43 results on '"Obernier K"'
Search Results
2. Contributors
- Author
-
Akassoglou, Katerina, primary, Allen, Nicola J., additional, Alsina, Fernando C., additional, Alunni, Alessandro, additional, Alvarez-Buylla, A., additional, Andrews, Madeline G., additional, Ang, S.-L., additional, Appel, B., additional, Arefin, Badrul, additional, Bahrampour, Shahrzad, additional, Bai, Q.-R., additional, Bally-Cuif, Laure, additional, Batista-Brito, Renata, additional, Baumgardt, Magnus, additional, Benito-Sipos, Jonathan, additional, Bergles, D.E., additional, Bhaduri, Aparna, additional, Blaess, S., additional, Bonney, Stephanie, additional, Bosze, Bernadett, additional, Breunig, Joshua J., additional, Brown, Nadean L., additional, Buffington, S.A., additional, Call, C.L., additional, Campbell, K., additional, Cardona, Astrid E., additional, Catela, Catarina, additional, Cebrián-Silla, A., additional, Cheng, Yi-Ting, additional, Chizhikov, Victor V., additional, Coolen, Marion, additional, Curt, Jesús Rodriguez, additional, Davalos, Dimitrios, additional, De Biase, L.M., additional, Deneen, Benjamin, additional, Durak, Omer, additional, Fame, Ryann M., additional, Fancy, Stephen P.J., additional, Fishell, Gord, additional, Foucher, Isabelle, additional, Fuentealba, L., additional, Gage, Fred H., additional, Galas, Ludovic, additional, Grande, Andrew W., additional, Grove, Elizabeth A., additional, Haigh, J.L., additional, Hébert, Jean, additional, Hobert, Oliver, additional, Hufnagel, Robert B., additional, Huttner, Wieland B., additional, Itoh, Yasuhiro, additional, Jessen, K.R., additional, Johnson, Jane E., additional, Karzbrun, Eyal, additional, Komuro, Yutaro, additional, Komuro, Hitoshi, additional, Kriegstein, Arnold R., additional, Lambert, J.T., additional, Long, Katherine R., additional, López-Bendito, Guillermina, additional, MacDonald, Jessica L., additional, Macklis, Jeffrey D., additional, Marchetto, Maria Carolina, additional, Martini, Francisco J., additional, Matise, Michael P., additional, Merkle, F.T., additional, Meunier, A., additional, Millen, Kathleen J., additional, Miller, Robert H., additional, Mirsky, R., additional, Mishra, Swati, additional, Molofsky, Anna Victoria, additional, Monedero Cobeta, Ignacio, additional, Monk, K., additional, Monuki, Edwin S., additional, Nakafuku, Masato, additional, Nakamura, Harukazu, additional, Nelson, Branden R., additional, Nord, A.S., additional, Obernier, K., additional, Ohno, Nobuhiko, additional, Ozkan, Abdulkadir, additional, Parkinson, David B., additional, Peter, Manuel, additional, Pleasure, Samuel J., additional, Rasband, M.N., additional, Reiner, Orly, additional, Rowitch, D.H., additional, Rubenstein, J.L.R., additional, Sardar, Debosmita, additional, Sarkar, Anindita, additional, Sawamoto, K., additional, Sharma, Kamal, additional, Shen, Q., additional, Siegenthaler, Julie A., additional, Silver, Debra L., additional, Spassky, N., additional, Stott, S.R.W., additional, Stratmann, Johannes, additional, Subramanian, L., additional, Svaren, John, additional, Szewczyk, Lukasz Mateusz, additional, Temple, S., additional, Thor, Stefan, additional, Tole, Shubha, additional, Valdez, Gregorio, additional, Vaudry, David, additional, Ward, Claire, additional, Wegner, Michael, additional, and Yaghmaeian Salmani, Behzad, additional
- Published
- 2020
- Full Text
- View/download PDF
3. Evolution of enhanced innate immune evasion by the SARS-CoV-2 Alpha variant
- Author
-
Thorne LG, Bouhaddou M, Reuschl AK, Alvarez Z, Polacco B, Pelin A, Batra J, Whelan MVX, Hosmillo M, Fossati A, Ragazzini R, Jungreis I, Ummadi M, Rojc A, Turner J, Bischof ML, Obernier K, Braberg H, Soucheray M, Richards A, Chen KH, Harjai B, Memon D, Hiatt J, Jahun A, Fabius JM, Goodfellow IG, Takeuchi Y, Bonfanti P, Shokat J, Jura N, Klim Verba K, Noursadeghi M, Beltrao P, Kellis M, Swaney DL
- Published
- 2022
- Full Text
- View/download PDF
4. FP2-6 Modelling neonatal intraventricular haemorrhage using organotypic samples from the wall of the lateral ventricle
- Author
-
Dawes, WJ, primary, Obernier, K, additional, and Tisdall, M, additional
- Published
- 2019
- Full Text
- View/download PDF
5. Axonal Control of the Adult Neural Stem Cell Niche
- Author
-
Tong CK, Chen J, Cebrian-Silla A, Mirzadeh Z, Obernier K, Guinto CD, Tecott LH, Garcia-Verdugo JM, Kriegstein A, and Alvarez-Buylla A
- Subjects
nervous system - Abstract
The ventricular-subventricular zone (V-SVZ) is an extensive germinal niche containing neural stem cells (NSCs) in the walls of the lateral ventricles of the adult brain. How the adult brain's neural activity influences the behavior of adult NSCs remains largely unknown. We show that serotonergic (5HT) axons originating from a small group of neurons in the raphe form an extensive plexus on most of the ventricular walls. Electron microscopy revealed intimate contacts between 5HT axons and NSCs (B1) or ependymal cells (E1) and these cells were labeled by a transsynaptic viral tracer injected into the raphe. B1 cells express the 5HT receptors 2C and 5A. Electrophysiology showed that activation of these receptors in B1 cells induced small inward currents. Intraventricular infusion of 5HT2C agonist or antagonist increased or decreased V-SVZ proliferation, respectively. These results indicate that supraependymal 5HT axons directly interact with NSCs to regulate neurogenesis via 5HT2C.
- Published
- 2014
6. Neural Stem Cell Relay from B1 to B2 cells in the adult mouse Ventricular-Subventricular Zone.
- Author
-
Cebrian-Silla A, Assis Nascimento M, Mancia W, Gonzalez-Granero S, Romero-Rodriguez R, Obernier K, Steffen DM, Lim DA, Garcia-Verdugo JM, and Alvarez-Buylla A
- Abstract
Neurogenesis and gliogenesis continue in the Ventricular-Subventricular Zone (V-SVZ) of the adult rodent brain. B1 cells are astroglial cells derived from radial glia that function as primary progenitors or neural stem cells (NSCs) in the V-SVZ. B1 cells, which have a small apical contact with the ventricle, decline in numbers during early postnatal life, yet neurogenesis continues into adulthood. Here we found that a second population of V-SVZ astroglial cells (B2 cells), that do not contact the ventricle, function as NSCs in the adult brain. B2 cell numbers increase postnatally, remain constant in 12-month-old mice and decrease by 18 months. Transcriptomic analysis of ventricular-contacting and non-contacting B cells revealed key molecular differences to distinguish B1 from B2 cells. Transplantation and lineage tracing of B2 cells demonstrate their function as primary progenitors for adult neurogenesis. This study reveals how NSC function is relayed from B1 to B2 progenitors to maintain adult neurogenesis.
- Published
- 2024
- Full Text
- View/download PDF
7. Predicted mechanistic impacts of human protein missense variants.
- Author
-
Jänes J, Müller M, Selvaraj S, Manoel D, Stephenson J, Gonçalves C, Lafita A, Polacco B, Obernier K, Alasoo K, Lemos MC, Krogan N, Martin M, Saraiva LR, Burke D, and Beltrao P
- Abstract
Genome sequencing efforts have led to the discovery of tens of millions of protein missense variants found in the human population with the majority of these having no annotated role and some likely contributing to trait variation and disease. Sequence-based artificial intelligence approaches have become highly accurate at predicting variants that are detrimental to the function of proteins but they do not inform on mechanisms of disruption. Here we combined sequence and structure-based methods to perform proteome-wide prediction of deleterious variants with information on their impact on protein stability, protein-protein interactions and small-molecule binding pockets. AlphaFold2 structures were used to predict approximately 100,000 small-molecule binding pockets and stability changes for over 200 million variants. To inform on protein-protein interfaces we used AlphaFold2 to predict structures for nearly 500,000 protein complexes. We illustrate the value of mechanism-aware variant effect predictions to study the relation between protein stability and abundance and the structural properties of interfaces underlying trans protein quantitative trait loci (pQTLs). We characterised the distribution of mechanistic impacts of protein variants found in patients and experimentally studied example disease linked variants in FGFR1., Competing Interests: Conflict of interests Aleix Lafita is employed by GSK. The Krogan Laboratory has received research support from Vir Biotechnology, F. Hoffmann-La Roche, and Rezo Therapeutics. Nevan Krogan has a financially compensated consulting agreement with Maze Therapeutics. Nevan is the President and is on the Board of Directors of Rezo Therapeutics, and he is a shareholder in Tenaya Therapeutics, Maze Therapeutics, Rezo Therapeutics, and Interline Therapeutics.
- Published
- 2024
- Full Text
- View/download PDF
8. Cell Maps for Artificial Intelligence: AI-Ready Maps of Human Cell Architecture from Disease-Relevant Cell Lines.
- Author
-
Clark T, Mohan J, Schaffer L, Obernier K, Al Manir S, Churas CP, Dailamy A, Doctor Y, Forget A, Hansen JN, Hu M, Lenkiewicz J, Levinson MA, Marquez C, Nourreddine S, Niestroy J, Pratt D, Qian G, Thaker S, Bélisle-Pipon JC, Brandt C, Chen J, Ding Y, Fodeh S, Krogan N, Lundberg E, Mali P, Payne-Foster P, Ratcliffe S, Ravitsky V, Sali A, Schulz W, and Ideker T
- Abstract
This article describes the Cell Maps for Artificial Intelligence (CM4AI) project and its goals, methods, standards, current datasets, software tools , status, and future directions. CM4AI is the Functional Genomics Data Generation Project in the U.S. National Institute of Health's (NIH) Bridge2AI program. Its overarching mission is to produce ethical, AI-ready datasets of cell architecture, inferred from multimodal data collected for human cell lines, to enable transformative biomedical AI research.
