo-González, Priamo A, Ordonez, Alvaro A, Blecha, Joseph, Rosenberg, Oren S, Flavell, Robert R, Engel, Joanne, Jain, Sanjay K, Ohliger, Michael A, and Wilson, David M
- Subjects
POSITRON emission tomography ,RADIOACTIVE tracers ,ANIMAL disease models ,MAGNETIC resonance imaging ,COMPUTED tomography - Abstract
Background Vertebral discitis-osteomyelitis (VDO) is a devastating infection of the spine that is challenging to distinguish from noninfectious mimics using computed tomography and magnetic resonance imaging. We and others have developed novel metabolism-targeted positron emission tomography (PET) radiotracers for detecting living Staphylococcus aureus and other bacteria in vivo, but their head-to-head performance in a well-validated VDO animal model has not been reported. Methods We compared the performance of several PET radiotracers in a rat model of VDO. [
11 C]PABA and [18 F]FDS were assessed for their ability to distinguish S aureus , the most common non-tuberculous pathogen VDO, from Escherichia coli. Results In the rat S aureus VDO model, [11 C]PABA could detect as few as 103 bacteria and exhibited the highest signal-to-background ratio, with a 20-fold increased signal in VDO compared to uninfected tissues. In a proof-of-concept experiment, detection of bacterial infection and discrimination between S aureus and E coli was possible using a combination of [11 C]PABA and [18 F]FDS. Conclusions Our work reveals that several bacteria-targeted PET radiotracers had sufficient signal to background in a rat model of S aureus VDO to be potentially clinically useful. [11 C]PABA was the most promising tracer investigated and warrants further investigation in human VDO. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
10. Structure of a new DNA-binding domain which regulates pathogenesis in a wide variety of fungi
- Author
-
Lohse, Matthew B., Rosenberg, Oren S., Cox, Jeffery S., Stroud, Robert M., Finer-Moore, Janet S., and Johnson, Alexander D.
- Published
- 2014
11. Chemoenzymatic Syntheses of Fluorine-18-Labeled Disaccharides from [18F] FDG Yield Potent Sensors of Living Bacteria In Vivo.
- Author
-
Sorlin, Alexandre M., López-Álvarez, Marina, Rabbitt, Sarah J., Alanizi, Aryn A., Shuere, Rebecca, Bobba, Kondapa Naidu, Blecha, Joseph, Sakhamuri, Sasank, Evans, Michael J., Bayles, Kenneth W., Flavell, Robert R., Rosenberg, Oren S., Sriram, Renuka, Desmet, Tom, Nidetzky, Bernd, Engel, Joanne, Ohliger, Michael A., Fraser, James S., and Wilson, David M.
- Published
- 2023
- Full Text
- View/download PDF
12. EspR, a key regulator of Mycobacterium tuberculosis virulence, adopts a unique dimeric structure among helix-turn-helix proteins
- Author
-
Rosenberg, Oren S., Dovey, Cole, Tempesta, Michael, Robbins, Rebecca A., Finer-Moore, Janet S., Stroud, Robert M., and Cox, Jeffery S.
- Published
- 2011
13. Treatment History and Treatment Dose Are Important Determinants of Sulfadoxine-Pyrimethamine Efficacy in Children with Uncomplicated Malaria in Western Kenya
- Author
-
Terlouw, Dianne J., Courval, Jeanne M., Kolczak, Margarette S., Rosenberg, Oren S., Oloo, Aggrey J., Kager, Piet A., Lal, Altaf A., Nahlen, Bernard L., and ter Kuile, Feiko O.
- Published
- 2003
14. Imaging joint infections using D-methyl-11C-methionine PET/MRI: initial experience in humans.
- Author
-
Polvoy, Ilona, Seo, Youngho, Parker, Matthew, Stewart, Megan, Siddiqua, Khadija, Manacsa, Harrison S., Ravanfar, Vahid, Blecha, Joseph, Hope, Thomas A., Vanbrocklin, Henry, Flavell, Robert R., Barry, Jeffrey, Hansen, Erik, Villanueva-Meyer, Javier E., Engel, Joanne, Rosenberg, Oren S., Wilson, David M., and Ohliger, Michael A.
- Subjects
JOINT infections ,ARTIFICIAL joints ,BACTERIAL cell walls ,ARTHROPLASTY ,RADIATION dosimetry ,MAGNETIC resonance imaging - Abstract
Purpose: Non-invasive imaging is a key clinical tool for detection and treatment monitoring of infections. Existing clinical imaging techniques are frequently unable to distinguish infection from tumors or sterile inflammation. This challenge is well-illustrated by prosthetic joint infections that often complicate joint replacements. D-methyl-
11 C-methionine (D-11 C-Met) is a new bacteria-specific PET radiotracer, based on an amino acid D-enantiomer, that is rapidly incorporated into the bacterial cell wall. In this manuscript, we describe the biodistribution, radiation dosimetry, and initial human experience using D-11 C-Met in patients with suspected prosthetic joint infections. Methods: 614.5 ± 100.2 MBq of D-11 C-Met was synthesized using an automated in-loop radiosynthesis method and administered to six healthy volunteers and five patients with suspected prosthetic joint infection, who were studied by PET/MRI. Time-activity curves were used to calculate residence times for each source organ. Absorbed doses to each organ and body effective doses were calculated using OLINDA/EXM 1.1 with both ICRP 60 and ICRP 103 tissue weighting factors. SUVmax and SUVpeak were calculated for volumes of interest (VOIs) in joints with suspected infection, the unaffected contralateral joint, blood pool, and soft tissue background. A two-tissue compartment model was used for kinetic modeling. Results: D-11 C-Met was well tolerated in all subjects. The tracer showed clearance from both urinary (rapid) and hepatobiliary (slow) pathways as well as low effective doses. Moreover, minimal background was observed in both organs with resident micro-flora and target organs, such as the spine and musculoskeletal system. Additionally, D-11 C-Met showed increased focal uptake in areas of suspected infection, demonstrated by a significantly higher SUVmax and SUVpeak calculated from VOIs of joints with suspected infections compared to the contralateral joints, blood pool, and background (P < 0.01). Furthermore, higher distribution volume and binding potential were observed in suspected infections compared to the unaffected joints. Conclusion: D-11 C-Met has a favorable radiation profile, minimal background uptake, and fast urinary extraction. Furthermore, D-11 C-Met showed increased uptake in areas of suspected infection, making this a promising approach. Validation in larger clinical trials with a rigorous gold standard is still required. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
15. Comparative host-coronavirus protein interaction networks reveal pan-viral disease mechanisms
- Author
-
Gordon, David E., Hiatt, Joseph, Bouhaddou, Mehdi, Rezelj, Veronica V., Ulferts, Svenja, Braberg, Hannes, Jureka, Alexander S., Obernier, Kirsten, Guo, Jeffrey Z., Batra, Jyoti, Kaake, Robyn M., Weckstein, Andrew R., Owens, Tristan W., Gupta, Meghna, Pourmal, Sergei, Titus, Erron W., Cakir, Merve, Soucheray, Margaret, McGregor, Michael, Cakir, Zeynep, Jang, Gwendolyn, O’Meara, Matthew J., Tummino, Tia A., Zhang, Ziyang, Foussard, Helene, Rojc, Ajda, Zhou, Yuan, Kuchenov, Dmitry, Hüttenhain, Ruth, Xu, Jiewei, Eckhardt, Manon, Swaney, Danielle L., Fabius, Jacqueline M., Ummadi, Manisha, Tutuncuoglu, Beril, Rathore, Ujjwal, Modak, Maya, Haas, Paige, Haas, Kelsey M., Naing, Zun Zar Chi, Pulido, Ernst H., Shi, Ying, Barrio-Hernandez, Inigo, Memon, Danish, Petsalaki, Eirini, Dunham, Alistair, Marrero, Miguel Correa, Burke, David, Koh, Cassandra, Vallet, Thomas, Silvas, Jesus A., Azumaya, Caleigh M., Billesbølle, Christian, Brilot, Axel F., Campbell, Melody G., Diallo, Amy, Dickinson, Miles Sasha, Diwanji, Devan, Herrera, Nadia, Hoppe, Nick, Kratochvil, Huong T., Liu, Yanxin, Merz, Gregory E., Moritz, Michelle, Nguyen, Henry C., Nowotny, Carlos, Puchades, Cristina, Rizo, Alexandrea N., Schulze-Gahmen, Ursula, Smith, Amber M., Sun, Ming, Young, Iris D., Zhao, Jianhua, Asarnow, Daniel, Biel, Justin, Bowen, Alisa, Braxton, Julian R., Chen, Jen, Chio, Cynthia M., Chio, Un Seng, Deshpande, Ishan, Doan, Loan, Faust, Bryan, Flores, Sebastian, Jin, Mingliang, Kim, Kate, Lam, Victor L., Li, Fei, Li, Junrui, Li, Yen-Li, Li, Yang, Liu, Xi, Lo, Megan, Lopez, Kyle E., Melo, Arthur A., Moss, Frank R., Nguyen, Phuong, Paulino, Joana, Pawar, Komal Ishwar, Peters, Jessica K., Pospiech, Thomas H., Safari, Maliheh, Sangwan, Smriti, Schaefer, Kaitlin, Thomas, Paul V., Thwin, Aye C., Trenker, Raphael, Tse, Eric, Tsui, Tsz Kin Martin, Wang, Feng, Whitis, Natalie, Yu, Zanlin, Zhang, Kaihua, Zhang, Yang, Zhou, Fengbo, Saltzberg, Daniel, Hodder, Anthony J., Shun-Shion, Amber S., Williams, Daniel M., White, Kris M., Rosales, Romel, Kehrer, Thomas, Miorin, Lisa, Moreno, Elena, Patel, Arvind H., Rihn, Suzannah, Khalid, Mir M., Vallejo-Gracia, Albert, Fozouni, Parinaz, Simoneau, Camille R., Roth, Theodore L., Wu, David, Karim, Mohd