1
|
Zhang X, Pang T, Zhang H, Horn M, Michlits G, Dyczynski M, Zhang L. The natural compound periplogenin suppresses the growth of prostate carcinoma cells by directly targeting ATP1A1. Sci Rep 2024; 14:20509. [PMID: 39227746 PMCID: PMC11372130 DOI: 10.1038/s41598-024-71722-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024] Open
Abstract
Natural compounds constitute a major resource for the development of medicines for multiple diseases. While many natural compounds show strong biological activity, the mechanisms that confer clinical benefits are often elusive and have been attributed to multiple pathways. Periplogenin (PPG), a natural compound isolated from Cortex periplocae, exhibits strong anti-tumor activities in several human cancer cell lines. However, its molecular mode of action remained unclear. In this study, we leveraged a forward genetic screening approach in DU145 prostate cancer cells to uncover the molecular target of PPG using chemical mutagenesis. Next generation sequencing revealed that a single amino acid substitution at amino acid 804 in ATP1A1 (ATPase Na + /K + Transporting Subunit Alpha 1) confers resistance to the cytotoxic activity of PPG. Mechanistically, ATP1A1 T804 forms a hydrogen bond with PPG which is abolished by the T804A substitution in ATP1A1, resulting in resistance to PPG treatment in vitro. Importantly, in vivo, PPG strongly suppressed tumor development in a DU145 xenograft model whereas DU145 xenograft tumors carrying a ATP1A1-T804A mutation were largely unaffected by the treatment. These findings demonstrate that PPG suppresses the growth of DU145 prostate cancer cells in vitro and in vivo by directly binding to ATP1A1 and highlight the power of our unbiased forward genetic screening approach to uncover direct drug target structures at single amino acid resolution.
Collapse
Affiliation(s)
- Xinquan Zhang
- Angal Biotechnology Co., Ltd., Life Health Town, National High-Tech Development Zone, Suzhou, China
| | - Tinglin Pang
- Angal Biotechnology Co., Ltd., Life Health Town, National High-Tech Development Zone, Suzhou, China
| | - Haifeng Zhang
- Angal Biotechnology Co., Ltd., Life Health Town, National High-Tech Development Zone, Suzhou, China
| | | | | | | | - Liqun Zhang
- Angal Biotechnology Co., Ltd., Life Health Town, National High-Tech Development Zone, Suzhou, China.
| |
Collapse
|
2
|
Monteil VM, Wright SC, Dyczynski M, Kellner MJ, Appelberg S, Platzer SW, Ibrahim A, Kwon H, Pittarokoilis I, Mirandola M, Michlits G, Devignot S, Elder E, Abdurahman S, Bereczky S, Bagci B, Youhanna S, Aastrup T, Lauschke VM, Salata C, Elaldi N, Weber F, Monserrat N, Hawman DW, Feldmann H, Horn M, Penninger JM, Mirazimi A. Crimean-Congo haemorrhagic fever virus uses LDLR to bind and enter host cells. Nat Microbiol 2024; 9:1499-1512. [PMID: 38548922 PMCID: PMC11153131 DOI: 10.1038/s41564-024-01672-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 03/11/2024] [Indexed: 06/07/2024]
Abstract
Climate change and population densities accelerated transmission of highly pathogenic viruses to humans, including the Crimean-Congo haemorrhagic fever virus (CCHFV). Here we report that the Low Density Lipoprotein Receptor (LDLR) is a critical receptor for CCHFV cell entry, playing a vital role in CCHFV infection in cell culture and blood vessel organoids. The interaction between CCHFV and LDLR is highly specific, with other members of the LDLR protein family failing to bind to or neutralize the virus. Biosensor experiments demonstrate that LDLR specifically binds the surface glycoproteins of CCHFV. Importantly, mice lacking LDLR exhibit a delay in CCHFV-induced disease. Furthermore, we identified the presence of Apolipoprotein E (ApoE) on CCHFV particles. Our findings highlight the essential role of LDLR in CCHFV infection, irrespective of ApoE presence, when the virus is produced in tick cells. This discovery holds profound implications for the development of future therapies against CCHFV.