- Published
- 2024
- Full Text
- View/download PDF
9. A foundational atlas of autism protein interactions reveals molecular convergence.
- Author
-
Wang B, Vartak R, Zaltsman Y, Naing ZZC, Hennick KM, Polacco BJ, Bashir A, Eckhardt M, Bouhaddou M, Xu J, Sun N, Lasser MC, Zhou Y, McKetney J, Guiley KZ, Chan U, Kaye JA, Chadha N, Cakir M, Gordon M, Khare P, Drake S, Drury V, Burke DF, Gonzalez S, Alkhairy S, Thomas R, Lam S, Morris M, Bader E, Seyler M, Baum T, Krasnoff R, Wang S, Pham P, Arbalaez J, Pratt D, Chag S, Mahmood N, Rolland T, Bourgeron T, Finkbeiner S, Swaney DL, Bandyopadhay S, Ideker T, Beltrao P, Willsey HR, Obernier K, Nowakowski TJ, Hüttenhain R, State MW, Willsey AJ, and Krogan NJ
- Abstract
Translating high-confidence (hc) autism spectrum disorder (ASD) genes into viable treatment targets remains elusive. We constructed a foundational protein-protein interaction (PPI) network in HEK293T cells involving 100 hcASD risk genes, revealing over 1,800 PPIs (87% novel). Interactors, expressed in the human brain and enriched for ASD but not schizophrenia genetic risk, converged on protein complexes involved in neurogenesis, tubulin biology, transcriptional regulation, and chromatin modification. A PPI map of 54 patient-derived missense variants identified differential physical interactions, and we leveraged AlphaFold-Multimer predictions to prioritize direct PPIs and specific variants for interrogation in Xenopus tropicalis and human forebrain organoids. A mutation in the transcription factor FOXP1 led to reconfiguration of DNA binding sites and altered development of deep cortical layer neurons in forebrain organoids. This work offers new insights into molecular mechanisms underlying ASD and describes a powerful platform to develop and test therapeutic strategies for many genetically-defined conditions.
- Published
- 2024
- Full Text
- View/download PDF
10. SARS-CoV-2 variants evolve convergent strategies to remodel the host response.
- Author
-
Bouhaddou M, Reuschl AK, Polacco BJ, Thorne LG, Ummadi MR, Ye C, Rosales R, Pelin A, Batra J, Jang GM, Xu J, Moen JM, Richards AL, Zhou Y, Harjai B, Stevenson E, Rojc A, Ragazzini R, Whelan MVX, Furnon W, De Lorenzo G, Cowton V, Syed AM, Ciling A, Deutsch N, Pirak D, Dowgier G, Mesner D, Turner JL, McGovern BL, Rodriguez ML, Leiva-Rebollo R, Dunham AS, Zhong X, Eckhardt M, Fossati A, Liotta NF, Kehrer T, Cupic A, Rutkowska M, Mena I, Aslam S, Hoffert A, Foussard H, Olwal CO, Huang W, Zwaka T, Pham J, Lyons M, Donohue L, Griffin A, Nugent R, Holden K, Deans R, Aviles P, Lopez-Martin JA, Jimeno JM, Obernier K, Fabius JM, Soucheray M, Hüttenhain R, Jungreis I, Kellis M, Echeverria I, Verba K, Bonfanti P, Beltrao P, Sharan R, Doudna JA, Martinez-Sobrido L, Patel AH, Palmarini M, Miorin L, White K, Swaney DL, Garcia-Sastre A, Jolly C, Zuliani-Alvarez L, Towers GJ, and Krogan NJ
- Subjects
- Humans, Immunity, Innate genetics, Pandemics, COVID-19 virology, SARS-CoV-2 genetics
- Abstract
SARS-CoV-2 variants of concern (VOCs) emerged during the COVID-19 pandemic. Here, we used unbiased systems approaches to study the host-selective forces driving VOC evolution. We discovered that VOCs evolved convergent strategies to remodel the host by modulating viral RNA and protein levels, altering viral and host protein phosphorylation, and rewiring virus-host protein-protein interactions. Integrative computational analyses revealed that although Alpha, Beta, Gamma, and Delta ultimately converged to suppress interferon-stimulated genes (ISGs), Omicron BA.1 did not. ISG suppression correlated with the expression of viral innate immune antagonist proteins, including Orf6, N, and Orf9b, which we mapped to specific mutations. Later Omicron subvariants BA.4 and BA.5 more potently suppressed innate immunity than early subvariant BA.1, which correlated with Orf6 levels, although muted in BA.4 by a mutation that disrupts the Orf6-nuclear pore interaction. Our findings suggest that SARS-CoV-2 convergent evolution overcame human adaptive and innate immune barriers, laying the groundwork to tackle future pandemics., Competing Interests: Declaration of interests The Krogan Laboratory received research support from Vir Biotechnology, F. Hoffmann-La Roche, and Rezo Therapeutics. N.J.K. has previously held financially compensated consulting agreements with the Icahn School of Medicine at Mount Sinai, New York and Twist Bioscience Corp. He currently has financially compensated consulting agreements with Maze Therapeutics, Interline Therapeutics, Rezo Therapeutics, and GEn1E Lifesciences, Inc. He is on the Board of Directors of Rezo Therapeutics and is a shareholder in Tenaya Therapeutics, Maze Therapeutics, Rezo Therapeutics, and Interline Therapeutics. The A.G.-S. laboratory received research support from Pfizer, Senhwa Biosciences, Kenall Manufacturing, Blade Therapeutics, Avimex, Johnson & Johnson, Dynavax, 7Hills Pharma, PharmaMar, ImmunityBio, Accurius, Nanocomposix, Hexamer, N-fold LLC, Model Medicines, Atea Pharma, Applied Biological Laboratories, and Merck. A.G.-S. has consulting agreements for the following companies involving cash and/or stock: Castlevax, Amovir, Vivaldi Biosciences, Contrafect, 7Hills Pharma, Avimex, Vaxalto, Pagoda, Accurius, Esperovax, Farmak, Applied Biological Laboratories, PharmaMar, Paratus, CureLab Oncology, CureLab Veterinary, Synairgen, and Pfizer. A.G.-S. has been an invited speaker in meeting events organized by Seqirus, Janssen, Abbott, and Astrazeneca. A.G.-S. is inventor on patents and patent applications on the use of antivirals and vaccines for the treatment and prevention of virus infections and cancer, owned by the Icahn School of Medicine at Mount Sinai, New York. M.B. is a financially compensated scientific advisor for GEn1E Lifesciences. C.Y. and L.M.-S. are co-inventors on a patent application directed to reverse genetics approaches to generate recombinant SARS-CoV-2. The Regents of the University of California have patents issued and pending for CRISPR technologies on which J.A.D. is an inventor. J.A.D. is a co-founder of Caribou Biosciences, Editas Medicine, Scribe Therapeutics, Intellia Therapeutics, and Mammoth Biosciences. J.A.D. is a scientific advisory board member of Vertex, Caribou Biosciences, Intellia Therapeutics, Scribe Therapeutics, Mammoth Biosciences, Algen Biotechnologies, Felix Biosciences, The Column Group, and Inari. J.A.D. is Chief Science Advisor to Sixth Street, a Director at Johnson & Johnson, Altos, and Tempus, and has research projects sponsored by Apple Tree Partners and Roche. John Pham, Molly Lyons, Laura Donahue, Aliesha Griffin, Rebecca Nugent, Kevin Holden, and Robert Deans are employees and shareholders of Synthego Corporation. D.L.S. has financially compensated consulting agreements with Maze Therapeutics and Rezo Therapeutics. P.A., J.A.L.-M., and J.M.J. are employees and shareholders of Pharma Mar, S.A. (Madrid, Spain). J.A.L.-M. is a co-inventor of a patent for Plitidepsin (WO2008135793A1). J.M.J. holds stocks of Pangaea Oncology, has a non-remunerated role in the Scientific Advisory Board, and holds stocks of Promontory Therapeutics, and is a co-inventor of two patents for Plitidepsin (WO99-42125)., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Publisher Correction: Evolution of enhanced innate immune evasion by SARS-CoV-2.
- Author
-
Thorne LG, Bouhaddou M, Reuschl AK, Zuliani-Alvarez L, Polacco B, Pelin A, Batra J, Whelan MVX, Hosmillo M, Fossati A, Ragazzini R, Jungreis I, Ummadi M, Rojc A, Turner J, Bischof ML, Obernier K, Braberg H, Soucheray M, Richards A, Chen KH, Harjai B, Memon D, Hiatt J, Rosales R, McGovern BL, Jahun A, Fabius JM, White K, Goodfellow IG, Takeuchi Y, Bonfanti P, Shokat K, Jura N, Verba K, Noursadeghi M, Beltrao P, Kellis M, Swaney DL, García-Sastre A, Jolly C, Towers GJ, and Krogan NJ
- Published
- 2022
- Full Text
- View/download PDF
12. Nests of dividing neuroblasts sustain interneuron production for the developing human brain.
- Author
-
Paredes MF, Mora C, Flores-Ramirez Q, Cebrian-Silla A, Del Dosso A, Larimer P, Chen J, Kang G, Gonzalez Granero S, Garcia E, Chu J, Delgado R, Cotter JA, Tang V, Spatazza J, Obernier K, Ferrer Lozano J, Vento M, Scott J, Studholme C, Nowakowski TJ, Kriegstein AR, Oldham MC, Hasenstaub A, Garcia-Verdugo JM, Alvarez-Buylla A, and Huang EJ
- Subjects
- Animals, Animals, Newborn, Cell Movement, Cell Proliferation, Cerebral Cortex cytology, Cerebral Cortex embryology, Cerebral Cortex growth & development, GABAergic Neurons cytology, GABAergic Neurons physiology, Gene Expression Profiling, Gestational Age, Humans, Interneurons cytology, Median Eminence cytology, Median Eminence growth & development, Mice, Neural Stem Cells transplantation, Prosencephalon embryology, Prosencephalon growth & development, Transplantation, Heterologous, Interneurons physiology, Median Eminence embryology, Neural Stem Cells physiology, Neurogenesis, Prosencephalon cytology
- Abstract
The human cortex contains inhibitory interneurons derived from the medial ganglionic eminence (MGE), a germinal zone in the embryonic ventral forebrain. How this germinal zone generates sufficient interneurons for the human brain remains unclear. We found that the human MGE (hMGE) contains nests of proliferative neuroblasts with ultrastructural and transcriptomic features that distinguish them from other progenitors in the hMGE. When dissociated hMGE cells are transplanted into the neonatal mouse brain, they reform into nests containing proliferating neuroblasts that generate young neurons that migrate extensively into the mouse forebrain and mature into different subtypes of functional interneurons. Together, these results indicate that the nest organization and sustained proliferation of neuroblasts in the hMGE provide a mechanism for the extended production of interneurons for the human forebrain.