Anisul, Ghoussaini, Maya, Dunham, Ian, Berardi, Francesco, Weigang, Sebastian, Chazal, Maxime, Park, Jisoo, Logue, James, McGrath, Marisa, Weston, Stuart, Haupt, Robert, Hastie, C. James, Elliott, Matthew, Brown, Fiona, Burness, Kerry A., Reid, Elaine, Dorward, Mark, Johnson, Clare, Wilkinson, Stuart G., Geyer, Anna, Giesel, Daniel M., Baillie, Carla, Raggett, Samantha, Leech, Hannah, Toth, Rachel, Goodman, Nicola, Keough, Kathleen C., Lind, Abigail L., Klesh, Reyna J., Hemphill, Kafi R., Carlson-Stevermer, Jared, Oki, Jennifer, Holden, Kevin, Maures, Travis, Pollard, Katherine S., Sali, Andrej, Agard, David A., Cheng, Yifan, Fraser, James S., Frost, Adam, Jura, Natalia, Kortemme, Tanja, Manglik, Aashish, Southworth, Daniel R., Stroud, Robert M., Alessi, Dario R., Davies, Paul, Frieman, Matthew B., Ideker, Trey, Abate, Carmen, Jouvenet, Nolwenn, Kochs, Georg, Shoichet, Brian, Ott, Melanie, Palmarini, Massimo, Shokat, Kevan M., García-Sastre, Adolfo, Rassen, Jeremy A., Grosse, Robert, Rosenberg, Oren S., Verba, Kliment A., Basler, Christopher F., Vignuzzi, Marco, Peden, Andrew A., Beltrao, Pedro, Krogan, Nevan J., Trinidad, Donovan, Damas, Joana, Hughes, Graham M., Painter, Corrie A., Persky, Nicole S., Corbo, Marco, Kirilenko, Bodgan, Hiller, Michael, Koepfli, Klaus-Peter, Kaplow, Irene, Wirthlin, Morgan, Pfenning, Andreas R., Zhao, Huabin, Genereux, Diane P., Swofford, Ross, Lind, Abigail, Ryderq, Oliver A., Nweeia, Martin T., Meadows, Jennifer, Dong, Michael, Wallerman, Ola, Marinescu, Vikki, Lindblad-Toh, Kerstin, Ray, David A., Power, Sarahjane, Teeling, Emma C., Chauhan, Gaurav, Li, Shirley Xue, Karlsson, Elinor K., Lewin, Harris A., Centre National de la Recherche Scientifique (CNRS), Quantitative Biosciences Institute [UC San Francisco, USA] (QBI), University of California [San Francisco] (UC San Francisco), University of California (UC)-University of California (UC), Populations virales et Pathogenèse - Viral Populations and Pathogenesis, and Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Infectious Medicine ,Middle East respiratory syndrome coronavirus ,Viral protein ,viruses ,[SDV]Life Sciences [q-bio] ,Infektionsmedicin ,medicine.disease_cause ,Conserved sequence ,Microbiology in the medical area ,03 medical and health sciences ,Mitochondrial membrane transport protein ,0302 clinical medicine ,Protein structure ,Pandemic ,medicine ,Mikrobiologi inom det medicinska området ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Coronavirus ,0303 health sciences ,Multidisciplinary ,biology ,virus diseases ,Virology ,3. Good health ,biology.protein ,Viral disease ,030217 neurology & neurosurgery - Abstract
INTRODUCTION The emergence of three lethal coronaviruses in Jennifer Meadows, Michael Dong, Ola Wallerman, Vikki Marinescu & Kerstin Lindblad-Toh ingår i gruppen Zoonomia Consortium
- Published
- 2020
- Full Text
- View/download PDF
16. Structure of the Autoinhibited Kinase Domain of CaMKII and SAXS Analysis of the Holoenzyme
- Author
-
Rosenberg, Oren S., Deindl, Sebastian, Sung, Rou-Jia, Nairn, Angus C., and Kuriyan, John
- Subjects
Threonine -- Analysis ,Peptides -- Analysis ,Protein kinases -- Analysis ,Calmodulin -- Analysis ,Enzymes -- Analysis ,Crystals -- Structure ,Crystals -- Analysis ,Biological sciences - Abstract
To link to full-text access for this article, visit this link: http://dx.doi.org/10.1016/j.cell.2005.10.029 Byline: Oren S. Rosenberg (1)(2)(3)(5), Sebastian Deindl (1)(2)(3), Rou-Jia Sung (1)(2)(3), Angus C. Nairn (6), John Kuriyan (1)(2)(3)(4) Abstract: Ca.sup.2+/calmodulin-dependent protein kinase-II (CaMKII) is unique among protein kinases for its dodecameric assembly and its complex response to Ca.sup.2+. The crystal structure of the autoinhibited kinase domain of CaMKII, determined at 1.8 A resolution, reveals an unexpected dimeric organization in which the calmodulin-responsive regulatory segments form a coiled-coil strut that blocks peptide and ATP binding to the otherwise intrinsically active kinase domains. A threonine residue in the regulatory segment, which when phosphorylated renders CaMKII calmodulin independent, is held apart from the catalytic sites by the organization of the dimer. This ensures a strict Ca.sup.2+ dependence for initial activation. The structure of the kinase dimer, when combined with small-angle X-ray scattering data for the holoenzyme, suggests that inactive CaMKII forms tightly packed autoinhibited assemblies that convert upon activation into clusters of loosely tethered and independent kinase domains. Author Affiliation: (1) Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA (2) Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA (3) Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA (4) Physical Biosciences Division, Lawrence Berkeley National Lab, Berkeley, CA 94720, USA (5) Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06508, USA (6) Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508, USA Article History: Received 9 August 2005; Revised 2 October 2005; Accepted 19 October 2005 Article Note: (miscellaneous) Published: December 1, 2005
- Published
- 2005
17. Immune Reconstitution Reactions in Human Immunodeficiency Virus–Negative Patients: Report of a Case and Review of the Literature
- Author
-
Scharschmidt, Tiffany C., Amerson, Erin H., Rosenberg, Oren S., Jacobs, Richard A., McCalmont, Timothy H., and Shinkai, Kanade
- Published
- 2013
- Full Text
- View/download PDF
18. Oligomerization states of the association domain and the holoenyzme of Ca2+/CaM kinase II
- Author
-
Rosenberg, Oren S., Deindl, Sebastian, Comolli, Luis R., Hoelz, André, Downing, Kenneth H., Nairn, Angus C., and Kuriyan, John
- Published
- 2006
19. Nuclear imaging of bacterial infection- state of the art and future directions.
- Author
-
Polvoy, Ilona, Flavell, Robert R., Rosenberg, Oren S., Ohliger, Michael A., and Wilson, David M.
- Published
- 2020
- Full Text
- View/download PDF
20. Small Molecule Sensors Targeting the Bacterial Cell Wall.
- Author
-
Parker, Matthew F. L., Flavell, Robert R., Luu, Justin M., Rosenberg, Oren S., Ohliger, Michael A., and Wilson, David M.
- Published
- 2020
- Full Text
- View/download PDF
21. Arabinofuranose‐derived positron‐emission tomography radiotracers for detection of pathogenic microorganisms.
- Author
-
Kalita, Mausam, Parker, Matthew F.L., Luu, Justin M., Stewart, Megan N., Blecha, Joseph E., VanBrocklin, Henry F., Evans, Michael J., Flavell, Robert R., Rosenberg, Oren S., Ohliger, Michael A., and Wilson, David M.
- Subjects
PATHOGENIC microorganisms ,RADIOACTIVE tracers ,DETECTION of microorganisms ,POSITRON emission tomography ,RADIOCHEMICAL purification - Abstract
PURPOSE: Detection of bacteria‐specific metabolism via positron emission tomography (PET) is an emerging strategy to image human pathogens, with dramatic implications for clinical practice. In silico and in vitro screening tools have recently been applied to this problem, with several monosaccharides including l‐arabinose showing rapid accumulation in Escherichia coli and other organisms. Our goal for this study was to evaluate several synthetically viable arabinofuranose‐derived 18F analogs for their incorporation into pathogenic bacteria. PROCEDURES: We synthesized four radiolabeled arabinofuranose‐derived sugars: 2‐deoxy‐2‐[18F]fluoro‐arabinofuranoses (d‐2‐18F‐AF and l‐2‐18F‐AF) and 5‐deoxy‐5‐[18F]fluoro‐arabinofuranoses (d‐5‐18F‐AF and l‐5‐18F‐AF). The arabinofuranoses were synthesized from 18F‐ via triflated, peracetylated precursors analogous to the most common radiosynthesis of 2‐deoxy‐2‐[18F]fluoro‐d‐glucose ([18F]FDG). These radiotracers were screened for their uptake into E. coli and Staphylococcus aureus. Subsequently, the sensitivity of d‐2‐18F‐AF and l‐2‐18F‐AF to key human pathogens was investigated in vitro. RESULTS: All 18F radiotracer targets were synthesized in high radiochemical purity. In the screening study, d‐2‐18F‐AF and l‐2‐18F‐AF showed greater accumulation in E. coli than in S. aureus. When evaluated in a panel of pathologic microorganisms, both d‐2‐18F‐AF and l‐2‐18F‐AF demonstrated sensitivity to most gram‐positive and gram‐negative bacteria. CONCLUSIONS: Arabinofuranose‐derived 18F PET radiotracers can be synthesized with high radiochemical purity. Our study showed absence of bacterial accumulation for 5‐substitued analogs, a finding that may have mechanistic implications for related tracers. Both d‐2‐18F‐AF and l‐2‐18F‐AF showed sensitivity to most gram‐negative and gram‐positive organisms. Future in vivo studies will evaluate the diagnostic accuracy of these radiotracers in animal models of infection. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. Oligomerization states of the association domain and the holoenyzme of Ca2+/CaM kinase II.