Collapse
Affiliation(s)
- Vanessa M Monteil
- Unit of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | - Shane C Wright
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Matheus Dyczynski
- Acus Laboratories GmbH, Cologne, Germany
- JLP Health GmbH, Vienna, Austria
| | - Max J Kellner
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
- Vienna Biocenter PhD Program, a Doctoral School of the University of Vienna and the Medical University of Vienna, Vienna, Austria
| | | | - Sebastian W Platzer
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
- Vienna Biocenter PhD Program, a Doctoral School of the University of Vienna and the Medical University of Vienna, Vienna, Austria
| | | | - Hyesoo Kwon
- National Veterinary Institute, Uppsala, Sweden
| | | | - Mattia Mirandola
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Stephanie Devignot
- Unit of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | | | | | | | - Binnur Bagci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Sivas Cumhuriyet University, Sivas, Turkey
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- University Tübingen, Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Nazif Elaldi
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Cumhuriyet University, Sivas, Turkey
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Gießen, Germany
| | - Nuria Monserrat
- University of Barcelona, Barcelona, Spain
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - David W Hawman
- Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, USA
| | - Heinz Feldmann
- Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, USA
| | - Moritz Horn
- Acus Laboratories GmbH, Cologne, Germany
- JLP Health GmbH, Vienna, Austria
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- Helmholtz Centre for Infection Research, Braunschweig, Germany.
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Ali Mirazimi
- Unit of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden.
- Public Health Agency of Sweden, Solna, Sweden.
- National Veterinary Institute, Uppsala, Sweden.
| |
Collapse
|
3
|
Monteil V, Kwon H, John L, Salata C, Jonsson G, Vorrink SU, Appelberg S, Youhanna S, Dyczynski M, Leopoldi A, Leeb N, Volz J, Hagelkruys A, Kellner MJ, Devignot S, Michlits G, Foong-Sobis M, Weber F, Lauschke VM, Horn M, Feldmann H, Elling U, Penninger JM, Mirazimi A. Identification of CCZ1 as an essential lysosomal trafficking regulator in Marburg and Ebola virus infections. Nat Commun 2023; 14:6785. [PMID: 37880247 PMCID: PMC10600203 DOI: 10.1038/s41467-023-42526-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023] Open
Abstract
Marburg and Ebola filoviruses are two of the deadliest infectious agents and several outbreaks have occurred in the last decades. Although several receptors and co-receptors have been reported for Ebola virus, key host factors remain to be elucidated. In this study, using a haploid cell screening platform, we identify the guanine nucleotide exchange factor CCZ1 as a key host factor in the early stage of filovirus replication. The critical role of CCZ1 for filovirus infections is validated in 3D primary human hepatocyte cultures and human blood-vessel organoids, both critical target sites for Ebola and Marburg virus tropism. Mechanistically, CCZ1 controls early to late endosomal trafficking of these viruses. In addition, we report that CCZ1 has a role in the endosomal trafficking of endocytosis-dependent SARS-CoV-2 infections, but not in infections by Lassa virus, which enters endo-lysosomal trafficking at the late endosome stage. Thus, we have identified an essential host pathway for filovirus infections in cell lines and engineered human target tissues. Inhibition of CCZ1 nearly completely abolishes Marburg and Ebola infections. Thus, targeting CCZ1 could potentially serve as a promising drug target for controlling infections caused by various viruses, such as SARS-CoV-2, Marburg, and Ebola.
Collapse
Affiliation(s)
- Vanessa Monteil
- Karolinska Institute and Karolinska University Hospital, Department of Laboratory Medicine, Unit of Clinical Microbiology, Stockholm, Sweden
| | - Hyesoo Kwon
- National Veterinary Institute, Uppsala, Sweden
| | - Lijo John
- National Veterinary Institute, Uppsala, Sweden
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Gustav Jonsson
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, A-1030, Vienna, Austria
| | - Sabine U Vorrink
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Matheus Dyczynski
- Acus Laboratories GmbH, Cologne, Germany
- JLP Health GmbH, Vienna, Austria
| | - Alexandra Leopoldi
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Nicole Leeb
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Jennifer Volz
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Astrid Hagelkruys
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Max J Kellner
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, A-1030, Vienna, Austria
| | - Stéphanie Devignot
- Karolinska Institute and Karolinska University Hospital, Department of Laboratory Medicine, Unit of Clinical Microbiology, Stockholm, Sweden
| | - Georg Michlits
- Acus Laboratories GmbH, Cologne, Germany
- JLP Health GmbH, Vienna, Austria
| | - Michelle Foong-Sobis
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- University Tübingen, Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Moritz Horn
- Acus Laboratories GmbH, Cologne, Germany
- JLP Health GmbH, Vienna, Austria
| | - Heinz Feldmann
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ulrich Elling
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ali Mirazimi
- Karolinska Institute and Karolinska University Hospital, Department of Laboratory Medicine, Unit of Clinical Microbiology, Stockholm, Sweden.