- Published
- 2022
- Full Text
- View/download PDF
13. Preclinical and randomized phase I studies of plitidepsin in adults hospitalized with COVID-19.
- Author
-
Varona JF, Landete P, Lopez-Martin JA, Estrada V, Paredes R, Guisado-Vasco P, Fernandez de Orueta L, Torralba M, Fortun J, Vates R, Barberan J, Clotet B, Ancochea J, Carnevali D, Cabello N, Porras L, Gijon P, Monereo A, Abad D, Zuñiga S, Sola I, Rodon J, Vergara-Alert J, Izquierdo-Useros N, Fudio S, Pontes MJ, de Rivas B, Giron de Velasco P, Nieto A, Gomez J, Aviles P, Lubomirov R, Belgrano A, Sopesen B, White KM, Rosales R, Yildiz S, Reuschl AK, Thorne LG, Jolly C, Towers GJ, Zuliani-Alvarez L, Bouhaddou M, Obernier K, McGovern BL, Rodriguez ML, Enjuanes L, Fernandez-Sousa JM, Krogan NJ, Jimeno JM, and Garcia-Sastre A
- Subjects
- Adult, Aged, COVID-19 virology, Cell Line, Tumor, Depsipeptides adverse effects, Depsipeptides pharmacology, Drug Evaluation, Preclinical methods, Female, Humans, Kaplan-Meier Estimate, Length of Stay statistics & numerical data, Male, Middle Aged, Neutropenia chemically induced, Peptides, Cyclic adverse effects, Peptides, Cyclic pharmacology, SARS-CoV-2 physiology, Treatment Outcome, Viral Load drug effects, Depsipeptides therapeutic use, Hospitalization statistics & numerical data, Peptides, Cyclic therapeutic use, SARS-CoV-2 drug effects, COVID-19 Drug Treatment
- Abstract
Plitidepsin, a marine-derived cyclic-peptide, inhibits SARS-CoV-2 replication at nanomolar concentrations by targeting the host protein eukaryotic translation elongation factor 1A. Here, we show that plitidepsin distributes preferentially to lung over plasma, with similar potency against across several SARS-CoV-2 variants in preclinical studies. Simultaneously, in this randomized, parallel, open-label, proof-of-concept study (NCT04382066) conducted in 10 Spanish hospitals between May and November 2020, 46 adult hospitalized patients with confirmed SARS-CoV-2 infection received either 1.5 mg (n = 15), 2.0 mg (n = 16), or 2.5 mg (n = 15) plitidepsin once daily for 3 d. The primary objective was safety; viral load kinetics, mortality, need for increased respiratory support, and dose selection were secondary end points. One patient withdrew consent before starting procedures; 45 initiated treatment; one withdrew because of hypersensitivity. Two Grade 3 treatment-related adverse events were observed (hypersensitivity and diarrhea). Treatment-related adverse events affecting more than 5% of patients were nausea (42.2%), vomiting (15.6%), and diarrhea (6.7%). Mean viral load reductions from baseline were 1.35, 2.35, 3.25, and 3.85 log
10 at days 4, 7, 15, and 31. Nonmechanical invasive ventilation was required in 8 of 44 evaluable patients (16.0%); six patients required intensive care support (13.6%), and three patients (6.7%) died (COVID-19-related). Plitidepsin has a favorable safety profile in patients with COVID-19., (© 2022 Varona et al.)- Published
- 2022
- Full Text
- View/download PDF
14. Target Discovery for Host-Directed Antiviral Therapies: Application of Proteomics Approaches.
- Author
-
Cakir M, Obernier K, Forget A, and Krogan NJ
- Abstract
Current epidemics, such as AIDS or flu, and the emergence of new threatening pathogens, such as the one causing the current coronavirus disease 2019 (COVID-19) pandemic, represent major global health challenges. While vaccination is an important part of the arsenal to counter the spread of viral diseases, it presents limitations and needs to be complemented by efficient therapeutic solutions. Intricate knowledge of host-pathogen interactions is a powerful tool to identify host-dependent vulnerabilities that can be exploited to dampen viral replication. Such host-directed antiviral therapies are promising and are less prone to the development of drug-resistant viral strains. Here, we first describe proteomics-based strategies that allow the rapid characterization of host-pathogen interactions. We then discuss how such data can be exploited to help prioritize compounds with potential host-directed antiviral activity that can be tested in preclinical models.
- Published
- 2021
- Full Text
- View/download PDF
15. A protein network map of head and neck cancer reveals PIK3CA mutant drug sensitivity.
- Author
-
Swaney DL, Ramms DJ, Wang Z, Park J, Goto Y, Soucheray M, Bhola N, Kim K, Zheng F, Zeng Y, McGregor M, Herrington KA, O'Keefe R, Jin N, VanLandingham NK, Foussard H, Von Dollen J, Bouhaddou M, Jimenez-Morales D, Obernier K, Kreisberg JF, Kim M, Johnson DE, Jura N, Grandis JR, Gutkind JS, Ideker T, and Krogan NJ
- Subjects
- Animals, Carcinoma, Squamous Cell genetics, Cell Line, Tumor, Cell Movement, Female, Head and Neck Neoplasms genetics, Humans, Intracellular Signaling Peptides and Proteins metabolism, Mice, Mice, Nude, Microfilament Proteins metabolism, Mutation, Receptor, Fibroblast Growth Factor, Type 3 metabolism, Xenograft Model Antitumor Assays, Carcinoma, Squamous Cell metabolism, Class I Phosphatidylinositol 3-Kinases genetics, Class I Phosphatidylinositol 3-Kinases metabolism, Drug Resistance, Neoplasm genetics, Head and Neck Neoplasms metabolism, Protein Interaction Maps
- Abstract
We outline a framework for elucidating tumor genetic complexity through multidimensional protein-protein interaction maps and apply it to enhancing our understanding of head and neck squamous cell carcinoma. This network uncovers 771 interactions from cancer and noncancerous cell states, including WT and mutant protein isoforms. Prioritization of cancer-enriched interactions reveals a previously unidentified association of the fibroblast growth factor receptor tyrosine kinase 3 with Daple, a guanine-nucleotide exchange factor, resulting in activation of Gαi- and p21-activated protein kinase 1/2 to promote cancer cell migration. Additionally, we observe mutation-enriched interactions between the human epidermal growth factor receptor 3 (HER3) receptor tyrosine kinase and PIK3CA (the alpha catalytic subunit of phosphatidylinositol 3-kinase) that can inform the response to HER3 inhibition in vivo. We anticipate that the application of this framework will be valuable for translating genetic alterations into a molecular and clinical understanding of the underlying biology of many disease areas.
- Published
- 2021
- Full Text
- View/download PDF
16. Single-cell analysis of the ventricular-subventricular zone reveals signatures of dorsal and ventral adult neurogenesis.
- Author
-
Cebrian-Silla A, Nascimento MA, Redmond SA, Mansky B, Wu D, Obernier K, Romero Rodriguez R, Gonzalez-Granero S, García-Verdugo JM, Lim DA, and Álvarez-Buylla A
- Subjects
- Animals, Female, Male, Mice, Mice, Transgenic, Microdissection, Neural Stem Cells chemistry, Neural Stem Cells cytology, Single-Cell Analysis, Lateral Ventricles cytology, Neural Stem Cells metabolism, Neurogenesis genetics, Transcriptome genetics
- Abstract
The ventricular-subventricular zone (V-SVZ), on the walls of the lateral ventricles, harbors the largest neurogenic niche in the adult mouse brain. Previous work has shown that neural stem/progenitor cells (NSPCs) in different locations within the V-SVZ produce different subtypes of new neurons for the olfactory bulb. The molecular signatures that underlie this regional heterogeneity remain largely unknown. Here, we present a single-cell RNA-sequencing dataset of the adult mouse V-SVZ revealing two populations of NSPCs that reside in largely non-overlapping domains in either the dorsal or ventral V-SVZ. These regional differences in gene expression were further validated using a single-nucleus RNA-sequencing reference dataset of regionally microdissected domains of the V-SVZ and by immunocytochemistry and RNAscope localization. We also identify two subpopulations of young neurons that have gene expression profiles consistent with a dorsal or ventral origin. Interestingly, a subset of genes are dynamically expressed, but maintained, in the ventral or dorsal lineages. The study provides novel markers and territories to understand the region-specific regulation of adult neurogenesis., Competing Interests: AC, MN, SR, BM, DW, KO, RR, SG, JG, DL No competing interests declared, AÁ Cofounder and on the Scientific Advisory Board of Neurona Therapeutics., (© 2021, Cebrian-Silla et al.)
- Published
- 2021
- Full Text
- View/download PDF
17. Evolution of enhanced innate immune evasion by the SARS-CoV-2 B.1.1.7 UK variant.
- Author
-
Thorne LG, Bouhaddou M, Reuschl AK, Zuliani-Alvarez L, Polacco B, Pelin A, Batra J, Whelan MVX, Ummadi M, Rojc A, Turner J, Obernier K, Braberg H, Soucheray M, Richards A, Chen KH, Harjai B, Memon D, Hosmillo M, Hiatt J, Jahun A, Goodfellow IG, Fabius JM, Shokat K, Jura N, Verba K, Noursadeghi M, Beltrao P, Swaney DL, Garcia-Sastre A, Jolly C, Towers GJ, and Krogan NJ
- Abstract
Emergence of SARS-CoV-2 variants, including the globally successful B.1.1.7 lineage, suggests viral adaptations to host selective pressures resulting in more efficient transmission. Although much effort has focused on Spike adaptation for viral entry and adaptive immune escape, B.1.1.7 mutations outside Spike likely contribute to enhance transmission. Here we used unbiased abundance proteomics, phosphoproteomics, mRNA sequencing and viral replication assays to show that B.1.1.7 isolates more effectively suppress host innate immune responses in airway epithelial cells. We found that B.1.1.7 isolates have dramatically increased subgenomic RNA and protein levels of Orf9b and Orf6, both known innate immune antagonists. Expression of Orf9b alone suppressed the innate immune response through interaction with TOM70, a mitochondrial protein required for RNA sensing adaptor MAVS activation, and Orf9b binding and activity was regulated via phosphorylation. We conclude that B.1.1.7 has evolved beyond the Spike coding region to more effectively antagonise host innate immune responses through upregulation of specific subgenomic RNA synthesis and increased protein expression of key innate immune antagonists. We propose that more effective innate immune antagonism increases the likelihood of successful B.1.1.7 transmission, and may increase in vivo replication and duration of infection.