- Author
-
Rosenberg, Oren S., Deindl, Sebastian, Comolli, Luis R., Hoelz, André, Downing, Kenneth H., Nairn, Angus C., and Kuriyan, John
- Subjects
- *
PROTEIN kinases , *PEPTIDE hormones , *GROWTH factors , *CAENORHABDITIS , *CAENORHABDITIS elegans , *ELECTRON microscopy , *FUNGUS-bacterium relationships , *PROTEOLYSIS - Abstract
Ca2+/calmodulin activated protein kinase II (CaMKII) is an oligomeric protein kinase with a unique holoenyzme architecture. The subunits of CaMKII are bound together into the holoenzyme by the association domain, a C-terminal region of ≈ 140 residues in the CaMKII polypeptide. Single particle analyses of electron micrographs have suggested previously that the holoenyzme forms a dodecamer that contains two stacked 6-fold symmetric rings. In contrast, a recent crystal structure of the isolated association domain of mouse CaMKIIα has revealed a tetradecameric assembly with two stacked 7-fold symmetric rings. In this study, we have determined the crystal structure of the Caenorhabditis elegans CaMKII association domain and it too forms a tetradecamer. We also show by electron microscopy that in its fully assembled form the CaMKII holoenzyme is a dodecamer but without the kinase domains, either from expression of the isolated association domain in bacteria or following their removal by proteolysis, the association domains form a tetradecamer. We speculate that the holoenzyme is held in its 6-fold symmetric state by the interactions of the N-terminal ≈ 1–335 residues and that the removal of this region allows the association domain to convert into a more stable 7-fold symmetric form. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
23. A Mechanism for Tunable Autoinhibition in the Structure of a Human Ca2+/Calmodulin- Dependent Kinase II Holoenzyme
- Author
-
Chao, Luke H., Stratton, Margaret M., Lee, Il-Hyung, Rosenberg, Oren S., Levitz, Joshua, Mandell, Daniel J., Kortemme, Tanja, Groves, Jay T., Schulman, Howard, and Kuriyan, John
- Subjects
- *
CALCIUM-binding proteins , *PROTEIN kinases , *MOLECULAR structure , *ENZYME inhibitors , *CALMODULIN , *CARRIER proteins - Abstract
Summary: Calcium/calmodulin-dependent kinase II (CaMKII) forms a highly conserved dodecameric assembly that is sensitive to the frequency of calcium pulse trains. Neither the structure of the dodecameric assembly nor how it regulates CaMKII are known. We present the crystal structure of an autoinhibited full-length human CaMKII holoenzyme, revealing an unexpected compact arrangement of kinase domains docked against a central hub, with the calmodulin-binding sites completely inaccessible. We show that this compact docking is important for the autoinhibition of the kinase domains and for setting the calcium response of the holoenzyme. Comparison of CaMKII isoforms, which differ in the length of the linker between the kinase domain and the hub, demonstrates that these interactions can be strengthened or weakened by changes in linker length. This equilibrium between autoinhibited states provides a simple mechanism for tuning the calcium response without changes in either the hub or the kinase domains. PaperFlick: Display Omitted [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
24. Modulating Pathogenesis with Mobile-CRISPRi.
- Author
-
Jiuxin Qu, Prasad, Neha K., Yu, Michelle A., Shuyan Chen, Lyden, Amy, Herrera, Nadia, Silvis, Melanie R., Crawford, Emily, Looney, Mark R., Peters, Jason M., and Rosenberg, Oren S.
- Abstract
Conditionally essential (CE) genes are required by pathogenic bacteria to establish and maintain infections. CE genes encode virulence factors, such as secretion systems and effector proteins, as well as biosynthetic enzymes that produce metabolites not found in the host environment. Due to their outsized importance in pathogenesis, CE gene products are attractive targets for the next generation of antimicrobials. However, the precise manipulation of CE gene expression in the context of infection is technically challenging, limiting our ability to understand the roles of CE genes in pathogenesis and accordingly design effective inhibitors. We previously developed a suite of CRISPR interference-based gene knockdown tools that are transferred by conjugation and stably integrate into bacterial genomes that we call Mobile-CRISPRi. Here, we show the efficacy of Mobile-CRISPRi in controlling CE gene expression in an animal infection model. We optimize Mobile-CRISPRi in Pseudomonas aeruginosa for use in a murine model of pneumonia by tuning the expression of CRISPRi components to avoid nonspecific toxicity. As a proof of principle, we demonstrate that knock down of a CE gene encoding the type III secretion system (T3SS) activator ExsA blocks effector protein secretion in culture and attenuates virulence in mice. We anticipate that Mobile-CRISPRi will be a valuable tool to probe the function of CE genes across many bacterial species and pathogenesis models. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
25. Deep mutational scanning of EccD 3 reveals the molecular basis of its essentiality in the mycobacterium ESX secretion system.
- Author
-
Trinidad DD, Macdonald CB, Rosenberg OS, Fraser JS, and Coyote-Maestas W
- Abstract
Tuberculosis remains the deadliest infectious disease in the world and requires novel therapeutic targets. The ESX-3 secretion system, which is essential for iron and zinc homeostasis and thus M. tuberculosis survival, is a promising target. In this study, we perform a deep mutational scan on the ESX-3 core protein EccD
3 in the model organism M. smegmatis . We systematically investigated the functional roles of 145 residues across the soluble ubiquitin-like domain, the conformationally distinct flexible linker, and selected transmembrane helices of EccD3 . Our data combined with structural comparisons to ESX-5 complexes support a model where EccD3 stabilizes the complex, with the hinge motif within the linker being particularly sensitive to disruption. Our study is the first deep mutational scan in mycobacteria, which could help guide drug development toward novel treatment of tuberculosis. This study underscores the importance of context-specific mutational analyses for discovering essential protein interactions within mycobacterial systems., Competing Interests: Conflicts of interest: OSR is an employee and equity holder in GSK plc. JSF is a consultant for, has equity in, and receives research support from Relay Therapeutics.- Published
- 2024
- Full Text
- View/download PDF
26. Bioorthogonal Radiolabeling of Azide-Modified Bacteria Using [ 18 F]FB-sulfo-DBCO.
- Author
-
Alanizi AA, Sorlin AM, Parker MFL, López-Álvarez M, Qin H, Lee SH, Blecha J, Rosenberg OS, Engel J, Ohliger MA, Flavell RR, and Wilson DM
- Subjects
- Humans, Animals, Mice, Tissue Distribution, Positron-Emission Tomography, Bacteria, Amino Acids, Alanine, Fluorine Radioisotopes chemistry, Azides chemistry, Peptidoglycan
- Abstract
Purpose: This study was motivated by the need for better positron emission tomography (PET)-compatible tools to image bacterial infection. Our previous efforts have targeted bacteria-specific metabolism via assimilation of carbon-11 labeled d-amino acids into the bacterial cell wall. Since the chemical determinants of this incorporation are not fully understood, we sought a high-throughput method to label d-amino acid derived structures with fluorine-18. Our strategy employed a chemical biology approach, whereby an azide (-N
3 ) bearing d-amino acid is incorporated into peptidoglycan muropeptides, with subsequent "click" cycloaddition with an18 F-labeled strained cyclooctyne partner. Procedures: A water-soluble,18 F-labeled and dibenzocyclooctyne (DBCO)-derived radiotracer ([18 F]FB-sulfo-DBCO) was synthesized. This tracer was incubated with pathogenic bacteria treated with azide-bearing d-amino acids, and incorporated18 F was determined via gamma counting. In vitro uptake in bacteria previously treated with azide-modified d-amino acids was compared to that in cultures treated with amino acid controls. The biodistribution of [18 F]FB-sulfo-DBCO was studied in a cohort of healthy mice with implications for future in vivo imaging. Results: The new strain-promoted azide-alkyne cycloaddition (SPAAC) radiotracer [18 F]FB-sulfo-DBCO was synthesized with high radiochemical yield and purity via N -succinimidyl 4-[18 F]fluorobenzoate ([18 F]SFB). Accumulation of [18 F]FB-sulfo-DBCO was significantly higher in several bacteria treated with azide-modified d-amino acids than in controls; for example, we observed 7 times greater [18 F]FB-sulfo-DBCO ligation in Staphylococcus aureus cultures incubated with 3-azido-d-alanine versus those incubated with d-alanine. Conclusions: The SPAAC radiotracer [18 F]FB-sulfo-DBCO was validated in vitro via metabolic labeling of azide-bearing peptidoglycan muropeptides. d-Amino acid-derived PET radiotracers may be more efficiently screened via [18 F]FB-sulfo-DBCO modification.- Published