- National Veterinary Institute, Uppsala, Sweden.
- Public Health Agency of Sweden, Solna, Sweden.
| |
Collapse
|
4
|
Kroef V, Ruegenberg S, Horn M, Allmeroth K, Ebert L, Bozkus S, Miethe S, Elling U, Schermer B, Baumann U, Denzel MS. GFPT2/GFAT2 and AMDHD2 act in tandem to control the hexosamine pathway. eLife 2022; 11:69223. [PMID: 35229715 PMCID: PMC8970586 DOI: 10.7554/elife.69223] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 02/28/2022] [Indexed: 11/25/2022] Open
Abstract
The hexosamine biosynthetic pathway (HBP) produces the essential metabolite UDP-GlcNAc and plays a key role in metabolism, health, and aging. The HBP is controlled by its rate-limiting enzyme glutamine fructose-6-phosphate amidotransferase (GFPT/GFAT) that is directly inhibited by UDP-GlcNAc in a feedback loop. HBP regulation by GFPT is well studied but other HBP regulators have remained obscure. Elevated UDP-GlcNAc levels counteract the glycosylation toxin tunicamycin (TM), and thus we screened for TM resistance in haploid mouse embryonic stem cells (mESCs) using random chemical mutagenesis to determine alternative HBP regulation. We identified the N-acetylglucosamine deacetylase AMDHD2 that catalyzes a reverse reaction in the HBP and its loss strongly elevated UDP-GlcNAc. To better understand AMDHD2, we solved the crystal structure and found that loss-of-function (LOF) is caused by protein destabilization or interference with its catalytic activity. Finally, we show that mESCs express AMDHD2 together with GFPT2 instead of the more common paralog GFPT1. Compared with GFPT1, GFPT2 had a much lower sensitivity to UDP-GlcNAc inhibition, explaining how AMDHD2 LOF resulted in HBP activation. This HBP configuration in which AMDHD2 serves to balance GFPT2 activity was also observed in other mESCs and, consistently, the GFPT2:GFPT1 ratio decreased with differentiation of human embryonic stem cells. Taken together, our data reveal a critical function of AMDHD2 in limiting UDP-GlcNAc production in cells that use GFPT2 for metabolite entry into the HBP.
Collapse
Affiliation(s)
- Virginia Kroef
- Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Sabine Ruegenberg
- Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Kira Allmeroth
- Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | | | - Stephan Miethe
- Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Ulrich Elling
- Vienna Biocenter, Austrian Academy of Sciences, Vienna, Austria
| | | | | | | |
Collapse
|
5
|
Ali Khan A, Raess M, de Angelis MH. Moving forward with forward genetics: A summary of the INFRAFRONTIER Forward Genetics Panel Discussion. F1000Res 2021; 10:456. [PMID: 34900227 PMCID: PMC8634052 DOI: 10.12688/f1000research.25369.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 11/20/2022] Open
Abstract
In the last few decades, forward genetics approaches have been extensively used to identify gene function. Essentially, forward genetics is the elucidation of the genetic basis of a specific phenotype by screening a population containing random genomic modifications that alter gene function. These approaches have shed light on some essential gene functions in development and disease and have expanded the realm of understanding for genetic disorders. Due to the availability of efficient mutagenesis methods, phenotyping techniques, reliable validation, comprehensive sequence information and translational potential, mouse models are favored for forward genetics approaches. However, in this post-genomic CRISPR-Cas9 era, the relevance and future of forward genetics was brought into question. With more than 7300 mouse strains archived and close interactions with several leading mouse researchers around the world, INFRAFRONTIER - the European Research Infrastructure for mouse models organised a panel discussion on forward genetics at the International Mammalian Genome Conference 2018 to discuss the future of forward genetics as well as challenges faced by researchers using this approach in the current research environment. The commentary presents an overview of this discussion.