- Published
- 2021
- Full Text
- View/download PDF
18. Plitidepsin has a positive therapeutic index in adult patients with COVID-19 requiring hospitalization.
- Author
-
Varona JF, Landete P, Lopez-Martin JA, Estrada V, Paredes R, Guisado-Vasco P, de Orueta LF, Torralba M, Fortún J, Vates R, Barberán J, Clotet B, Ancochea J, Carnevali D, Cabello N, Porras L, Gijón P, Monereo A, Abad D, Zúñiga S, Sola I, Rodon J, Izquierdo-Useros N, Fudio S, Pontes MJ, de Rivas B, Girón de Velasco P, Sopesén B, Nieto A, Gómez J, Avilés P, Lubomirov R, White KM, Rosales R, Yildiz S, Reuschl AK, Thorne LG, Jolly C, Towers GJ, Zuliani-Alvarez L, Bouhaddou M, Obernier K, Enjuanes L, Fernández-Sousa JM, Krogan NJ, Jimeno JM, and García-Sastre A
- Abstract
Plitidepsin is a marine-derived cyclic-peptide that inhibits SARS-CoV-2 replication at low nanomolar concentrations by the targeting of host protein eEF1A (eukaryotic translation-elongation-factor-1A). We evaluated a model of intervention with plitidepsin in hospitalized COVID-19 adult patients where three doses were assessed (1.5, 2 and 2.5 mg/day for 3 days, as a 90-minute intravenous infusion) in 45 patients (15 per dose-cohort). Treatment was well tolerated, with only two Grade 3 treatment-related adverse events observed (hypersensitivity and diarrhea). The discharge rates by Days 8 and 15 were 56.8% and 81.8%, respectively, with data sustaining dose-effect. A mean 4.2 log10 viral load reduction was attained by Day 15. Improvement in inflammation markers was also noted in a seemingly dose-dependent manner. These results suggest that plitidepsin impacts the outcome of patients with COVID-19., One-Sentence Summary: Plitidepsin, an inhibitor of SARS-Cov-2 in vitro , is safe and positively influences the outcome of patients hospitalized with COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
19. Plitidepsin has potent preclinical efficacy against SARS-CoV-2 by targeting the host protein eEF1A.
- Author
-
White KM, Rosales R, Yildiz S, Kehrer T, Miorin L, Moreno E, Jangra S, Uccellini MB, Rathnasinghe R, Coughlan L, Martinez-Romero C, Batra J, Rojc A, Bouhaddou M, Fabius JM, Obernier K, Dejosez M, Guillén MJ, Losada A, Avilés P, Schotsaert M, Zwaka T, Vignuzzi M, Shokat KM, Krogan NJ, and García-Sastre A
- Subjects
- Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate pharmacology, Adenosine Monophosphate therapeutic use, Alanine analogs & derivatives, Alanine pharmacology, Alanine therapeutic use, Animals, Antiviral Agents therapeutic use, COVID-19 prevention & control, COVID-19 virology, Coronavirus Nucleocapsid Proteins biosynthesis, Coronavirus Nucleocapsid Proteins genetics, Depsipeptides administration & dosage, Depsipeptides therapeutic use, Drug Evaluation, Preclinical, Female, HEK293 Cells, Humans, Lung virology, Mice, Inbred C57BL, Mutation, Peptides, Cyclic, Phosphoproteins biosynthesis, Phosphoproteins genetics, RNA, Viral biosynthesis, RNA, Viral genetics, SARS-CoV-2 genetics, SARS-CoV-2 physiology, Virus Replication drug effects, Mice, Antiviral Agents pharmacology, Depsipeptides pharmacology, Peptide Elongation Factor 1 antagonists & inhibitors, SARS-CoV-2 drug effects, COVID-19 Drug Treatment
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral proteins interact with the eukaryotic translation machinery, and inhibitors of translation have potent antiviral effects. We found that the drug plitidepsin (aplidin), which has limited clinical approval, possesses antiviral activity (90% inhibitory concentration = 0.88 nM) that is more potent than remdesivir against SARS-CoV-2 in vitro by a factor of 27.5, with limited toxicity in cell culture. Through the use of a drug-resistant mutant, we show that the antiviral activity of plitidepsin against SARS-CoV-2 is mediated through inhibition of the known target eEF1A (eukaryotic translation elongation factor 1A). We demonstrate the in vivo efficacy of plitidepsin treatment in two mouse models of SARS-CoV-2 infection with a reduction of viral replication in the lungs by two orders of magnitude using prophylactic treatment. Our results indicate that plitidepsin is a promising therapeutic candidate for COVID-19., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
20. Host-directed therapies against early-lineage SARS-CoV-2 retain efficacy against B.1.1.7 variant.
- Author
-
Reuschl AK, Thorne LG, Zuliani-Alvarez L, Bouhaddou M, Obernier K, Hiatt J, Soucheray M, Turner J, Fabius JM, Nguyen GT, Swaney DL, Rosales R, White KM, Avilés P, Kirby IT, Melnyk JE, Shi Y, Zhang Z, Shokat KM, García-Sastre A, Jolly C, Towers GJ, and Krogan NJ
- Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in millions of deaths worldwide and massive societal and economic burden. Recently, a new variant of SARS-CoV-2, known as B.1.1.7, was first detected in the United Kingdom and is spreading in several other countries, heightening public health concern and raising questions as to the resulting effectiveness of vaccines and therapeutic interventions. We and others previously identified host-directed therapies with antiviral efficacy against SARS-CoV-2 infection. Less prone to the development of therapy resistance, host-directed drugs represent promising therapeutic options to combat emerging viral variants as host genes possess a lower propensity to mutate compared to viral genes. Here, in the first study of the full-length B.1.1.7 variant virus , we find two host-directed drugs, plitidepsin (aplidin; inhibits translation elongation factor eEF1A) and ralimetinib (inhibits p38 MAP kinase cascade), as well as remdesivir, to possess similar antiviral activity against both the early-lineage SARS-CoV-2 and the B.1.1.7 variant, evaluated in both human gastrointestinal and lung epithelial cell lines. We find that plitidepsin is over an order of magnitude more potent than remdesivir against both viruses. These results highlight the importance of continued development of host-directed therapeutics to combat current and future coronavirus variant outbreaks., Competing Interests: Competing Interests The García-Sastre Laboratory has received research support from Pfizer, Senhwa Biosciences and 7Hills Pharma. Adolfo García-Sastre has consulting agreements for the following companies involving cash and/or stock: Vivaldi Biosciences, Contrafect, 7Hills Pharma, Avimex, Vaxalto, Accurius and Esperovax. The Krogan Laboratory receives funding from Roche and VIR and Nevan Krogan has consulting agreements with Maze Therapeutics and Interline Therapeutics. 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, Revolution Medicines, Type6 Therapeutics, Venthera, Wellspring Biosciences (Araxes Pharma), Turning Point Therapeutics, Ikena, Nextech. Pablo Avilés is an employee and shareholder of PharmaMar, SA (Madrid, Spain).
- Published
- 2021
- Full Text
- View/download PDF
21. Comparative host-coronavirus protein interaction networks reveal pan-viral disease mechanisms.
- Author
-
Gordon DE, Hiatt J, Bouhaddou M, Rezelj VV, Ulferts S, Braberg H, Jureka AS, Obernier K, Guo JZ, Batra J, Kaake RM, Weckstein AR, Owens TW, Gupta M, Pourmal S, Titus EW, Cakir M, Soucheray M, McGregor M, Cakir Z, Jang G, O'Meara MJ, Tummino TA, Zhang Z, Foussard H, Rojc A, Zhou Y, Kuchenov D, Hüttenhain R, Xu J, Eckhardt M, Swaney DL, Fabius JM, Ummadi M, Tutuncuoglu B, Rathore U, Modak M, Haas P, Haas KM, Naing ZZC, Pulido EH, Shi Y, Barrio-Hernandez I, Memon D, Petsalaki E, Dunham A, Marrero MC, Burke D, Koh C, Vallet T, Silvas JA, Azumaya CM, Billesbølle C, Brilot AF, Campbell MG, Diallo A, Dickinson MS, Diwanji D, Herrera N, Hoppe N, Kratochvil HT, Liu Y, Merz GE, Moritz M, Nguyen HC, Nowotny C, Puchades C, Rizo AN, Schulze-Gahmen U, Smith AM, Sun M, Young ID, Zhao J, Asarnow D, Biel J, Bowen A, Braxton JR, Chen J, Chio CM, Chio US, Deshpande I, Doan L, Faust B, Flores S, Jin M, Kim K, Lam VL, Li F, Li J, Li YL, Li Y, Liu X, Lo M, Lopez KE, Melo AA, Moss FR 3rd, Nguyen P, Paulino J, Pawar KI, Peters JK, Pospiech TH Jr, Safari M, Sangwan S, Schaefer K, Thomas PV, Thwin AC, Trenker R, Tse E, Tsui TKM, Wang F, Whitis N, Yu Z, Zhang K, Zhang Y, Zhou F, Saltzberg D, Hodder AJ, Shun-Shion AS, Williams DM, White KM, Rosales R, Kehrer T, Miorin L, Moreno E, Patel AH, Rihn S, Khalid MM, Vallejo-Gracia A, Fozouni P, Simoneau CR, Roth TL, Wu D, Karim MA, Ghoussaini M, Dunham I, Berardi F, Weigang S, Chazal M, Park J, Logue J, McGrath M, Weston S, Haupt R, Hastie CJ, Elliott M, Brown F, Burness KA, Reid E, Dorward M, Johnson C, Wilkinson SG, Geyer A, Giesel DM, Baillie C, Raggett S, Leech H, Toth R, Goodman N, Keough KC, Lind AL, Klesh RJ, Hemphill KR, Carlson-Stevermer J, Oki J, Holden K, Maures T, Pollard KS, Sali A, Agard DA, Cheng Y, Fraser JS, Frost A, Jura N, Kortemme T, Manglik A, Southworth DR, Stroud RM, Alessi DR, Davies P, Frieman MB, Ideker T, Abate C, Jouvenet N, Kochs G, Shoichet B, Ott M, Palmarini M, Shokat KM, García-Sastre A, Rassen JA, Grosse R, Rosenberg OS, Verba KA, Basler CF, Vignuzzi M, Peden AA, Beltrao P, and Krogan NJ
- Subjects
- Conserved Sequence, Coronavirus Nucleocapsid Proteins genetics, Cryoelectron Microscopy, Humans, Mitochondrial Membrane Transport Proteins genetics, Mitochondrial Precursor Protein Import Complex Proteins, Phosphoproteins genetics, Phosphoproteins metabolism, Protein Conformation, COVID-19 metabolism, Coronavirus Nucleocapsid Proteins metabolism, Host Microbial Interactions, Mitochondrial Membrane Transport Proteins metabolism, Protein Interaction Maps, Severe acute respiratory syndrome-related coronavirus metabolism, SARS-CoV-2 metabolism, Severe Acute Respiratory Syndrome metabolism
- Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a grave threat to public health and the global economy. SARS-CoV-2 is closely related to the more lethal but less transmissible coronaviruses SARS-CoV-1 and Middle East respiratory syndrome coronavirus (MERS-CoV). Here, we have carried out comparative viral-human protein-protein interaction and viral protein localization analyses for all three viruses. Subsequent functional genetic screening identified host factors that functionally impinge on coronavirus proliferation, including Tom70, a mitochondrial chaperone protein that interacts with both SARS-CoV-1 and SARS-CoV-2 ORF9b, an interaction we structurally characterized using cryo-electron microscopy. Combining genetically validated host factors with both COVID-19 patient genetic data and medical billing records identified molecular mechanisms and potential drug treatments that merit further molecular and clinical study., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