- 2024
- Full Text
- View/download PDF
27. Investigating Pseudomonas aeruginosa Gene Function During Pathogenesis Using Mobile-CRISPRi.
- Author
-
Yu MA, Banta AB, Ward RD, Prasad NK, Kwon MS, Rosenberg OS, and Peters JM
- Subjects
- Animals, Mice, Clustered Regularly Interspaced Short Palindromic Repeats, Gene Silencing, Phenotype, Pseudomonas aeruginosa genetics, CRISPR-Cas Systems
- Abstract
CRISPR interference (CRISPRi) is a robust gene silencing technique that is ideal for targeting essential and conditionally essential (CE) genes. CRISPRi is especially valuable for investigating gene function in pathogens such as P. aeruginosa where essential and CE genes underlie clinically important phenotypes such as antibiotic susceptibility and virulence. To facilitate the use of CRISPRi in diverse bacteria-including P. aeruginosa-we developed a suite of modular, mobilizable, and integrating vectors we call, "Mobile-CRISPRi." We further optimized Mobile-CRISPRi for use in P. aeruginosa mouse models of acute lung infection by expressing the CRISPRi machinery at low levels constitutively, enabling partial knockdown of essential and CE genes without the need for an exogenous inducer. Here, we describe protocols for creating Mobile-CRISPRi knockdown strains and testing their phenotypes in a mouse pneumonia model of P. aeruginosa infection. In addition, we provide comprehensive guide RNA designs to target genes in common laboratory strains of P. aeruginosa and other Pseudomonas species., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
28. Chemoenzymatic Syntheses of Fluorine-18-Labeled Disaccharides from [ 18 F] FDG Yield Potent Sensors of Living Bacteria In Vivo .
- Author
-
Sorlin AM, López-Álvarez M, Rabbitt SJ, Alanizi AA, Shuere R, Bobba KN, Blecha J, Sakhamuri S, Evans MJ, Bayles KW, Flavell RR, Rosenberg OS, Sriram R, Desmet T, Nidetzky B, Engel J, Ohliger MA, Fraser JS, and Wilson DM
- Subjects
- Humans, Cellobiose, Staphylococcus aureus, Positron-Emission Tomography methods, Bacteria, Fluorodeoxyglucose F18, Trehalose
- Abstract
Chemoenzymatic techniques have been applied extensively to pharmaceutical development, most effectively when routine synthetic methods fail. The regioselective and stereoselective construction of structurally complex glycans is an elegant application of this approach that is seldom applied to positron emission tomography (PET) tracers. We sought a method to dimerize 2-deoxy-[
18 F]-fluoro-d-glucose ([18 F]FDG), the most common tracer used in clinical imaging, to form [18 F]-labeled disaccharides for detecting microorganisms in vivo based on their bacteria-specific glycan incorporation. When [18 F]FDG was reacted with β-d-glucose-1-phosphate in the presence of maltose phosphorylase, the α-1,4- and α-1,3-linked products 2-deoxy-[18 F]-fluoro-maltose ([18 F]FDM) and 2-deoxy-2-[18 F]-fluoro-sakebiose ([18 F]FSK) were obtained. This method was further extended with the use of trehalose (α,α-1,1), laminaribiose (β-1,3), and cellobiose (β-1,4) phosphorylases to synthesize 2-deoxy-2-[18 F]fluoro-trehalose ([18 F]FDT), 2-deoxy-2-[18 F]fluoro-laminaribiose ([18 F]FDL), and 2-deoxy-2-[18 F]fluoro-cellobiose ([18 F]FDC). We subsequently tested [18 F]FDM and [18 F]FSK in vitro , showing accumulation by several clinically relevant pathogens including Staphylococcus aureus and Acinetobacter baumannii , and demonstrated their specific uptake in vivo. Both [18 F]FDM and [18 F]FSK were stable in human serum with high accumulation in preclinical infection models. The synthetic ease and high sensitivity of [18 F]FDM and [18 F]FSK to S. aureus including methicillin-resistant (MRSA) strains strongly justify clinical translation of these tracers to infected patients. Furthermore, this work suggests that chemoenzymatic radiosyntheses of complex [18 F]FDG-derived oligomers will afford a wide array of PET radiotracers for infectious and oncologic applications.- Published
- 2023
- Full Text
- View/download PDF
29. Chemoenzymatic syntheses of fluorine-18-labeled disaccharides from [ 18 F]FDG yield potent sensors of living bacteria in vivo .
- Author
-
Sorlin AM, López-Álvarez M, Rabbitt SJ, Alanizi AA, Shuere R, Bobba KN, Blecha J, Sakhamuri S, Evans MJ, Bayles KW, Flavell RR, Rosenberg OS, Sriram R, Desmet T, Nidetzky B, Engel J, Ohliger MA, Fraser JS, and Wilson DM
- Abstract
Chemoenzymatic techniques have been applied extensively to pharmaceutical development, most effectively when routine synthetic methods fail. The regioselective and stereoselective construction of structurally complex glycans is an elegant application of this approach, that is seldom applied to positron emission tomography (PET) tracers. We sought a method to dimerize 2-deoxy-[
18 F]-fluoro-D-glucose ([18 F]FDG), the most common tracer used in clinical imaging, to form [18 F]-labeled disaccharides for detecting microorganisms in vivo based on their bacteria-specific glycan incorporation. When [18 F]FDG was reacted with β-D-glucose-1-phosphate in the presence of maltose phosphorylase, both the α-1,4 and α-1,3-linked products 2-deoxy-[18 F]-fluoro-maltose ([18 F]FDM) and 2-deoxy-2-[18 F]-fluoro-sakebiose ([18 F]FSK) were obtained. This method was further extended with the use of trehalose (α,α-1,1), laminaribiose (β-1,3), and cellobiose (β-1,4) phosphorylases to synthesize 2-deoxy-2-[18 F]fluoro-trehalose ([18 F]FDT), 2-deoxy-2-[18 F]fluoro-laminaribiose ([18 F]FDL), and 2-deoxy-2-[18 F]fluoro-cellobiose ([18 F]FDC). We subsequently tested [18 F]FDM and [18 F]FSK in vitro, showing accumulation by several clinically relevant pathogens including Staphylococcus aureus and Acinetobacter baumannii, and demonstrated their specific uptake in vivo. The lead sakebiose-derived tracer [18 F]FSK was stable in human serum and showed high uptake in preclinical models of myositis and vertebral discitis-osteomyelitis. Both the synthetic ease, and high sensitivity of [18 F]FSK to S. aureus including methicillin-resistant (MRSA) strains strongly justify clinical translation of this tracer to infected patients. Furthermore, this work suggests that chemoenzymatic radiosyntheses of complex [18 F]FDG-derived oligomers will afford a wide array of PET radiotracers for infectious and oncologic applications.- Published
- 2023
- Full Text
- View/download PDF
30. Computational pipeline provides mechanistic understanding of Omicron variant of concern neutralizing engineered ACE2 receptor traps.
- Author
-
Remesh SG, Merz GE, Brilot AF, Chio US, Rizo AN, Pospiech TH Jr, Lui I, Laurie MT, Glasgow J, Le CQ, Zhang Y, Diwanji D, Hernandez E, Lopez J, Mehmood H, Pawar KI, Pourmal S, Smith AM, Zhou F, DeRisi J, Kortemme T, Rosenberg OS, Glasgow A, Leung KK, Wells JA, and Verba KA
- Subjects
- Humans, SARS-CoV-2, Antibodies, Monoclonal, Protein Binding, Antibodies, Neutralizing, Angiotensin-Converting Enzyme 2, COVID-19
- Abstract
The SARS-CoV-2 Omicron variant, with 15 mutations in Spike receptor-binding domain (Spike-RBD), renders virtually all clinical monoclonal antibodies against WT SARS-CoV-2 ineffective. We recently engineered the SARS-CoV-2 host entry receptor, ACE2, to tightly bind WT-RBD and prevent viral entry into host cells ("receptor traps"). Here we determine cryo-EM structures of our receptor traps in complex with stabilized Spike ectodomain. We develop a multi-model pipeline combining Rosetta protein modeling software and cryo-EM to allow interface energy calculations even at limited resolution and identify interface side chains that allow for high-affinity interactions between our ACE2 receptor traps and Spike-RBD. Our structural analysis provides a mechanistic rationale for the high-affinity (0.53-4.2 nM) binding of our ACE2 receptor traps to Omicron-RBD confirmed with biolayer interferometry measurements. Finally, we show that ACE2 receptor traps potently neutralize Omicron and Delta pseudotyped viruses, providing alternative therapeutic routes to combat this evolving virus., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