Collapse
Affiliation(s)
- Asrar Ali Khan
- INFRAFRONTIER GmbH, Neuherberg / Munich, Bavaria, 85764, Germany
| | - Michael Raess
- INFRAFRONTIER GmbH, Neuherberg / Munich, Bavaria, 85764, Germany
| | | |
Collapse
|
6
|
Derisbourg MJ, Wester LE, Baddi R, Denzel MS. Mutagenesis screen uncovers lifespan extension through integrated stress response inhibition without reduced mRNA translation. Nat Commun 2021; 12:1678. [PMID: 33723245 PMCID: PMC7960713 DOI: 10.1038/s41467-021-21743-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Protein homeostasis is modulated by stress response pathways and its deficiency is a hallmark of aging. The integrated stress response (ISR) is a conserved stress-signaling pathway that tunes mRNA translation via phosphorylation of the translation initiation factor eIF2. ISR activation and translation initiation are finely balanced by eIF2 kinases and by the eIF2 guanine nucleotide exchange factor eIF2B. However, the role of the ISR during aging remains poorly understood. Using a genomic mutagenesis screen for longevity in Caenorhabditis elegans, we define a role of eIF2 modulation in aging. By inhibiting the ISR, dominant mutations in eIF2B enhance protein homeostasis and increase lifespan. Consistently, full ISR inhibition using phosphorylation-defective eIF2α or pharmacological ISR inhibition prolong lifespan. Lifespan extension through impeding the ISR occurs without a reduction in overall protein synthesis. Instead, we observe changes in the translational efficiency of a subset of mRNAs, of which the putative kinase kin-35 is required for lifespan extension. Evidently, lifespan is limited by the ISR and its inhibition may provide an intervention in aging.
Collapse
Affiliation(s)
| | - Laura E Wester
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Ruth Baddi
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- CECAD - Cluster of Excellence, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
7
|
Monteil V, Dyczynski M, Lauschke VM, Kwon H, Wirnsberger G, Youhanna S, Zhang H, Slutsky AS, Hurtado del Pozo C, Horn M, Montserrat N, Penninger JM, Mirazimi A. Human soluble ACE2 improves the effect of remdesivir in SARS-CoV-2 infection. EMBO Mol Med 2021; 13:e13426. [PMID: 33179852 PMCID: PMC7799356 DOI: 10.15252/emmm.202013426] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/21/2022] Open
Abstract
There is a critical need for safe and effective drugs for COVID-19. Only remdesivir has received authorization for COVID-19 and has been shown to improve outcomes but not decrease mortality. However, the dose of remdesivir is limited by hepatic and kidney toxicity. ACE2 is the critical cell surface receptor for SARS-CoV-2. Here, we investigated additive effect of combination therapy using remdesivir with recombinant soluble ACE2 (high/low dose) on Vero E6 and kidney organoids, targeting two different modalities of SARS-CoV-2 life cycle: cell entry via its receptor ACE2 and intracellular viral RNA replication. This combination treatment markedly improved their therapeutic windows against SARS-CoV-2 in both models. By using single amino-acid resolution screening in haploid ES cells, we report a singular critical pathway required for remdesivir toxicity, namely, Adenylate Kinase 2. The data provided here demonstrate that combining two therapeutic modalities with different targets, common strategy in HIV treatment, exhibit strong additive effects at sub-toxic concentrations. Our data lay the groundwork for the study of combinatorial regimens in future COVID-19 clinical trials.