22. The Global Phosphorylation Landscape of SARS-CoV-2 Infection.
- Author
-
Bouhaddou M, Memon D, Meyer B, White KM, Rezelj VV, Correa Marrero M, Polacco BJ, Melnyk JE, Ulferts S, Kaake RM, Batra J, Richards AL, Stevenson E, Gordon DE, Rojc A, Obernier K, Fabius JM, Soucheray M, Miorin L, Moreno E, Koh C, Tran QD, Hardy A, Robinot R, Vallet T, Nilsson-Payant BE, Hernandez-Armenta C, Dunham A, Weigang S, Knerr J, Modak M, Quintero D, Zhou Y, Dugourd A, Valdeolivas A, Patil T, Li Q, Hüttenhain R, Cakir M, Muralidharan M, Kim M, Jang G, Tutuncuoglu B, Hiatt J, Guo JZ, Xu J, Bouhaddou S, Mathy CJP, Gaulton A, Manners EJ, Félix E, Shi Y, Goff M, Lim JK, McBride T, O'Neal MC, Cai Y, Chang JCJ, Broadhurst DJ, Klippsten S, De Wit E, Leach AR, Kortemme T, Shoichet B, Ott M, Saez-Rodriguez J, tenOever BR, Mullins RD, Fischer ER, Kochs G, Grosse R, García-Sastre A, Vignuzzi M, Johnson JR, Shokat KM, Swaney DL, Beltrao P, and Krogan NJ
- Subjects
- A549 Cells, Angiotensin-Converting Enzyme 2, Animals, Antiviral Agents pharmacology, COVID-19, Caco-2 Cells, Casein Kinase II antagonists & inhibitors, Casein Kinase II metabolism, Chlorocebus aethiops, Coronavirus Infections virology, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases metabolism, HEK293 Cells, Host-Pathogen Interactions, Humans, Pandemics, Peptidyl-Dipeptidase A genetics, Peptidyl-Dipeptidase A metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors pharmacology, Phosphorylation, Pneumonia, Viral virology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases metabolism, SARS-CoV-2, Spike Glycoprotein, Coronavirus metabolism, Vero Cells, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, p38 Mitogen-Activated Protein Kinases metabolism, Axl Receptor Tyrosine Kinase, Betacoronavirus metabolism, Coronavirus Infections metabolism, Drug Evaluation, Preclinical methods, Pneumonia, Viral metabolism, Proteomics methods
- Abstract
The causative agent of the coronavirus disease 2019 (COVID-19) pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected millions and killed hundreds of thousands of people worldwide, highlighting an urgent need to develop antiviral therapies. Here we present a quantitative mass spectrometry-based phosphoproteomics survey of SARS-CoV-2 infection in Vero E6 cells, revealing dramatic rewiring of phosphorylation on host and viral proteins. SARS-CoV-2 infection promoted casein kinase II (CK2) and p38 MAPK activation, production of diverse cytokines, and shutdown of mitotic kinases, resulting in cell cycle arrest. Infection also stimulated a marked induction of CK2-containing filopodial protrusions possessing budding viral particles. Eighty-seven drugs and compounds were identified by mapping global phosphorylation profiles to dysregulated kinases and pathways. We found pharmacologic inhibition of the p38, CK2, CDK, AXL, and PIKFYVE kinases to possess antiviral efficacy, representing potential COVID-19 therapies., Competing Interests: Declaration of Interests The Krogan laboratory has received research support from Vir Biotechnology and F. Hoffmann-La Roche. K.M.S. has consulting agreements for the following companies involving cash and/or stock compensation: Black Diamond Therapeutics, BridGene Biosciences, Denali Therapeutics, Dice Molecules, eFFECTOR Therapeutics (zotatifin and tomivosertib), Erasca, Genentech/Roche, Janssen Pharmaceuticals, Kumquat Biosciences, Kura Oncology, Merck, Mitokinin, Petra Pharma, Qulab Inc. Revolution Medicines (WDB002), Type6 Therapeutics, Venthera, and Wellspring Biosciences (Araxes Pharma)., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing.
- Author
-
Gordon DE, Jang GM, Bouhaddou M, Xu J, Obernier K, White KM, O'Meara MJ, Rezelj VV, Guo JZ, Swaney DL, Tummino TA, Hüttenhain 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, Meyer B, Roesch F, Vallet T, Mac Kain A, Miorin L, Moreno E, Naing ZZC, Zhou Y, Peng S, Shi Y, Zhang Z, Shen W, Kirby IT, Melnyk JE, Chorba JS, Lou K, Dai SA, Barrio-Hernandez I, Memon D, Hernandez-Armenta C, Lyu J, Mathy CJP, Perica T, Pilla KB, Ganesan SJ, Saltzberg DJ, Rakesh R, Liu X, Rosenthal SB, Calviello L, Venkataramanan S, Liboy-Lugo J, Lin Y, Huang XP, Liu Y, Wankowicz SA, Bohn M, Safari M, Ugur FS, Koh C, Savar NS, Tran QD, Shengjuler D, Fletcher SJ, O'Neal MC, Cai Y, Chang JCJ, Broadhurst DJ, Klippsten S, Sharp PP, Wenzell NA, Kuzuoglu-Ozturk D, Wang HY, Trenker R, Young JM, Cavero DA, Hiatt J, Roth TL, Rathore U, Subramanian A, Noack J, Hubert M, Stroud RM, Frankel AD, Rosenberg OS, Verba KA, Agard DA, Ott M, Emerman M, Jura N, von Zastrow M, Verdin E, Ashworth A, Schwartz O, d'Enfert C, Mukherjee S, Jacobson M, Malik HS, Fujimori DG, Ideker T, Craik CS, Floor SN, Fraser JS, Gross JD, Sali A, Roth BL, Ruggero D, Taunton J, Kortemme T, Beltrao P, Vignuzzi M, García-Sastre A, Shokat KM, Shoichet BK, and Krogan NJ
- Subjects
- Animals, Antiviral Agents classification, Antiviral Agents pharmacology, Betacoronavirus genetics, Betacoronavirus metabolism, Betacoronavirus pathogenicity, COVID-19, Chlorocebus aethiops, Cloning, Molecular, Coronavirus Infections immunology, Coronavirus Infections virology, Drug Evaluation, Preclinical, HEK293 Cells, Host-Pathogen Interactions drug effects, Humans, Immunity, Innate, Mass Spectrometry, Pandemics, Pneumonia, Viral immunology, Pneumonia, Viral virology, Protein Binding, Protein Biosynthesis drug effects, Protein Domains, Protein Interaction Mapping, Receptors, sigma metabolism, SARS-CoV-2, SKP Cullin F-Box Protein Ligases metabolism, Vero Cells, Viral Proteins genetics, COVID-19 Drug Treatment, Betacoronavirus drug effects, Coronavirus Infections drug therapy, Coronavirus Infections metabolism, Drug Repositioning, Molecular Targeted Therapy, Pneumonia, Viral drug therapy, Pneumonia, Viral metabolism, Protein Interaction Maps, Viral Proteins metabolism
- Abstract
A newly described coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the causative agent of coronavirus disease 2019 (COVID-19), has infected over 2.3 million people, led to the death of more than 160,000 individuals and caused worldwide social and economic disruption
1,2 . There are no antiviral drugs with proven clinical efficacy for the treatment of COVID-19, nor are there any vaccines that prevent infection with SARS-CoV-2, and efforts to develop drugs and vaccines are hampered by the limited knowledge of the molecular details of how SARS-CoV-2 infects cells. Here we cloned, tagged and expressed 26 of the 29 SARS-CoV-2 proteins in human cells and identified the human proteins that physically associated with each of the SARS-CoV-2 proteins using affinity-purification mass spectrometry, identifying 332 high-confidence protein-protein interactions between SARS-CoV-2 and human proteins. Among these, we identify 66 druggable human proteins or host factors targeted by 69 compounds (of which, 29 drugs are approved by the US Food and Drug Administration, 12 are in clinical trials and 28 are preclinical compounds). We screened a subset of these in multiple viral assays and found two sets of pharmacological agents that displayed antiviral activity: inhibitors of mRNA translation and predicted regulators of the sigma-1 and sigma-2 receptors. Further studies of these host-factor-targeting agents, including their combination with drugs that directly target viral enzymes, could lead to a therapeutic regimen to treat COVID-19.- Published
- 2020
- Full Text
- View/download PDF
24. 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
25. Development of Ependymal and Postnatal Neural Stem Cells and Their Origin from a Common Embryonic Progenitor.
- Author
-
Redmond SA, Figueres-Oñate M, Obernier K, Nascimento MA, Parraguez JI, López-Mascaraque L, Fuentealba LC, and Alvarez-Buylla A
- Subjects
- Animals, Humans, Mice, Ependyma embryology, Neural Stem Cells metabolism, Neurogenesis genetics
- Abstract
The adult mouse brain contains an extensive neurogenic niche in the lateral walls of the lateral ventricles. This epithelium, which has a unique pinwheel organization, contains multiciliated ependymal (E1) cells and neural stem cells (B1). This postnatal germinal epithelium develops from the embryonic ventricular zone, but the lineage relationship between E1 and B1 cells remains unknown. Distinct subpopulations of radial glia (RG) cells in late embryonic and early postnatal development either expand their apical domain >11-fold to form E1 cells or retain small apical domains that coalesce into the centers of pinwheels to form B1 cells. Using independent methods of lineage tracing, we show that individual RG cells can give rise to clones containing E1 and B1 cells. This study reveals key developmental steps in the formation of the postnatal germinal niche and the shared cellular origin of E1 and B1 cells., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
26. Neural stem cells: origin, heterogeneity and regulation in the adult mammalian brain.
- Author
-
Obernier K and Alvarez-Buylla A
- Subjects
- Animals, Cell Communication, Cell Differentiation, Cell Lineage, Embryonic Stem Cells physiology, Hippocampus embryology, Humans, Interneurons physiology, Lateral Ventricles embryology, Mice, Neurons physiology, Olfactory Bulb embryology, Sequence Analysis, RNA, Signal Transduction, Single-Cell Analysis, Transcriptome, Adult Stem Cells physiology, Hippocampus physiology, Lateral Ventricles physiology, Neural Stem Cells physiology, Neurogenesis physiology, Olfactory Bulb physiology
- Abstract
In the adult rodent brain, neural stem cells (NSCs) persist in the ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ), which are specialized niches in which young neurons for the olfactory bulb (OB) and hippocampus, respectively, are generated. Recent studies have significantly modified earlier views on the mechanisms of NSC self-renewal and neurogenesis in the adult brain. Here, we discuss the molecular control, heterogeneity, regional specification and cell division modes of V-SVZ NSCs, and draw comparisons with NSCs in the SGZ. We highlight how V-SVZ NSCs are regulated by local signals from their immediate neighbors, as well as by neurotransmitters and factors that are secreted by distant neurons, the choroid plexus and vasculature. We also review recent advances in single cell RNA analyses that reveal the complexity of adult neurogenesis. These findings set the stage for a better understanding of adult neurogenesis, a process that one day may inspire new approaches to brain repair., (© 2019. Published by The Company of Biologists Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