31. Structure and dynamics of the essential endogenous mycobacterial polyketide synthase Pks13.
- Author
-
Kim SK, Dickinson MS, Finer-Moore J, Guan Z, Kaake RM, Echeverria I, Chen J, Pulido EH, Sali A, Krogan NJ, Rosenberg OS, and Stroud RM
- Abstract
Mycobacterium tuberculosis is currently the leading cause of death by any bacterial infection
1 . The mycolic acid layer of the cell wall is essential for viability and virulence, and the enzymes responsible for its synthesis are therefore front line targets for antimycobacterial drug development2,3 . Polyketide synthase 13 (Pks13) is a module comprised of a closely symmetric parallel dimer of chains, each encoding several enzymatic and transport functions, that carries out the condensation of two different very long chain fatty acids to produce mycolic acids that are essential components of the mycobacterial cell wall. Consequently individual enzymatic domains of Pks13 are targets for antimycobacterial drug development4 . To understand this machinery, we sought to determine the structure and domain trajectories of the dimeric multi-enzyme Pks13, a 2×198,426 Dalton complex, from protein purified endogenously from mycobacteria under normal growth conditions, to capture it with normal substrates bound trapped 'in action'. Structures of the multi-domain assembly revealed by cryogenic electron microscopy (cryoEM) define the ketosynthase (KS), linker, and acyltransferase (AT) domains, each at atomic resolution (1.8Å), with bound substrates defined at 2.4Å and 2.9Å resolution. Image classification reveals two distinct structures with alternate locations of the N-terminal acyl carrier protein (termed ACP1a, ACP1b) seen at 3.6Å and 4.6Å resolution respectively. These two structures suggest plausible intermediate states, related by a ~60Å movement of ACP1, on the pathway for substrate delivery from the fatty acyl-ACP ligase (FadD32) to the ketosynthase domain. The linking sequence between ACP1 and the KS includes an 11 amino acid sequence with 6 negatively charged side chains that lies in different positively charged grooves on the KS in ACP1a versus ACP1b structures. This charge complementarity between the extended chain and the grooves suggests some stabilization of these two distinct orientations. Other domains are visible at lower resolution and indicate flexibility relative to the KS-AT core. The chemical structures of three bound endogenous long chain fatty acid substrates with their proximal regions defined in the structures were determined by electrospray ionization mass spectrometry. The domain proximities were probed by chemical cross-linking and identified by mass spectrometry. These were incorporated into integrative structure modeling to define multiple domain configurations that transport the very long fatty acid chains throughout the multistep Pks13 mediated synthetic pathway., Competing Interests: Competing interests: The Krogan Laboratory has received research support from Vir Biotechnology, F. Hoffmann-La Roche, and Rezo Therapeutics. Nevan Krogan has financially compensated consulting agreements with the Icahn School of Medicine at Mount Sinai, New York, Maze Therapeutics, Interline Therapeutics, Rezo Therapeutics, GEn1E Lifesciences, Inc. and Twist Bioscience Corp. He is on the Board of Directors of Rezo Therapeutics and is a shareholder in Tenaya Therapeutics, Maze Therapeutics, Rezo Therapeutics, and Interline Therapeutics.- Published
- 2023
- Full Text
- View/download PDF
32. Workshop-based learning and networking: a scalable model for research capacity strengthening in low- and middle-income countries.
- Author
-
Perier C, Nasinghe E, Charles I, Ssetaba LJ, Ahyong V, Bangs D, Beatty PR, Czudnochowski N, Diallo A, Dugan E, Fabius JM, Fong Baker H, Gardner J, Isaacs S, Joanah B, Kalantar K, Kateete D, Knight M, Krasilnikov M, Krogan NJ, Langelier C, Lee E, Li LM, Licht D, Lien K, Lyons Z, Mboowa G, Mwebaza I, Mwesigwa S, Nalwadda G, Nichols R, Penaranda ME, Petnic S, Phelps M, Popper SJ, Rape M, Reingold A, Robbins R, Rosenberg OS, Savage DF, Schildhauer S, Settles ML, Sserwadda I, Stanley S, Tato CM, Tsitsiklis A, Van Dis E, Vanaerschot M, Vinden J, Cox JS, Joloba ML, and Schaletzky J
- Subjects
- Capacity Building, Humans, Poverty, Students, Universities, Developing Countries, Global Health
- Abstract
Science education and research have the potential to drive profound change in low- and middle-income countries (LMICs) through encouraging innovation, attracting industry, and creating job opportunities. However, in LMICs, research capacity is often limited, and acquisition of funding and access to state-of-the-art technologies is challenging. The Alliance for Global Health and Science (the Alliance) was founded as a partnership between the University of California, Berkeley (USA) and Makerere University (Uganda), with the goal of strengthening Makerere University's capacity for bioscience research. The flagship program of the Alliance partnership is the MU/UCB Biosciences Training Program, an in-country, hands-on workshop model that trains a large number of students from Makerere University in infectious disease and molecular biology research. This approach nucleates training of larger and more diverse groups of students, development of mentoring and bi-directional research partnerships, and support of the local economy. Here, we describe the project, its conception, implementation, challenges, and outcomes of bioscience research workshops. We aim to provide a blueprint for workshop implementation, and create a valuable resource for bioscience research capacity strengthening in LMICs.
- Published
- 2022
- Full Text
- View/download PDF
33. Computational pipeline provides mechanistic understanding of Omicron variant of concern neutralizing engineered ACE2 receptor traps.
- Author
-
Remesh SG, Merz GE, Brilot AF, Chio US, Rizo AN, Pospiech TH Jr, Lui I, Laurie MT, Glasgow J, Le CQ, Zhang Y, Diwanji D, Hernandez E, Lopez J, Pawar KI, Pourmal S, Smith AM, Zhou F, DeRisi J, Kortemme T, Rosenberg OS, Glasgow A, Leung KK, Wells JA, and Verba KA
- Abstract
The SARS-CoV-2 Omicron variant, with 15 mutations in Spike receptor binding domain (Spike-RBD), renders virtually all clinical monoclonal antibodies against WT SARS-CoV-2 ineffective. We recently engineered the SARS-CoV-2 host entry receptor, ACE2, to tightly bind WT-Spike-RBD and prevent viral entry into host cells ("receptor traps"). Here we determine cryo-EM structures of our receptor traps in complex with full length Spike. We develop a multi-model pipeline combining Rosetta protein modeling software and cryo-EM to allow interface energy calculations even at limited resolution and identify interface side chains that allow for high affinity interactions between our ACE2 receptor traps and Spike-RBD. Our structural analysis provides a mechanistic rationale for the high affinity (0.53 - 4.2nM) binding of our ACE2 receptor traps to Omicron-RBD confirmed with biolayer interferometry measurements. Finally, we show that ACE2 receptor traps potently neutralize Omicron- and Delta-pseudotyped viruses, providing alternative therapeutic routes to combat this evolving virus.
- Published
- 2022
- Full Text
- View/download PDF
34. Discovery Proteomics Analysis Determines That Driver Oncogenes Suppress Antiviral Defense Pathways Through Reduction in Interferon-β Autocrine Stimulation.
- Author
-
Solomon PE, Kirkemo LL, Wilson GM, Leung KK, Almond MH, Sayles LC, Sweet-Cordero EA, Rosenberg OS, Coon JJ, and Wells JA
- Subjects
- Animals, Antiviral Restriction Factors, Carcinogenesis, Cell Line, Tumor, Humans, Proto-Oncogene Proteins p21(ras) genetics, SARS-CoV-2, COVID-19 immunology, Interferon-beta immunology, Oncogenes, Proteomics
- Abstract
Since the discovery of oncogenes, there has been tremendous interest to understand their mechanistic basis and to develop broadly actionable therapeutics. Some of the most frequently activated oncogenes driving diverse cancers are c-MYC, EGFR, HER2, AKT, KRAS, BRAF, and MEK. Using a reductionist approach, we explored how cellular proteomes are remodeled in isogenic cell lines engineered with or without these driver oncogenes. The most striking discovery for all oncogenic models was the systematic downregulation of scores of antiviral proteins regulated by type 1 interferon. These findings extended to cancer cell lines and patient-derived xenograft models of highly refractory pancreatic cancer and osteosarcoma driven by KRAS and MYC oncogenes. The oncogenes reduced basal expression of and autocrine stimulation by type 1 interferon causing remarkable convergence on common phenotypic and functional profiles. In particular, there was dramatically lower expression of dsRNA sensors including DDX58 (RIG-I) and OAS proteins, which resulted in attenuated functional responses when the oncogenic cells were treated with the dsRNA mimetic, polyI:C, and increased susceptibility to infection with an RNA virus shown using SARS-CoV-2. Our reductionist approach provides molecular and functional insights connected to immune evasion hallmarks in cancers and suggests therapeutic opportunities., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
35. Leaks in the Pipeline: a Failure Analysis of Gram-Negative Antibiotic Development from 2010 to 2020.
- Author
-
Prasad NK, Seiple IB, Cirz RT, and Rosenberg OS
- Subjects
- Drug Resistance, Multiple, Bacterial, Gram-Negative Bacteria, Humans, United States, United States Food and Drug Administration, Anti-Bacterial Agents pharmacology, Anti-Bacterial Agents therapeutic use, Gram-Negative Bacterial Infections drug therapy, Gram-Negative Bacterial Infections microbiology
- Abstract
The World Health Organization (WHO) has warned that our current arsenal of antibiotics is not innovative enough to face impending infectious diseases, especially those caused by multidrug-resistant Gram-negative pathogens. Although the current preclinical pipeline is well stocked with novel candidates, the last U.S. Food and Drug Administration (FDA)-approved antibiotic with a novel mechanism of action against Gram-negative bacteria was discovered nearly 60 years ago. Of all the antibiotic candidates that initiated investigational new drug (IND) applications in the 2000s, 17% earned FDA approval within 12 years, while an overwhelming 62% were discontinued in that time frame. These "leaks" in the clinical pipeline, where compounds with clinical potential are abandoned during clinical development, indicate that scientific innovations are not reaching the clinic and providing benefits to patients. This is true for not only novel candidates but also candidates from existing antibiotic classes with clinically validated targets. By identifying the sources of the leaks in the clinical pipeline, future developmental efforts can be directed toward strategies that are more likely to flow into clinical use. In this review, we conduct a detailed failure analysis of clinical candidates with Gram-negative activity that have fallen out of the clinical pipeline over the past decade. Although limited by incomplete data disclosure from companies engaging in antibiotic development, we attempt to distill the developmental challenges faced by each discontinued candidate. It is our hope that this insight can help de-risk antibiotic development and bring new, effective antibiotics to the clinic.