Collapse
Affiliation(s)
- Vanessa Monteil
- Department of Laboratory MedicineUnit of Clinical MicrobiologyKarolinska InstituteStockholmSweden
| | | | - Volker M Lauschke
- Department of Physiology and PharmacologyKarolinska InstituteStockholmSweden
| | | | | | - Sonia Youhanna
- Department of Physiology and PharmacologyKarolinska InstituteStockholmSweden
| | - Haibo Zhang
- Keenan Research Centre for Biomedical Science at Li Ka Shing Knowledge Institute of St. Michael’s HospitalUniversity of TorontoTorontoONCanada
| | - Arthur S Slutsky
- Keenan Research Centre for Biomedical Science at Li Ka Shing Knowledge Institute of St. Michael’s HospitalUniversity of TorontoTorontoONCanada
| | - Carmen Hurtado del Pozo
- Pluripotency for Organ RegenerationInstitute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Technology (BIST)BarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Centro de Investigación Biomédica en Red en BioingenieríaBiomateriales y NanomedicinaMadridSpain
| | - Moritz Horn
- Acus Laboratories GmbHCologneGermany
- JLP Health GmbHViennaAustria
| | - Nuria Montserrat
- Pluripotency for Organ RegenerationInstitute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Technology (BIST)BarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Centro de Investigación Biomédica en Red en BioingenieríaBiomateriales y NanomedicinaMadridSpain
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
- Department of Medical GeneticsLife Sciences InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Ali Mirazimi
- Department of Laboratory MedicineUnit of Clinical MicrobiologyKarolinska InstituteStockholmSweden
- National Veterinary InstituteUppsalaSweden
| |
Collapse
|
8
|
Pagliaro L, Marchesini M, Roti G. Targeting oncogenic Notch signaling with SERCA inhibitors. J Hematol Oncol 2021; 14:8. [PMID: 33407740 PMCID: PMC7789735 DOI: 10.1186/s13045-020-01015-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
P-type ATPase inhibitors are among the most successful and widely prescribed therapeutics in modern pharmacology. Clinical transition has been safely achieved for H+/K+ ATPase inhibitors such as omeprazole and Na+/K+-ATPase inhibitors like digoxin. However, this is more challenging for Ca2+-ATPase modulators due to the physiological role of Ca2+ in cardiac dynamics. Over the past two decades, sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) modulators have been studied as potential chemotherapy agents because of their Ca2+-mediated pan-cancer lethal effects. Instead, recent evidence suggests that SERCA inhibition suppresses oncogenic Notch1 signaling emerging as an alternative to γ-secretase modulators that showed limited clinical activity due to severe side effects. In this review, we focus on how SERCA inhibitors alter Notch1 signaling and show that Notch on-target-mediated antileukemia properties of these molecules can be achieved without causing overt Ca2+ cellular overload.
Collapse
Affiliation(s)
- Luca Pagliaro
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Matteo Marchesini
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy.
| |
Collapse
|
9
|
Small-molecule inhibitors of human mitochondrial DNA transcription. Nature 2020; 588:712-716. [PMID: 33328633 DOI: 10.1038/s41586-020-03048-z] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/22/2020] [Indexed: 01/31/2023]
Abstract
Altered expression of mitochondrial DNA (mtDNA) occurs in ageing and a range of human pathologies (for example, inborn errors of metabolism, neurodegeneration and cancer). Here we describe first-in-class specific inhibitors of mitochondrial transcription (IMTs) that target the human mitochondrial RNA polymerase (POLRMT), which is essential for biogenesis of the oxidative phosphorylation (OXPHOS) system1-6. The IMTs efficiently impair mtDNA transcription in a reconstituted recombinant system and cause a dose-dependent inhibition of mtDNA expression and OXPHOS in cell lines. To verify the cellular target, we performed exome sequencing of mutagenized cells and identified a cluster of amino acid substitutions in POLRMT that cause resistance to IMTs. We obtained a cryo-electron microscopy (cryo-EM) structure of POLRMT bound to an IMT, which further defined the allosteric binding site near the active centre cleft of POLRMT. The growth of cancer cells and the persistence of therapy-resistant cancer stem cells has previously been reported to depend on OXPHOS7-17, and we therefore investigated whether IMTs have anti-tumour effects. Four weeks of oral treatment with an IMT is well-tolerated in mice and does not cause OXPHOS dysfunction or toxicity in normal tissues, despite inducing a strong anti-tumour response in xenografts of human cancer cells. In summary, IMTs provide a potent and specific chemical biology tool to study the role of mtDNA expression in physiology and disease.
Collapse
|
10
|
Schirle M, Jenkins JL. Contemporary Techniques for Target Deconvolution and Mode of Action Elucidation. PHENOTYPIC DRUG DISCOVERY 2020. [DOI: 10.1039/9781839160721-00083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The elucidation of the cellular efficacy target and mechanism of action of a screening hit remain key steps in phenotypic drug discovery. A large number of experimental and in silico approaches have been introduced to address these questions and are being discussed in this chapter with a focus on recent developments. In addition to practical considerations such as throughput and technological requirements, these approaches differ conceptually in the specific compound characteristic that they are focusing on, including physical and functional interactions, cellular response patterns as well as structural features. As a result, different approaches often provide complementary information and we describe a multipronged strategy that is frequently key to successful identification of the efficacy target but also other epistatic nodes and off-targets that together shape the overall cellular effect of a bioactive compound.