27. A tension-mediated glycocalyx-integrin feedback loop promotes mesenchymal-like glioblastoma.
- Author
-
Barnes JM, Kaushik S, Bainer RO, Sa JK, Woods EC, Kai F, Przybyla L, Lee M, Lee HW, Tung JC, Maller O, Barrett AS, Lu KV, Lakins JN, Hansen KC, Obernier K, Alvarez-Buylla A, Bergers G, Phillips JJ, Nam DH, Bertozzi CR, and Weaver VM
- Subjects
- Animals, Antineoplastic Agents, Alkylating therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Cell Survival drug effects, Feedback, Physiological drug effects, Glioblastoma drug therapy, Glioblastoma genetics, Humans, Mesenchymal Stem Cells drug effects, Mice, Nude, Neoplastic Stem Cells drug effects, Surface Tension, Temozolomide therapeutic use, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Brain Neoplasms metabolism, Glioblastoma metabolism, Glycocalyx metabolism, Integrins metabolism, Mesenchymal Stem Cells metabolism, Neoplastic Stem Cells metabolism
- Abstract
Glioblastoma multiforme (GBMs) are recurrent lethal brain tumours. Recurrent GBMs often exhibit mesenchymal, stem-like phenotypes that could explain their resistance to therapy. Analyses revealed that recurrent GBMs have increased tension and express high levels of glycoproteins that increase the bulkiness of the glycocalyx. Studies showed that a bulky glycocalyx potentiates integrin mechanosignalling and tissue tension and promotes a mesenchymal, stem-like phenotype in GBMs. Gain- and loss-of-function studies implicated integrin mechanosignalling as an inducer of GBM growth, survival, invasion and treatment resistance, and a mesenchymal, stem-like phenotype. Mesenchymal-like GBMs were highly contractile and expressed elevated levels of glycoproteins that expanded their glycocalyx, and they were surrounded by a stiff extracellular matrix that potentiated integrin mechanosignalling. Our findings suggest that there is a dynamic and reciprocal link between integrin mechanosignalling and a bulky glycocalyx, implying a causal link towards a mesenchymal, stem-like phenotype in GBMs. Strategies to ameliorate GBM tissue tension offer a therapeutic approach to reduce mortality due to GBM.
- Published
- 2018
- Full Text
- View/download PDF
28. A Glial Signature and Wnt7 Signaling Regulate Glioma-Vascular Interactions and Tumor Microenvironment.
- Author
-
Griveau A, Seano G, Shelton SJ, Kupp R, Jahangiri A, Obernier K, Krishnan S, Lindberg OR, Yuen TJ, Tien AC, Sabo JK, Wang N, Chen I, Kloepper J, Larrouquere L, Ghosh M, Tirosh I, Huillard E, Alvarez-Buylla A, Oldham MC, Persson AI, Weiss WA, Batchelor TT, Stemmer-Rachamimov A, Suvà ML, Phillips JJ, Aghi MK, Mehta S, Jain RK, and Rowitch DH
- Subjects
- Animals, Bevacizumab pharmacology, Blood-Brain Barrier metabolism, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Cell Line, Tumor, Gene Expression Regulation, Neoplastic drug effects, Glioma drug therapy, Glioma metabolism, Humans, Mice, Neoplasm Transplantation, Oligodendrocyte Transcription Factor 2 genetics, Temozolomide pharmacology, Tumor Cells, Cultured, Tumor Microenvironment, Wnt Proteins genetics, Wnt Signaling Pathway drug effects, Brain Neoplasms blood supply, Glioma blood supply, Oligodendrocyte Transcription Factor 2 metabolism, Oligodendroglia microbiology, Wnt Proteins metabolism
- Abstract
Gliomas comprise heterogeneous malignant glial and stromal cells. While blood vessel co-option is a potential mechanism to escape anti-angiogenic therapy, the relevance of glial phenotype in this process is unclear. We show that Olig2
+ oligodendrocyte precursor-like glioma cells invade by single-cell vessel co-option and preserve the blood-brain barrier (BBB). Conversely, Olig2-negative glioma cells form dense perivascular collections and promote angiogenesis and BBB breakdown, leading to innate immune cell activation. Experimentally, Olig2 promotes Wnt7b expression, a finding that correlates in human glioma profiling. Targeted Wnt7a/7b deletion or pharmacologic Wnt inhibition blocks Olig2+ glioma single-cell vessel co-option and enhances responses to temozolomide. Finally, Olig2 and Wnt7 become upregulated after anti-VEGF treatment in preclinical models and patients. Thus, glial-encoded pathways regulate distinct glioma-vascular microenvironmental interactions., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
29. Adult Neurogenesis Is Sustained by Symmetric Self-Renewal and Differentiation.
- Author
-
Obernier K, Cebrian-Silla A, Thomson M, Parraguez JI, Anderson R, Guinto C, Rodas Rodriguez J, Garcia-Verdugo JM, and Alvarez-Buylla A
- Subjects
- Animals, Cell Count, Humans, Interneurons cytology, Mice, Transgenic, Neural Stem Cells cytology, Neural Stem Cells metabolism, Time Factors, Cell Differentiation, Cell Self Renewal, Neurogenesis
- Abstract
Somatic stem cells have been identified in multiple adult tissues. Whether self-renewal occurs symmetrically or asymmetrically is key to understanding long-term stem cell maintenance and generation of progeny for cell replacement. In the adult mouse brain, neural stem cells (NSCs) (B1 cells) are retained in the walls of the lateral ventricles (ventricular-subventricular zone [V-SVZ]). The mechanism of B1 cell retention into adulthood for lifelong neurogenesis is unknown. Using multiple clonal labeling techniques, we show that the vast majority of B1 cells divide symmetrically. Whereas 20%-30% symmetrically self-renew and can remain in the niche for several months before generating neurons, 70%-80% undergo consuming divisions generating progeny, resulting in the depletion of B1 cells over time. This cellular mechanism decouples self-renewal from the generation of progeny. Limited rounds of symmetric self-renewal and consuming symmetric differentiation divisions can explain the levels of neurogenesis observed throughout life., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
30. Embryonic Origin of Postnatal Neural Stem Cells.
- Author
-
Fuentealba LC, Rompani SB, Parraguez JI, Obernier K, Romero R, Cepko CL, and Alvarez-Buylla A
- Subjects
- Adult Stem Cells classification, Animals, Mice, Neural Stem Cells classification, Prosencephalon cytology, Adult Stem Cells cytology, Cell Lineage, Embryo, Mammalian cytology, Neural Stem Cells cytology, Olfactory Bulb cytology
- Abstract
Adult neural stem/progenitor (B1) cells within the walls of the lateral ventricles generate different types of neurons for the olfactory bulb (OB). The location of B1 cells determines the types of OB neurons they generate. Here we show that the majority of mouse B1 cell precursors are produced between embryonic days (E) 13.5 and 15.5 and remain largely quiescent until they become reactivated postnatally. Using a retroviral library carrying over 100,000 genetic tags, we found that B1 cells share a common progenitor with embryonic cells of the cortex, striatum, and septum, but this lineage relationship is lost before E15.5. The regional specification of B1 cells is evident as early as E11.5 and is spatially linked to the production of neurons that populate different areas of the forebrain. This study reveals an early embryonic regional specification of postnatal neural stem cells and the lineage relationship between them and embryonic progenitor cells., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
31. Primary cilia are required in a unique subpopulation of neural progenitors.
- Author
-
Tong CK, Han YG, Shah JK, Obernier K, Guinto CD, and Alvarez-Buylla A
- Subjects
- Animals, Animals, Newborn, Brain embryology, Brain growth & development, Brain metabolism, Cell Proliferation, Embryonic Stem Cells classification, Embryonic Stem Cells metabolism, Embryonic Stem Cells ultrastructure, Female, Gene Knockdown Techniques, Glial Fibrillary Acidic Protein genetics, Glial Fibrillary Acidic Protein metabolism, Hedgehog Proteins physiology, Humans, Kinesins antagonists & inhibitors, Kinesins genetics, Kinesins metabolism, Mice, Mice, Transgenic, Nestin genetics, Nestin metabolism, Neural Stem Cells metabolism, Neurogenesis physiology, Pregnancy, Signal Transduction, Tumor Suppressor Proteins antagonists & inhibitors, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Cilia physiology, Neural Stem Cells classification, Neural Stem Cells ultrastructure
- Abstract
The apical domain of embryonic (radial glia) and adult (B1 cells) neural stem cells (NSCs) contains a primary cilium. This organelle has been suggested to function as an antenna for the detection of morphogens or growth factors. In particular, primary cilia are essential for Hedgehog (Hh) signaling, which plays key roles in brain development. Their unique location facing the ventricular lumen suggests that primary cilia in NSCs could play an important role in reception of signals within the cerebrospinal fluid. Surprisingly, ablation of primary cilia using conditional alleles for genes essential for intraflagellar transport [kinesin family member 3A (Kif3a) and intraflagellar transport 88 (Ift88)] and Cre drivers that are activated at early [Nestin; embryonic day 10.5 (E10.5)] and late [human glial fibrillary acidic protein (hGFAP); E13.5] stages of mouse neural development resulted in no apparent developmental defects. Neurogenesis in the ventricular-subventricular zone (V-SVZ) shortly after birth was also largely unaffected, except for a restricted ventral domain previously known to be regulated by Hh signaling. However, Kif3a and Ift88 genetic ablation also disrupts ependymal cilia, resulting in hydrocephalus by postnatal day 4. To directly study the role of B1 cells' primary cilia without the confounding effects of hydrocephalus, we stereotaxically targeted elimination of Kif3a from a subpopulation of radial glia, which resulted in ablation of primary cilia in a subset of B1 cells. Again, this experiment resulted in decreased neurogenesis only in the ventral V-SVZ. Primary cilia ablation led to disruption of Hh signaling in this subdomain. We conclude that primary cilia are required in a specific Hh-regulated subregion of the postnatal V-SVZ.