- Published
- 2022
- Full Text
- View/download PDF
36. CryoEM and AI reveal a structure of SARS-CoV-2 Nsp2, a multifunctional protein involved in key host processes.
- Author
-
Gupta M, Azumaya CM, Moritz M, Pourmal S, Diallo A, Merz GE, Jang G, Bouhaddou M, Fossati A, Brilot AF, Diwanji D, Hernandez E, Herrera N, Kratochvil HT, Lam VL, Li F, Li Y, Nguyen HC, Nowotny C, Owens TW, Peters JK, Rizo AN, Schulze-Gahmen U, Smith AM, Young ID, Yu Z, Asarnow D, Billesbølle C, Campbell MG, Chen J, Chen KH, Chio US, Dickinson MS, Doan L, Jin M, Kim K, Li J, Li YL, Linossi E, Liu Y, Lo M, Lopez J, Lopez KE, Mancino A, Moss FR 3rd, Paul MD, Pawar KI, Pelin A, Pospiech TH Jr, Puchades C, Remesh SG, Safari M, Schaefer K, Sun M, Tabios MC, Thwin AC, Titus EW, Trenker R, Tse E, Tsui TKM, Wang F, Zhang K, Zhang Y, Zhao J, Zhou F, Zhou Y, Zuliani-Alvarez L, Agard DA, Cheng Y, Fraser JS, Jura N, Kortemme T, Manglik A, Southworth DR, Stroud RM, Swaney DL, Krogan NJ, Frost A, Rosenberg OS, and Verba KA
- Abstract
The SARS-CoV-2 protein Nsp2 has been implicated in a wide range of viral processes, but its exact functions, and the structural basis of those functions, remain unknown. Here, we report an atomic model for full-length Nsp2 obtained by combining cryo-electron microscopy with deep learning-based structure prediction from AlphaFold2. The resulting structure reveals a highly-conserved zinc ion-binding site, suggesting a role for Nsp2 in RNA binding. Mapping emerging mutations from variants of SARS-CoV-2 on the resulting structure shows potential host-Nsp2 interaction regions. Using structural analysis together with affinity tagged purification mass spectrometry experiments, we identify Nsp2 mutants that are unable to interact with the actin-nucleation-promoting WASH protein complex or with GIGYF2, an inhibitor of translation initiation and modulator of ribosome-associated quality control. Our work suggests a potential role of Nsp2 in linking viral transcription within the viral replication-transcription complexes (RTC) to the translation initiation of the viral message. Collectively, the structure reported here, combined with mutant interaction mapping, provides a foundation for functional studies of this evolutionary conserved coronavirus protein and may assist future drug design.
- Published
- 2021
- Full Text
- View/download PDF
37. Nuclear Imaging of Bacterial Infection: The State of the Art and Future Directions.
- Author
-
Polvoy I, Flavell RR, Rosenberg OS, Ohliger MA, and Wilson DM
- Subjects
- Animals, Bacterial Infections drug therapy, Humans, Bacterial Infections diagnostic imaging, Diagnostic Imaging methods, Nuclear Medicine methods
- Abstract
Increased mortality rates from infectious diseases is a growing public health concern. Successful management of acute bacterial infections requires early diagnosis and treatment, which are not always easy to achieve. Structural imaging techniques such as CT and MRI are often applied to this problem. However, these methods generally rely on secondary inflammatory changes and are frequently not specific to infection. The use of nuclear medicine techniques can add crucial complementary information, allowing visualization of infectious pathophysiology beyond morphologic imaging. This review will discuss the current structural and functional imaging techniques used for the diagnosis of bacterial infection and their roles in different clinical scenarios. We will also present several new radiotracers in development, with an emphasis on probes targeting bacteria-specific metabolism. As highlighted by the current coronavirus disease 2019 epidemic, caused by the novel severe acute respiratory syndrome coronavirus 2, similar thinking may apply in imaging viral pathogens; for this case, prominent effects on host proteins, most notably angiotensin-converting enzyme 2, might also provide worthwhile imaging targets., (© 2020 by the Society of Nuclear Medicine and Molecular Imaging.)
- Published
- 2020
- Full Text
- View/download PDF
38. Engineered ACE2 receptor traps potently neutralize SARS-CoV-2.
- Author
-
Glasgow A, Glasgow J, Limonta D, Solomon P, Lui I, Zhang Y, Nix MA, Rettko NJ, Lim SA, Zha S, Yamin R, Kao K, Rosenberg OS, Ravetch JV, Wiita AP, Leung KK, Zhou XX, Hobman TC, Kortemme T, and Wells JA
- Abstract
An essential mechanism for SARS-CoV-1 and -2 infection begins with the viral spike protein binding to the human receptor protein angiotensin-converting enzyme II (ACE2). Here we describe a stepwise engineering approach to generate a set of affinity optimized, enzymatically inactivated ACE2 variants that potently block SARS-CoV-2 infection of cells. These optimized receptor traps tightly bind the receptor binding domain (RBD) of the viral spike protein and prevent entry into host cells. We first computationally designed the ACE2-RBD interface using a two-stage flexible protein backbone design process that improved affinity for the RBD by up to 12-fold. These designed receptor variants were affinity matured an additional 14-fold by random mutagenesis and selection using yeast surface display. The highest affinity variant contained seven amino acid changes and bound to the RBD 170-fold more tightly than wild-type ACE2. With the addition of the natural ACE2 collectrin domain and fusion to a human Fc domain for increased stabilization and avidity, the most optimal ACE2 receptor traps neutralized SARS-CoV-2 pseudotyped lentivirus and authentic SARS-CoV-2 virus with half-maximal inhibitory concentrations (IC50) in the 10-100 ng/ml range. Engineered ACE2 receptor traps offer a promising route to fighting infections by SARS-CoV-2 and other ACE2-utilizing coronaviruses, with the key advantage that viral resistance would also likely impair viral entry. Moreover, such traps can be predesigned for viruses with known entry receptors for faster therapeutic response without the need for neutralizing antibodies isolated or generated from convalescent patients.
- Published
- 2020
- Full Text
- View/download PDF
39. A SARS-CoV-2-Human Protein-Protein Interaction Map Reveals Drug Targets and Potential Drug-Repurposing.
- Author
-
Gordon DE, Jang GM, Bouhaddou M, Xu J, Obernier K, O'Meara MJ, Guo JZ, Swaney DL, Tummino TA, Huettenhain R, Kaake RM, Richards AL, Tutuncuoglu B, Foussard H, Batra J, Haas K, Modak M, Kim M, Haas P, Polacco BJ, Braberg H, Fabius JM, Eckhardt M, Soucheray M, Bennett MJ, Cakir M, McGregor MJ, Li Q, Naing ZZC, Zhou Y, Peng S, Kirby IT, Melnyk JE, Chorba JS, Lou K, Dai SA, Shen W, Shi Y, Zhang Z, Barrio-Hernandez I, Memon D, Hernandez-Armenta C, Mathy CJP, Perica T, Pilla KB, Ganesan SJ, Saltzberg DJ, Ramachandran R, Liu X, Rosenthal SB, Calviello L, Venkataramanan S, Liboy-Lugo J, Lin Y, Wankowicz SA, Bohn M, Sharp PP, Trenker R, Young JM, Cavero DA, Hiatt J, Roth TL, Rathore U, Subramanian A, Noack J, Hubert M, Roesch F, Vallet T, Meyer B, White KM, Miorin L, Rosenberg OS, Verba KA, Agard D, Ott M, Emerman M, Ruggero D, García-Sastre A, Jura N, von Zastrow M, Taunton J, Ashworth A, Schwartz O, Vignuzzi M, d'Enfert C, Mukherjee S, Jacobson M, Malik HS, Fujimori DG, Ideker T, Craik CS, Floor S, Fraser JS, Gross J, Sali A, Kortemme T, Beltrao P, Shokat K, Shoichet BK, and Krogan NJ
- Abstract
An outbreak of the novel coronavirus SARS-CoV-2, the causative agent of COVID-19 respiratory disease, has infected over 290,000 people since the end of 2019, killed over 12,000, and caused worldwide social and economic disruption
1,2 . There are currently no antiviral drugs with proven efficacy nor are there vaccines for its prevention. Unfortunately, the scientific community has little knowledge of the molecular details of SARS-CoV-2 infection. To illuminate this, we cloned, tagged and expressed 26 of the 29 viral proteins in human cells and identified the human proteins physically associated with each using affinity-purification mass spectrometry (AP-MS), which identified 332 high confidence SARS-CoV-2-human protein-protein interactions (PPIs). Among these, we identify 67 druggable human proteins or host factors targeted by 69 existing FDA-approved drugs, drugs in clinical trials and/or preclinical compounds, that we are currently evaluating for efficacy in live SARS-CoV-2 infection assays. The identification of host dependency factors mediating virus infection may provide key insights into effective molecular targets for developing broadly acting antiviral therapeutics against SARS-CoV-2 and other deadly coronavirus strains., Competing Interests: Conflicts: The Krogan Laboratory has received research support from Vir Biotechnology and F. Hoffmann-La Roche. Kevan Shokat has consulting agreements for the following companies involving cash and/or stock compensation: Black Diamond Therapeutics, BridGene Biosciences, Denali Therapeutics, Dice Molecules, eFFECTOR Therapeutics, Erasca, Genentech/Roche, Janssen Pharmaceuticals, Kumquat Biosciences, Kura Oncology, Merck, Mitokinin, Petra Pharma, Qulab Inc. Revolution Medicines, Type6 Therapeutics, Venthera, Wellspring Biosciences (Araxes Pharma). Jack Taunton is a cofounder and shareholder of Global Blood Therapeutics, Principia Biopharma, Kezar Life Sciences, and Cedilla Therapeutics. Jack Taunton and Phillip P. Sharp are listed as inventors on a provisional patent application describing PS3061.- Published