Collapse
Affiliation(s)
- Markus Schirle
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research Cambridge MA 02139 USA
| | - Jeremy L. Jenkins
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research Cambridge MA 02139 USA
| |
Collapse
|
11
|
Bonekamp NA, Peter B, Hillen HS, Felser A, Bergbrede T, Choidas A, Horn M, Unger A, Di Lucrezia R, Atanassov I, Li X, Koch U, Menninger S, Boros J, Habenberger P, Giavalisco P, Cramer P, Denzel MS, Nussbaumer P, Klebl B, Falkenberg M, Gustafsson CM, Larsson NG. Small-molecule inhibitors of human mitochondrial DNA transcription. Nature 2020. [PMID: 33328633 DOI: 10.1038/s41586-020-03048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Altered expression of mitochondrial DNA (mtDNA) occurs in ageing and a range of human pathologies (for example, inborn errors of metabolism, neurodegeneration and cancer). Here we describe first-in-class specific inhibitors of mitochondrial transcription (IMTs) that target the human mitochondrial RNA polymerase (POLRMT), which is essential for biogenesis of the oxidative phosphorylation (OXPHOS) system1-6. The IMTs efficiently impair mtDNA transcription in a reconstituted recombinant system and cause a dose-dependent inhibition of mtDNA expression and OXPHOS in cell lines. To verify the cellular target, we performed exome sequencing of mutagenized cells and identified a cluster of amino acid substitutions in POLRMT that cause resistance to IMTs. We obtained a cryo-electron microscopy (cryo-EM) structure of POLRMT bound to an IMT, which further defined the allosteric binding site near the active centre cleft of POLRMT. The growth of cancer cells and the persistence of therapy-resistant cancer stem cells has previously been reported to depend on OXPHOS7-17, and we therefore investigated whether IMTs have anti-tumour effects. Four weeks of oral treatment with an IMT is well-tolerated in mice and does not cause OXPHOS dysfunction or toxicity in normal tissues, despite inducing a strong anti-tumour response in xenografts of human cancer cells. In summary, IMTs provide a potent and specific chemical biology tool to study the role of mtDNA expression in physiology and disease.
Collapse
Affiliation(s)
- Nina A Bonekamp
- Department of Mitochondrial Biology, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Bradley Peter
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Hauke S Hillen
- Department of Molecular Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Andrea Felser
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Moritz Horn
- Metabolic and Genetic Regulation of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
- Acus Laboratories, Cologne, Germany
- JLP Health, Vienna, Austria
| | - Anke Unger
- Lead Discovery Center, Dortmund, Germany
| | | | - Ilian Atanassov
- Proteomics Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Xinping Li
- Proteomics Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Uwe Koch
- Lead Discovery Center, Dortmund, Germany
| | | | | | | | - Patrick Giavalisco
- Metabolomics Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Patrick Cramer
- Department of Molecular Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Martin S Denzel
- Metabolic and Genetic Regulation of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Bert Klebl
- Lead Discovery Center, Dortmund, Germany
| | - Maria Falkenberg
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Claes M Gustafsson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden.
| | - Nils-Göran Larsson
- Department of Mitochondrial Biology, Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
- Max Planck Institute for Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
12
|
Wilkinson IVL, Terstappen GC, Russell AJ. Combining experimental strategies for successful target deconvolution. Drug Discov Today 2020; 25:S1359-6446(20)30373-1. [PMID: 32971235 DOI: 10.1016/j.drudis.2020.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/10/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
Investment in phenotypic drug discovery has led to increased demand for rapid and robust target deconvolution to aid successful drug development. Although methods for target identification and mechanism of action (MoA) discovery are flourishing, they typically lead to lists of putative targets. Validating which target(s) are involved in the therapeutic mechanism of a compound poses a significant challenge, requiring direct binding, target engagement, and functional studies in relevant physiological contexts. A combination of orthogonal approaches can allow target identification beyond the proteome as well as aid prioritisation for resource-intensive target validation studies.