- Published
- 2014
- Full Text
- View/download PDF
32. Restricted nature of adult neural stem cells: re-evaluation of their potential for brain repair.
- Author
-
Obernier K, Tong CK, and Alvarez-Buylla A
- Abstract
Neural stem cells (NSCs) in the walls of the lateral ventricles continue to produce new neurons and oligodendrocytes throughout life. The identification of NSCs, long-range neuronal migration, and the integration of new neurons into fully formed mature neural circuits-all in the juvenile or adult brain-has dramatically changed concepts in neurodevelopment and suggests new strategies for brain repair. Yet, the latter has to be seen in perspective: NSCs in the adult are heterogeneous and highly regionally specified; young neurons derived from these primary progenitors migrate and integrate in specific brain regions. Neurogenesis appears to have a function in brain plasticity rather than brain repair. If similar processes could be induced in regions of the brain that are normally not a target of new neurons, therapeutic neuronal replacement may one day reinstate neural circuit plasticity and possibly repair broken neural circuits.
- Published
- 2014
- Full Text
- View/download PDF
33. Proliferation and cilia dynamics in neural stem cells prospectively isolated from the SEZ.
- Author
-
Khatri P, Obernier K, Simeonova IK, Hellwig A, Hölzl-Wenig G, Mandl C, Scholl C, Wölfl S, Winkler J, Gaspar JA, Sachinidis A, and Ciccolini F
- Subjects
- Animals, Animals, Newborn, Apoptosis, Blotting, Western, Cell Cycle, Cell Lineage, Cell Membrane metabolism, Cells, Cultured, Ependyma metabolism, ErbB Receptors metabolism, Fluorescent Antibody Technique, Gene Expression Profiling, Mice, Mice, Inbred C57BL, Neural Stem Cells metabolism, Neurogenesis physiology, Neurons metabolism, Oligonucleotide Array Sequence Analysis, Biomarkers metabolism, Cell Differentiation, Cell Proliferation, Cilia physiology, Ependyma cytology, Neural Stem Cells cytology, Neurons cytology
- Abstract
Neural stem cells (NSCs) generate new neurons in vivo and in vitro throughout adulthood and therefore are physiologically and clinically relevant. Unveiling the mechanisms regulating the lineage progression from NSCs to newborn neurons is critical for the transition from basic research to clinical application. However, the direct analysis of NSCs and their progeny is still elusive due to the problematic identification of the cells. We here describe the isolation of highly purified genetically unaltered NSCs and transit-amplifying precursors (TAPs) from the adult subependymal zone (SEZ). Using this approach we show that a primary cilium and high levels of epidermal growth factor receptor (EGFR) at the cell membrane characterize quiescent and cycling NSCs, respectively. However, we also observed non-ciliated quiescent NSCs and NSCs progressing into the cell cycle without up-regulating EGFR expression. Thus, the existence of NSCs displaying distinct molecular and structural conformations provides more flexibility to the regulation of quiescence and cell cycle progression.
- Published
- 2014
- Full Text
- View/download PDF
34. Molecular diversity subdivides the adult forebrain neural stem cell population.
- Author
-
Giachino C, Basak O, Lugert S, Knuckles P, Obernier K, Fiorelli R, Frank S, Raineteau O, Alvarez-Buylla A, and Taylor V
- Subjects
- Animals, Cell Growth Processes physiology, Humans, Mice, Mice, Transgenic, Neural Stem Cells metabolism, Neurogenesis, Neurons metabolism, Prosencephalon metabolism, Signal Transduction, Neural Stem Cells cytology, Neurons cytology, Prosencephalon cytology
- Abstract
Neural stem cells (NSCs) in the ventricular domain of the subventricular zone (V-SVZ) of rodents produce neurons throughout life while those in humans become largely inactive or may be lost during infancy. Most adult NSCs are quiescent, express glial markers, and depend on Notch signaling for their self-renewal and the generation of neurons. Using genetic markers and lineage tracing, we identified subpopulations of adult V-SVZ NSCs (type 1, 2, and 3) indicating a striking heterogeneity including activated, brain lipid binding protein (BLBP, FABP7) expressing stem cells. BLBP(+) NSCs are mitotically active components of pinwheel structures in the lateral ventricle walls and persistently generate neurons in adulthood. BLBP(+) NSCs express epidermal growth factor (EGF) receptor, proliferate in response to EGF, and are a major clonogenic population in the SVZ. We also find BLBP expressed by proliferative ventricular and subventricular progenitors in the fetal and postnatal human brain. Loss of BLBP(+) stem/progenitor cells correlates with reduced neurogenesis in aging rodents and postnatal humans. These findings of molecular heterogeneity and proliferative differences subdivide the NSC population and have implications for neurogenesis in the forebrain of mammals during aging., (Copyright © 2013 AlphaMed Press.)
- Published
- 2014
- Full Text
- View/download PDF
35. Lineage progression from stem cells to new neurons in the adult brain ventricular-subventricular zone.
- Author
-
Ponti G, Obernier K, and Alvarez-Buylla A
- Subjects
- Animals, Astrocytes cytology, Astrocytes metabolism, Cell Lineage, Cerebral Ventricles cytology, Cerebral Ventricles physiology, Glial Fibrillary Acidic Protein, Mice, Mitosis, Nerve Tissue Proteins metabolism, Neural Stem Cells metabolism, Neurons cytology, Neurons physiology, Neural Stem Cells cytology
- Published
- 2013
- Full Text
- View/download PDF
36. Cell cycle and lineage progression of neural progenitors in the ventricular-subventricular zones of adult mice.
- Author
-
Ponti G, Obernier K, Guinto C, Jose L, Bonfanti L, and Alvarez-Buylla A
- Subjects
- Adult Stem Cells cytology, Animals, Humans, Lateral Ventricles cytology, Mice, Mice, Transgenic, Neural Stem Cells cytology, Time Factors, Adult Stem Cells metabolism, Lateral Ventricles metabolism, Neural Stem Cells metabolism, S Phase physiology
- Abstract
Proliferating neural stem cells and intermediate progenitors persist in the ventricular-subventricular zone (V-SVZ) of the adult mammalian brain. This extensive germinal layer in the walls of the lateral ventricles is the site of birth of different types of interneurons destined for the olfactory bulb. The cell cycle dynamics of stem cells (B1 cells), intermediate progenitors (C cells), and neuroblasts (A cells) in the V-SVZ and the number of times these cells divide remain unknown. Using whole mounts of the walls of the lateral ventricles of adult mice and three cell cycle analysis methods using thymidine analogs, we determined the proliferation dynamics of B1, C, and A cells in vivo. Achaete-scute complex homolog (Ascl)1(+) C cells were heterogeneous with a cell cycle length (T(C)) of 18-25 h and a long S phase length (T(S)) of 14-17 h. After C cells, Doublecortin(+) A cells were the second-most common dividing cell type in the V-SVZ and had a T(C) of 18 h and T(S) of 9 h. Human glial fibrillary acidic protein (hGFAP)::GFP(+) B1 cells had a surprisingly short Tc of 17-18 h and a T(S) of 4 h. Progenitor population analysis suggests that following the initial division of B1 cells, C cells divide three times and A cells once, possibly twice. These data provide essential information on the dynamics of adult progenitor cell proliferation in the V-SVZ and how large numbers of new neurons continue to be produced in the adult mammalian brain.
- Published
- 2013
- Full Text
- View/download PDF
37. Adult neural stem cells bridge their niche.
- Author
-
Fuentealba LC, Obernier K, and Alvarez-Buylla A
- Subjects
- Adult, Adult Stem Cells physiology, Humans, Neural Stem Cells physiology, Signal Transduction, Adult Stem Cells cytology, Brain cytology, Neural Stem Cells cytology, Neurons cytology, Stem Cell Niche
- Abstract
Major developments in the neural stem cell (NSC) field in recent years provide new insights into the nature of the NSC niche. In this perspective, we integrate recent anatomical data on the organization of the two main neurogenic niches in the adult brain, the ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ), with signaling pathways that control the behavior of NSCs. NSCs in the adult brain stretch into physiologically distinct compartments of their niche. We propose how adult NSCs' morphology may allow these cells to integrate multiple signaling pathways arising from unique locations of their niche., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
38. Expression of Tlx in both stem cells and transit amplifying progenitors regulates stem cell activation and differentiation in the neonatal lateral subependymal zone.
- Author
-
Obernier K, Simeonova I, Fila T, Mandl C, Hölzl-Wenig G, Monaghan-Nichols P, and Ciccolini F
- Subjects
- Animals, Cell Differentiation physiology, Cell Growth Processes physiology, Female, Genotype, Lateral Ventricles growth & development, Mice, Neurogenesis, Pregnancy, Receptors, Cytoplasmic and Nuclear genetics, Lateral Ventricles cytology, Lateral Ventricles metabolism, Neural Stem Cells cytology, Neural Stem Cells metabolism, Receptors, Cytoplasmic and Nuclear biosynthesis
- Abstract
Niche homeostasis in the postnatal subependymal zone of the lateral ventricle (lSEZ) requires coordinated proliferation and differentiation of neural progenitor cells. The mechanisms regulating this balance are scarcely known. Recent observations indicate that the orphan nuclear receptor Tlx is an intrinsic factor essential in maintaining this balance. However, the effect of Tlx on gene expression depends on age and cell-type cues. Therefore, it is essential to establish its expression pattern at different developmental ages. Here, we show for the first time that in the neonatal lSEZ activated neural stem cells (NSCs) and especially transit-amplifying progenitors (TAPs) express Tlx and that its expression may be regulated at the posttranscriptional level. We also provide evidence that in both cell types Tlx affects gene expression in a positive and negative manner. In activated NSCs, but not in TAPs, absence of Tlx leads to overexpression of negative cell cycle regulators and impairment of proliferation. Moreover, in both cell types, the homeobox transcription factor Dlx2 is downregulated in the absence of Tlx. This is paralleled by increased expression of Olig2 in activated NSCs and glial fibrillary acidic protein in TAPs, indicating that in both populations Tlx decreases gliogenesis. Consistent with this, we found a higher proportion of cells expressing glial makers in the neonatal lSEZ of mutant mice than in the wild type counterpart. Thus, Tlx playing a dual role affects the expression of distinct genes in these two lSEZ cell types., (Copyright © 2011 AlphaMed Press.)