- 2020
- Full Text
- View/download PDF
40. Sensing Living Bacteria in Vivo Using d-Alanine-Derived 11 C Radiotracers.
- Author
-
Parker MFL, Luu JM, Schulte B, Huynh TL, Stewart MN, Sriram R, Yu MA, Jivan S, Turnbaugh PJ, Flavell RR, Rosenberg OS, Ohliger MA, and Wilson DM
- Abstract
Incorporation of d-amino acids into peptidoglycan is a unique metabolic feature of bacteria. Since d-amino acids are not metabolic substrates in most mammalian tissues, this difference can be exploited to detect living bacteria in vivo . Given the prevalence of d-alanine in peptidoglycan muropeptides, as well as its role in several antibiotic mechanisms, we targeted this amino acid for positron emission tomography (PET) radiotracer development. d-[3-
11 C]Alanine and the dipeptide d-[3-11 C]alanyl-d-alanine were synthesized via asymmetric alkylation of glycine-derived Schiff-base precursors with [11 C]methyl iodide in the presence of a cinchonidinium phase-transfer catalyst. In cell experiments, both tracers showed accumulation by a wide variety of both Gram-positive and Gram-negative pathogens including Staphylococcus aureus and Pseudomonas aeruginosa . In a mouse model of acute bacterial myositis, d-[3-11 C]alanine was accumulated by living microorganisms but was not taken up in areas of sterile inflammation. When compared to existing clinical nuclear imaging tools, specifically 2-deoxy-2-[18 F]fluoro-d-glucose and a gallium citrate radiotracer, d-alanine showed more bacteria-specific uptake. Decreased d-[3-11 C]alanine uptake was also observed in antibiotic-sensitive microbes after antimicrobial therapy, when compared to that in resistant organisms. Finally, prominent uptake of d-[3-11 C]alanine uptake was seen in rodent models of discitis-osteomyelitis and P. aeruginosa pneumonia. These data provide strong justification for clinical translation of d-[3-11 C]alanine to address a number of important human infections., Competing Interests: The authors declare no competing financial interest., (Copyright © 2020 American Chemical Society.)- Published
- 2020
- Full Text
- View/download PDF
41. The structure of the endogenous ESX-3 secretion system.
- Author
-
Poweleit N, Czudnochowski N, Nakagawa R, Trinidad DD, Murphy KC, Sassetti CM, and Rosenberg OS
- Subjects
- Bacterial Proteins genetics, Bacterial Proteins ultrastructure, Chromosomes chemistry, Chromosomes genetics, Epitopes chemistry, Epitopes genetics, Mycobacterium smegmatis ultrastructure, Operon genetics, Type VII Secretion Systems genetics, Type VII Secretion Systems ultrastructure, Bacterial Proteins chemistry, Mycobacterium smegmatis chemistry, Protein Transport genetics, Type VII Secretion Systems chemistry
- Abstract
The ESX (or Type VII) secretion systems are protein export systems in mycobacteria and many Gram-positive bacteria that mediate a broad range of functions including virulence, conjugation, and metabolic regulation. These systems translocate folded dimers of WXG100-superfamily protein substrates across the cytoplasmic membrane. We report the cryo-electron microscopy structure of an ESX-3 system, purified using an epitope tag inserted with recombineering into the chromosome of the model organism Mycobacterium smegmatis . The structure reveals a stacked architecture that extends above and below the inner membrane of the bacterium. The ESX-3 protomer complex is assembled from a single copy of the EccB
3 , EccC3 , and EccE3 and two copies of the EccD3 protein. In the structure, the protomers form a stable dimer that is consistent with assembly into a larger oligomer. The ESX-3 structure provides a framework for further study of these important bacterial transporters., Competing Interests: NP, NC, RN, DT, KM, CS, OR No competing interests declared, (© 2019, Poweleit et al.)- Published
- 2019
- Full Text
- View/download PDF
42. Modulating Pathogenesis with Mobile-CRISPRi.
- Author
-
Qu J, Prasad NK, Yu MA, Chen S, Lyden A, Herrera N, Silvis MR, Crawford E, Looney MR, Peters JM, and Rosenberg OS
- Subjects
- Animals, CRISPR-Associated Protein 9, Gene Knockdown Techniques, Genes, Bacterial, Immunoblotting, Male, Mice, Mice, Inbred C57BL, Pneumonia, Bacterial metabolism, Pseudomonas Infections metabolism, Pseudomonas aeruginosa genetics, Reverse Transcriptase Polymerase Chain Reaction, Type III Secretion Systems genetics, CRISPR-Cas Systems, Gene Editing methods, Pneumonia, Bacterial microbiology, Pseudomonas Infections microbiology, Pseudomonas aeruginosa pathogenicity
- Abstract
Conditionally essential (CE) genes are required by pathogenic bacteria to establish and maintain infections. CE genes encode virulence factors, such as secretion systems and effector proteins, as well as biosynthetic enzymes that produce metabolites not found in the host environment. Due to their outsized importance in pathogenesis, CE gene products are attractive targets for the next generation of antimicrobials. However, the precise manipulation of CE gene expression in the context of infection is technically challenging, limiting our ability to understand the roles of CE genes in pathogenesis and accordingly design effective inhibitors. We previously developed a suite of CRISPR interference-based gene knockdown tools that are transferred by conjugation and stably integrate into bacterial genomes that we call Mobile-CRISPRi. Here, we show the efficacy of Mobile-CRISPRi in controlling CE gene expression in an animal infection model. We optimize Mobile-CRISPRi in Pseudomonas aeruginosa for use in a murine model of pneumonia by tuning the expression of CRISPRi components to avoid nonspecific toxicity. As a proof of principle, we demonstrate that knock down of a CE gene encoding the type III secretion system (T3SS) activator ExsA blocks effector protein secretion in culture and attenuates virulence in mice. We anticipate that Mobile-CRISPRi will be a valuable tool to probe the function of CE genes across many bacterial species and pathogenesis models. IMPORTANCE Antibiotic resistance is a growing threat to global health. To optimize the use of our existing antibiotics and identify new targets for future inhibitors, understanding the fundamental drivers of bacterial growth in the context of the host immune response is paramount. Historically, these genetic drivers have been difficult to manipulate precisely, as they are requisite for pathogen survival. Here, we provide the first application of Mobile-CRISPRi to study conditionally essential virulence genes in mouse models of lung infection through partial gene perturbation. We envision the use of Mobile-CRISPRi in future pathogenesis models and antibiotic target discovery efforts., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
43. Imaging Active Infection in vivo Using D-Amino Acid Derived PET Radiotracers.
- Author
-
Neumann KD, Villanueva-Meyer JE, Mutch CA, Flavell RR, Blecha JE, Kwak T, Sriram R, VanBrocklin HF, Rosenberg OS, Ohliger MA, and Wilson DM
- Subjects
- Animals, Disease Models, Animal, Escherichia coli isolation & purification, Escherichia coli metabolism, Female, Isotope Labeling, Mice, Inbred CBA, Staphylococcus aureus isolation & purification, Staphylococcus aureus metabolism, Carbon Radioisotopes analysis, Escherichia coli Infections diagnostic imaging, Methionine metabolism, Positron-Emission Tomography methods, Staphylococcal Infections diagnostic imaging
- Abstract
Occult bacterial infections represent a worldwide health problem. Differentiating active bacterial infection from sterile inflammation can be difficult using current imaging tools. Present clinically viable methodologies either detect morphologic changes (CT/ MR), recruitment of immune cells (
111 In-WBC SPECT), or enhanced glycolytic flux seen in inflammatory cells (18 F-FDG PET). However, these strategies are often inadequate to detect bacterial infection and are not specific for living bacteria. Recent approaches have taken advantage of key metabolic differences between prokaryotic and eukaryotic organisms, allowing easier distinction between bacteria and their host. In this report, we exploited one key difference, bacterial cell wall biosynthesis, to detect living bacteria using a positron-labeled D-amino acid. After screening several14 C D-amino acids for their incorporation into E. coli in culture, we identified D-methionine as a probe with outstanding radiopharmaceutical potential. Based on an analogous procedure to that used for L-[methyl-11 C]methionine ([11 C] L-Met), we developed an enhanced asymmetric synthesis of D-[methyl-11 C]methionine ([11 C] D-Met), and showed that it can rapidly and selectively differentiate both E. coli and S. aureus infections from sterile inflammation in vivo. We believe that the ease of [11 C] D-Met radiosynthesis, coupled with its rapid and specific in vivo bacterial accumulation, make it an attractive radiotracer for infection imaging in clinical practice.- Published
- 2017
- Full Text
- View/download PDF
44. Chlamydia interfere with an interaction between the mannose-6-phosphate receptor and sorting nexins to counteract host restriction.