Collapse
Affiliation(s)
- Isabel V L Wilkinson
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Georg C Terstappen
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3PQ, UK
| | - Angela J Russell
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3PQ, UK.
| |
Collapse
|
13
|
Allmeroth K, Horn M, Kroef V, Miethe S, Müller RU, Denzel MS. Bortezomib resistance mutations in PSMB5 determine response to second-generation proteasome inhibitors in multiple myeloma. Leukemia 2020; 35:887-892. [PMID: 32690882 PMCID: PMC7932915 DOI: 10.1038/s41375-020-0989-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 01/20/2023]
Affiliation(s)
- Kira Allmeroth
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, D-50931, Cologne, Germany
| | - Moritz Horn
- Acus Laboratories GmbH c/o Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, D-50931, Cologne, Germany
| | - Virginia Kroef
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, D-50931, Cologne, Germany
| | - Stephan Miethe
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, D-50931, Cologne, Germany
| | - Roman-Ulrich Müller
- CECAD-Cluster of Excellence, University of Cologne, Joseph-Stelzmann-Str. 26, D-50931, Cologne, Germany.,Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, D-50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, D-50931, Cologne, Germany
| | - Martin S Denzel
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, D-50931, Cologne, Germany. .,CECAD-Cluster of Excellence, University of Cologne, Joseph-Stelzmann-Str. 26, D-50931, Cologne, Germany. .,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, D-50931, Cologne, Germany.
| |
Collapse
|
14
|
Marchesini M, Gherli A, Montanaro A, Patrizi L, Sorrentino C, Pagliaro L, Rompietti C, Kitara S, Heit S, Olesen CE, Møller JV, Savi M, Bocchi L, Vilella R, Rizzi F, Baglione M, Rastelli G, Loiacono C, La Starza R, Mecucci C, Stegmaier K, Aversa F, Stilli D, Lund Winther AM, Sportoletti P, Bublitz M, Dalby-Brown W, Roti G. Blockade of Oncogenic NOTCH1 with the SERCA Inhibitor CAD204520 in T Cell Acute Lymphoblastic Leukemia. Cell Chem Biol 2020; 27:678-697.e13. [PMID: 32386594 PMCID: PMC7305996 DOI: 10.1016/j.chembiol.2020.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 02/14/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022]
Abstract
The identification of SERCA (sarco/endoplasmic reticulum calcium ATPase) as a target for modulating gain-of-function NOTCH1 mutations in Notch-dependent cancers has spurred the development of this compound class for cancer therapeutics. Despite the innate toxicity challenge associated with SERCA inhibition, we identified CAD204520, a small molecule with better drug-like properties and reduced off-target Ca2+ toxicity compared with the SERCA inhibitor thapsigargin. In this work, we describe the properties and complex structure of CAD204520 and show that CAD204520 preferentially targets mutated over wild-type NOTCH1 proteins in T cell acute lymphoblastic leukemia (T-ALL) and mantle cell lymphoma (MCL). Uniquely among SERCA inhibitors, CAD204520 suppresses NOTCH1-mutated leukemic cells in a T-ALL xenografted model without causing cardiac toxicity. This study supports the development of SERCA inhibitors for Notch-dependent cancers and extends their application to cases with isolated mutations in the PEST degradation domain of NOTCH1, such as MCL or chronic lymphocytic leukemia (CLL).