- Published
- 2011
- Full Text
- View/download PDF
39. GABAA receptor signaling induces osmotic swelling and cell cycle activation of neonatal prominin+ precursors.
- Author
-
Cesetti T, Fila T, Obernier K, Bengtson CP, Li Y, Mandl C, Hölzl-Wenig G, and Ciccolini F
- Subjects
- AC133 Antigen, Animals, Animals, Newborn, Cell Proliferation, Cells, Cultured, Diazepam pharmacology, ErbB Receptors biosynthesis, ErbB Receptors metabolism, Mice, Mice, Inbred C57BL, Neural Stem Cells cytology, Neural Stem Cells drug effects, Osmotic Pressure physiology, Protein Precursors metabolism, gamma-Aminobutyric Acid metabolism, Antigens, CD metabolism, Cell Cycle drug effects, Glycoproteins metabolism, Neural Stem Cells metabolism, Peptides metabolism, Receptors, GABA-A metabolism, Signal Transduction drug effects
- Abstract
Signal-regulated changes in cell size affect cell division and survival and therefore are central to tissue morphogenesis and homeostasis. In this respect, GABA receptors (GABA(A)Rs) are of particular interest because allowing anions flow across the cell membrane modulates the osmolyte flux and the cell volume. Therefore, we have here investigated the hypothesis that GABA may regulate neural stem cell proliferation by inducing cell size changes. We found that, besides neuroblasts, also neural precursors in the neonatal murine subependymal zone sense GABA via GABA(A) Rs. However, unlike in neuroblasts, where it induced depolarization-mediated [Ca(2+)](i) increase, GABA(A) Rs activation in precursors caused hyperpolarization. This resulted in osmotic swelling and increased surface expression of epidermal growth factor receptors (EGFRs). Furthermore, activation of GABA(A) Rs signaling in vitro in the presence of EGF modified the expression of the cell cycle regulators, phosphatase and tensin homolog and cyclin D1, increasing the pool of cycling precursors without modifying cell cycle length. A similar effect was observed on treatment with diazepam. We also demonstrate that GABA and diazepam responsive precursors represent prominin(+) stem cells. Finally, we show that as in in vitro also in in vivo a short administration of diazepam promotes EGFR expression in prominin(+) stem cells causing activation and cell cycle entry. Thus, our data indicate that endogenous GABA is a part of a regulatory mechanism of size and cell cycle entry of neonatal stem cells. Our results also have potential implications for the therapeutic practices that involve exposure to GABA(A) Rs modulators during neurodevelopment., (Copyright © 2010 AlphaMed Press.)
- Published
- 2011
- Full Text
- View/download PDF
40. Multipotent precursors in the anterior and hippocampal subventricular zone display similar transcription factor signatures but their proliferation and maintenance are differentially regulated.
- Author
-
Carrillo-García C, Suh Y, Obernier K, Hölzl-Wenig G, Mandl C, and Ciccolini F
- Subjects
- Age Factors, Animals, Cell Differentiation physiology, Cell Proliferation, Cells, Cultured, ErbB Receptors genetics, Female, Mice, Mice, Inbred C57BL, Multipotent Stem Cells cytology, Neurons cytology, Organ Specificity, Pregnancy, Stem Cell Niche cytology, ErbB Receptors metabolism, Homeodomain Proteins metabolism, Multipotent Stem Cells physiology, Neurogenesis physiology, Neurons physiology, Stem Cell Niche physiology, Transcription Factors metabolism
- Abstract
Precursors within the subventricular zone (SVZ) exhibit regional variations in the expression of transcription factors important for the regulation of their proliferation and differentiation. In the anterior SVZ (aSVZ) the homeobox transcription factor distalless (Dlx)2 modulates both processes by promoting neural stem cell (NSC) activation as well as neurogenesis. Activated NSCs and transit-amplifying precursors (TAPs) in the aSVZ both express high levels of epidermal growth factor receptor (EGFR(high)) and form clones in response to exogenous EGF. EGF-responsive cells are also present in the hippocampal subependyma (hSVZ). However, it is not clear whether they represent NSCs or TAPs and whether their proliferation and differentiation are regulated as in the aSVZ. Here we have purified EGFR(high) cells from both the aSVZ and hSVZ at different ages. When isolated from perinatal tissue both populations were enriched in multipotent clonogenic precursors, which generated GABAergic neurons. Although they differed in absolute expression levels, activated NSCs and TAPs in both regions displayed similar signatures of transcription factor expression. However, activated NSCs were less frequent in the hSVZ than in the aSVZ. Furthermore, increasing age had a greater inhibitory effect on NSC proliferation in the hSVZ than in the aSVZ. This suggests that NSC activation is differentially regulated in the two regions. Consistent with this hypothesis, we found that in hippocampal precursors Dlx2 promoted neurogenesis but not NSC activation. Thus, most clonogenic EGFR(high) precursors in the hSVZ represent TAPs and NSC proliferation in the aSVZ and hSVZ is regulated by different mechanisms.
- Published
- 2010
- Full Text
- View/download PDF
41. Interaction between DLX2 and EGFR regulates proliferation and neurogenesis of SVZ precursors.
- Author
-
Suh Y, Obernier K, Hölzl-Wenig G, Mandl C, Herrmann A, Wörner K, Eckstein V, and Ciccolini F
- Subjects
- Animals, Cell Differentiation physiology, Cell Lineage, Cells, Cultured, Epidermal Growth Factor metabolism, ErbB Receptors genetics, Homeodomain Proteins genetics, Mice, Neurons cytology, Signal Transduction physiology, Stem Cell Niche cytology, Stem Cells cytology, Transcription Factors genetics, Cell Proliferation, ErbB Receptors metabolism, Homeodomain Proteins metabolism, Neurogenesis physiology, Neurons physiology, Stem Cell Niche physiology, Stem Cells physiology, Transcription Factors metabolism
- Abstract
In the postnatal subventricular zone (SVZ) neural stem cells (NSCs) give rise to transit-amplifying precursors (TAPs) expressing high levels of epidermal growth factor receptor (EGFR) that in turn generate neuroblasts. Both TAPs and neuroblasts express distal-less (DLX)2 homeobox transcription factor but the latter proliferate less. Modulation of its expression in vivo has revealed that DLX2 affects both neurogenesis and proliferation in the postnatal SVZ. However, the mechanisms underlying these effects are not clear. To investigate this issue we have here forced the expression of DLX2 in SVZ isolated NSCs growing in defined in vitro conditions. This analysis revealed that DLX2 affects the proliferation of SVZ precursors by regulating two distinct steps of neural lineage progression. Firstly, it promotes the lineage transition from NSCs to TAPs. Secondly it enhances the proliferative response of neuronal progenitors to EGF. Thus DLX2 and EGFR signalling interact at multiple levels to coordinate proliferation in the postnatal SVZ.
- Published
- 2009
- Full Text
- View/download PDF
42. Analysis of stem cell lineage progression in the neonatal subventricular zone identifies EGFR+/NG2- cells as transit-amplifying precursors.
- Author
-
Cesetti T, Obernier K, Bengtson CP, Fila T, Mandl C, Hölzl-Wenig G, Wörner K, Eckstein V, and Ciccolini F
- Subjects
- Animals, Animals, Newborn, Antigens metabolism, Brain metabolism, Cell Differentiation, ErbB Receptors metabolism, Flow Cytometry, Immunohistochemistry, Mice, Neurons metabolism, Patch-Clamp Techniques, Proteoglycans metabolism, Brain cytology, Cell Lineage, Neurogenesis physiology, Neurons cytology, Stem Cells cytology
- Abstract
In the adult subventricular zone (SVZ), astroglial stem cells generate transit-amplifying precursors (TAPs). Both stem cells and TAPs form clones in response to epidermal growth factor (EGF). However, in vivo, in the absence of sustained EGF receptor (EGFR) activation, TAPs divide a few times before differentiating into neuroblasts. The lack of suitable markers has hampered the analysis of stem cell lineage progression and associated functional changes in the neonatal germinal epithelium. Here we purified neuroblasts and clone-forming precursors from the neonatal SVZ using expression levels of EGFR and polysialylated neural cell adhesion molecule (PSANCAM). As in the adult SVZ, most neonatal clone-forming precursors did not express the neuroglia proteoglycan 2 (NG2) but displayed characteristics of TAPs, and only a subset exhibited antigenic characteristics of astroglial stem cells. Both precursors and neuroblasts were PSANCAM(+); however, neuroblasts also expressed doublecortin and functional voltage-dependent Ca(2+) channels. Neuroblasts and precursors had distinct outwardly rectifying K(+) current densities and passive membrane properties, particularly in precursors contacting each other, because of the contribution of gap junction coupling. Confirming the hypothesis that most are TAPs, cell tracing in brain slices revealed that within 2 days the majority of EGFR(+) cells had exited the cell cycle and differentiated into a progenitor displaying intermediate antigenic and functional properties between TAPs and neuroblasts. Thus, distinct functional and antigenic properties mark stem cell lineage progression in the neonatal SVZ.
- Published
- 2009
- Full Text
- View/download PDF
43. Functional characterization and transcriptome analysis of embryonic stem cell-derived contractile smooth muscle cells.
- Author
-
Potta SP, Liang H, Pfannkuche K, Winkler J, Chen S, Doss MX, Obernier K, Kamisetti N, Schulz H, Hübner N, Hescheler J, and Sachinidis A
- Subjects
- Actins genetics, Angiotensin II pharmacology, Animals, Calcium-Binding Proteins genetics, Calcium-Binding Proteins metabolism, Cell Differentiation genetics, Cells, Cultured, Desmin genetics, Desmin metabolism, Endothelins pharmacology, Hypertrophy pathology, KATP Channels genetics, KATP Channels metabolism, Mice, Mice, Transgenic, Microfilament Proteins genetics, Microfilament Proteins metabolism, Muscle Contraction genetics, Muscle Proteins genetics, Muscle Proteins metabolism, Myocytes, Smooth Muscle drug effects, Myosin Heavy Chains genetics, Myosin Heavy Chains metabolism, Patch-Clamp Techniques, Potassium Channels, Calcium-Activated genetics, Potassium Channels, Calcium-Activated metabolism, Calponins, Actins metabolism, Embryonic Stem Cells cytology, Gene Expression Profiling, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology
- Abstract
Complete transcriptome profiling of contractile smooth muscle cells (SMCs) differentiated from embryonic stem cells is crucial for the characterization of smooth muscle gene expression signatures and will contribute to defining biological and physiological processes in these cells. We have generated a transgenic embryonic stem cell line expressing both the puromycin acetyl transferase and enhanced green fluorescent protein cassettes under the control of the Acta2 promoter. Applying a specific monolayer culture protocol using retinoic acid, a puromycin-resistant and enhanced green fluorescent protein-positive Acta2(+) SMC population of 95% purity was isolated. Acta2(+) SMCs were characterized by semiquantitative and quantitative RT-PCR profiling of SMC markers and by microarray expression profiling, as well as by immunostaining for SMC-specific cytoskeletal proteins. Patch-clamp electrophysiological characterization of these cells identified SMC-specific channels such as the ATP-sensitive potassium channel and the Ca(2+)-activated potassium channel. Culturing of Acta2(+) SMCs in serum-containing medium resulted in a significant number of hypertrophic and binucleated cells failing to complete cell division. Functional characterization of the cells has been proved by stimulation of the cells with vasoactive agents, such as angiotensin II and endothelin. We concluded that our embryonic stem cell-derived SMC population possesses the contractile and hypertrophic phenotype of SMCs incapable of proliferation. This is the first study describing the complete transcriptome of ES-derived SMCs allowing identification of specific biological and physiological processes in the contractile phenotype SMCs and will contribute to the understanding of these processes in early SMCs derived from embryonic stem cells.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.