- Author
-
Elwell CA, Czudnochowski N, von Dollen J, Johnson JR, Nakagawa R, Mirrashidi K, Krogan NJ, Engel JN, and Rosenberg OS
- Subjects
- Animals, Bacterial Proteins chemistry, Bacterial Proteins genetics, Crystallography, X-Ray, DNA Mutational Analysis, Mice, Models, Molecular, Protein Conformation, Protein Interaction Mapping, Receptor, IGF Type 2 chemistry, Receptor, IGF Type 2 genetics, Sorting Nexins chemistry, Sorting Nexins genetics, Bacterial Proteins metabolism, Chlamydia trachomatis immunology, Chlamydia trachomatis physiology, Host-Pathogen Interactions, Immune Evasion, Receptor, IGF Type 2 metabolism, Sorting Nexins metabolism
- Abstract
Chlamydia trachomatis is an obligate intracellular pathogen that resides in a membrane-bound compartment, the inclusion. The bacteria secrete a unique class of proteins, Incs, which insert into the inclusion membrane and modulate the host-bacterium interface. We previously reported that IncE binds specifically to the Sorting Nexin 5 Phox domain (SNX5-PX) and disrupts retromer trafficking. Here, we present the crystal structure of the SNX5-PX:IncE complex, showing IncE bound to a unique and highly conserved hydrophobic groove on SNX5. Mutagenesis of the SNX5-PX:IncE binding surface disrupts a previously unsuspected interaction between SNX5 and the cation-independent mannose-6-phosphate receptor (CI-MPR). Addition of IncE peptide inhibits the interaction of CI-MPR with SNX5. Finally, C. trachomatis infection interferes with the SNX5:CI-MPR interaction, suggesting that IncE and CI-MPR are dependent on the same binding surface on SNX5. Our results provide new insights into retromer assembly and underscore the power of using pathogens to discover disease-related cell biology.
- Published
- 2017
- Full Text
- View/download PDF
45. Substrates Control Multimerization and Activation of the Multi-Domain ATPase Motor of Type VII Secretion.
- Author
-
Rosenberg OS, Dovala D, Li X, Connolly L, Bendebury A, Finer-Moore J, Holton J, Cheng Y, Stroud RM, and Cox JS
- Subjects
- Actinobacteria metabolism, Adenosine Triphosphatases chemistry, Adenosine Triphosphatases metabolism, Crystallography, X-Ray, Models, Molecular, Mycobacterium tuberculosis enzymology, Mycobacterium tuberculosis metabolism, Mycobacterium tuberculosis pathogenicity, Staphylococcus aureus enzymology, Staphylococcus aureus metabolism, Staphylococcus aureus pathogenicity, Virulence Factors chemistry, Actinobacteria enzymology, Bacterial Secretion Systems
- Abstract
Mycobacterium tuberculosis and Staphylococcus aureus secrete virulence factors via type VII protein secretion (T7S), a system that intriguingly requires all of its secretion substrates for activity. To gain insights into T7S function, we used structural approaches to guide studies of the putative translocase EccC, a unique enzyme with three ATPase domains, and its secretion substrate EsxB. The crystal structure of EccC revealed that the ATPase domains are joined by linker/pocket interactions that modulate its enzymatic activity. EsxB binds via its signal sequence to an empty pocket on the C-terminal ATPase domain, which is accompanied by an increase in ATPase activity. Surprisingly, substrate binding does not activate EccC allosterically but, rather, by stimulating its multimerization. Thus, the EsxB substrate is also an integral T7S component, illuminating a mechanism that helps to explain interdependence of substrates, and suggests a model in which binding of substrates modulates their coordinate release from the bacterium., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
46. The crystal structure of S. cerevisiae Sad1, a catalytically inactive deubiquitinase that is broadly required for pre-mRNA splicing.
- Author
-
Hadjivassiliou H, Rosenberg OS, and Guthrie C
- Subjects
- Amino Acid Sequence, Catalysis, Cell Cycle Proteins genetics, Checkpoint Kinase 2 genetics, Protein Conformation, RNA Precursors chemistry, RNA Precursors genetics, Saccharomyces cerevisiae chemistry, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Sequence Homology, Amino Acid, Spliceosomes chemistry, Spliceosomes genetics, Ubiquitin chemistry, Ubiquitin metabolism, Alternative Splicing genetics, Cell Cycle Proteins chemistry, Checkpoint Kinase 2 chemistry, Crystallography, X-Ray, Saccharomyces cerevisiae Proteins chemistry, Ubiquitin-Specific Proteases chemistry
- Abstract
Sad1 is an essential splicing factor initially identified in a genetic screen in Saccharomyces cerevisiae for snRNP assembly defects. Based on sequence homology, Sad1, or USP39 in humans, is predicted to comprise two domains: a zinc finger ubiquitin binding domain (ZnF-UBP) and an inactive ubiquitin-specific protease (iUSP) domain, both of which are well conserved. The role of these domains in splicing and their interaction with ubiquitin are unknown. We first used splicing microarrays to analyze Sad1 function in vivo and found that Sad1 is critical for the splicing of nearly all yeast intron-containing genes. By using in vitro assays, we then showed that it is required for the assembly of the active spliceosome. To gain structural insights into Sad1 function, we determined the crystal structure of the full-length protein at 1.8 Å resolution. In the structure, the iUSP domain forms the characteristic ubiquitin binding pocket, though with an amino acid substitution in the active site that results in complete inactivation of the enzymatic activity of the domain. The ZnF-UBP domain of Sad1 shares high structural similarly to other ZnF-UBPs; however, Sad1's ZnF-UBP does not possess the canonical ubiquitin binding motif. Given the precedents for ZnF-UBP domains to function as activators for their neighboring USP domains, we propose that Sad1's ZnF-UBP acts in a ubiquitin-independent capacity to recruit and/or activate Sad1's iUSP domain to interact with the spliceosome.
- Published
- 2014
- Full Text
- View/download PDF
47. A mechanism for tunable autoinhibition in the structure of a human Ca2+/calmodulin- dependent kinase II holoenzyme.
- Author
-
Chao LH, Stratton MM, Lee IH, Rosenberg OS, Levitz J, Mandell DJ, Kortemme T, Groves JT, Schulman H, and Kuriyan J
- Subjects
- Amino Acid Sequence, Animals, Crystallography, X-Ray, Holoenzymes chemistry, Holoenzymes metabolism, Humans, Models, Molecular, Protein Conformation, Protein Structure, Tertiary, Sequence Alignment, Calcium-Calmodulin-Dependent Protein Kinase Type 2 chemistry, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism
- Abstract
Calcium/calmodulin-dependent kinase II (CaMKII) forms a highly conserved dodecameric assembly that is sensitive to the frequency of calcium pulse trains. Neither the structure of the dodecameric assembly nor how it regulates CaMKII are known. We present the crystal structure of an autoinhibited full-length human CaMKII holoenzyme, revealing an unexpected compact arrangement of kinase domains docked against a central hub, with the calmodulin-binding sites completely inaccessible. We show that this compact docking is important for the autoinhibition of the kinase domains and for setting the calcium response of the holoenzyme. Comparison of CaMKII isoforms, which differ in the length of the linker between the kinase domain and the hub, demonstrates that these interactions can be strengthened or weakened by changes in linker length. This equilibrium between autoinhibited states provides a simple mechanism for tuning the calcium response without changes in either the hub or the kinase domains., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
48. Sulfadoxine-pyrimethamine in treatment of malaria in Western Kenya: increasing resistance and underdosing.
- Author
-
Terlouw DJ, Nahlen BL, Courval JM, Kariuki SK, Rosenberg OS, Oloo AJ, Kolczak MS, Hawley WA, Lal AA, and Kuile FO
- Subjects
- Anti-Infective Agents administration & dosage, Antimalarials administration & dosage, Body Weight, Drug Combinations, Drug Resistance, Female, Humans, Infant, Kenya, Male, Prospective Studies, Pyrimethamine administration & dosage, Sulfadoxine administration & dosage, Anti-Infective Agents therapeutic use, Antimalarials therapeutic use, Malaria drug therapy, Pyrimethamine therapeutic use, Sulfadoxine therapeutic use
- Abstract
Between 1993 and 1999, we monitored the efficacy of sulfadoxine-pyrimethamine in 1175 children aged <24 months receiving 2789 treatments for falciparum malaria in western Kenya using a widely deployed age-based dose regimen: infants, 125 plus 6.25 mg (sulfadoxine plus pyrimethamine); children aged 12 to 23 months; 250 plus 12.5 mg. Cumulative treatment failure by day 7, defined as early clinical failure by day 3 or presence of parasitemia on day 7, increased from 18% in 1993 to 1994 to 22% in 1997 to 1998 (P-trend test = 0.20). Based on body weight, the median dose received was 20 plus 1.00 mg/kg, and 73% of the treatments were given at lower than the recommended target dose of 25 plus 1.25 mg/kg. Underdosing accounted for 26% of cumulative treatment failures. After the dose was increased in 1998 (median, 36 plus 1.8 mg/kg), only 4.2% of patients received less than 25 plus 1.25 mg/kg and there was no association with treatment failure. However, the proportion of cumulative treatment failure continued to increase to 27% by 1999 (P-trend test = 0.03). These results raise concern about the longevity of sulfadoxine-pyrimethamine in these settings. Underdosing may have contributed to the rate at which sulfadoxine-pyrimethamine resistance developed in this area. Treatment guidelines should ensure that adequate doses are given from the initial deployment of antimalarials onward.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.