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Enzyme Inhibitors/chemical synthesis
- Enzyme Inhibitors/chemistry
- Enzyme Inhibitors/pharmacology
- Female
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred ICR
- Mice, Inbred NOD
- Mice, SCID
- Molecular Structure
- Mutation
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Receptor, Notch1/antagonists & inhibitors
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Matteo Marchesini
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Andrea Gherli
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Anna Montanaro
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Laura Patrizi
- University of Perugia, Department of Medicine, Hematology and Clinical Immunology, Perugia 06123, Italy
| | - Claudia Sorrentino
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Luca Pagliaro
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Chiara Rompietti
- University of Perugia, Department of Medicine, Hematology and Clinical Immunology, Perugia 06123, Italy
| | - Samuel Kitara
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Sabine Heit
- University of Oxford, Department of Biochemistry, Oxford OX1 3QU, UK
| | - Claus E Olesen
- Aarhus University, Department of Biomedicine, 8000 Aarhus C, Denmark
| | - Jesper V Møller
- Aarhus University, Department of Biomedicine, 8000 Aarhus C, Denmark
| | - Monia Savi
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parma 43124, Italy
| | - Leonardo Bocchi
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parma 43124, Italy
| | - Rocchina Vilella
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parma 43124, Italy
| | - Federica Rizzi
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy; INBB - Biostructures and Biosystems National Institute, Rome 00136, Italy
| | - Marilena Baglione
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Giorgia Rastelli
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Caterina Loiacono
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Roberta La Starza
- University of Perugia, Department of Medicine, Hematology and Clinical Immunology, Perugia 06123, Italy
| | - Cristina Mecucci
- University of Perugia, Department of Medicine, Hematology and Clinical Immunology, Perugia 06123, Italy
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; The Broad Institute, Cambridge, MA 02142, USA
| | - Franco Aversa
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy
| | - Donatella Stilli
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parma 43124, Italy
| | | | - Paolo Sportoletti
- University of Perugia, Department of Medicine, Hematology and Clinical Immunology, Perugia 06123, Italy
| | - Maike Bublitz
- University of Oxford, Department of Biochemistry, Oxford OX1 3QU, UK
| | | | - Giovanni Roti
- University of Parma, Department of Medicine and Surgery, Parma 43126, Italy.
| |
Collapse
|
15
|
Schmidt FI. From atoms to physiology: what it takes to really understand inflammasomes. J Physiol 2019; 597:5335-5348. [PMID: 31490557 DOI: 10.1113/jp277027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/29/2019] [Indexed: 12/22/2022] Open
Abstract
Rapid inflammatory responses to cytosolic threats are mediated by inflammasomes - large macromolecular signalling complexes that control the activation of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18, as well as cell death by pyroptosis. Different inflammasome sensors are activated by diverse direct and indirect signals, and subsequently nucleate the polymerization of the adaptor molecule ASC to form signalling platforms macroscopically observed as ASC specks. Caspase-1 is autocatalytically activated at these sites and subsequently matures pro-inflammatory cytokines and the pore-forming effector molecule gasdermin D. While most molecules and basic assembly principles have been deduced from reductionist experimental systems, we still lack fundamental information on the structure and regulation of these complexes in their physiological environment and in the interplay with other signalling pathways. In this review, novel experimental approaches are proposed, including some that rely on nanobodies and single domain antibodies, to understand inflammasome assembly and regulation in the context of the relevant tissues or cells.
Collapse
|
16
|
Unbiased Forward Genetic Screening with Chemical Mutagenesis to Uncover Drug-Target Interactions. Methods Mol Biol 2019. [PMID: 30912013 DOI: 10.1007/978-1-4939-9145-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The steadily increasing throughput in next-generation sequencing technologies is revolutionizing a number of fields in biology. One application requiring massive parallel sequencing is forward genetic screening based on chemical mutagenesis. Such screens interrogate the entire genome in an entirely unbiased fashion and can be applied to a number of research questions. CRISPR/Cas9-based screens, which are largely limited to a gene's loss of function, have already been very successful in identifying drug targets and pathways related to the drug's mode of action. By inducing single nucleotide changes using an alkylating reagent, it is possible to generate amino acid changes that perturb the interaction between a drug and its direct target, resulting in drug resistance. This chemogenomic approach combined with latest sequencing technologies allows deconvolution of drug targets and characterization of drug-target binding interfaces at amino acid resolution, therefore nicely complementing existing biochemical approaches. Here we describe a general protocol for a chemical mutagenesis-based forward genetic screen applicable for drug-target deconvolution.
Collapse
|
17
|
Volz JC, Schuller N, Elling U. Using Functional Genetics in Haploid Cells for Drug Target Identification. Methods Mol Biol 2019; 1953:3-21. [PMID: 30912012 DOI: 10.1007/978-1-4939-9145-7_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Pooled genetic screens are a powerful tool to identify targets for drug development as well as chemogenetic interactions. Various complementary methods for mutagenesis are available to generate highly complex cell populations, including mRNA knockdown, directed genome editing, as well as random genome mutagenesis. With the availability of a growing number of haploid mammalian cell lines, random mutagenesis is becoming increasingly powerful and represents an attractive alternative, e.g., to CRISPR-based screening. This chapter provides a step-by-step protocol for performing haploid gene trap screens.
Collapse
Affiliation(s)
- Jennifer C Volz
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Nicole Schuller
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Ulrich Elling
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|