1
|
Soto OB, Ramirez CS, Koyani R, Rodriguez-Palomares IA, Dirmeyer JR, Grajeda B, Roy S, Cox MB. Structure and function of the TPR-domain immunophilins FKBP51 and FKBP52 in normal physiology and disease. J Cell Biochem 2024; 125:e30406. [PMID: 37087733 PMCID: PMC10903107 DOI: 10.1002/jcb.30406] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/22/2023] [Accepted: 04/04/2023] [Indexed: 04/24/2023]
Abstract
Coordinated cochaperone interactions with Hsp90 and associated client proteins are crucial for a multitude of signaling pathways in normal physiology, as well as in disease settings. Research on the molecular mechanisms regulated by the Hsp90 multiprotein complexes has demonstrated increasingly diverse roles for cochaperones throughout Hsp90-regulated signaling pathways. Thus, the Hsp90-associated cochaperones have emerged as attractive therapeutic targets in a wide variety of disease settings. The tetratricopeptide repeat (TPR)-domain immunophilins FKBP51 and FKBP52 are of special interest among the Hsp90-associated cochaperones given their Hsp90 client protein specificity, ubiquitous expression across tissues, and their increasingly important roles in neuronal signaling, intracellular calcium release, peptide bond isomerization, viral replication, steroid hormone receptor function, and cell proliferation to name a few. This review summarizes the current knowledge of the structure and molecular functions of TPR-domain immunophilins FKBP51 and FKBP52, recent findings implicating these immunophilins in disease, and the therapeutic potential of targeting FKBP51 and FKBP52 for the treatment of disease.
Collapse
Affiliation(s)
- Olga B. Soto
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Christian S. Ramirez
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Rina Koyani
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Isela A. Rodriguez-Palomares
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Jessica R. Dirmeyer
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Brian Grajeda
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Sourav Roy
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Marc B. Cox
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968
| |
Collapse
|
2
|
Dreizler JK, Meyners C, Hausch F. Toward Dual Targeting of Catalytic and Gatekeeper Pockets in Cyclophilins Using a Macrocyclic Scaffold. ACS Med Chem Lett 2024; 15:2012-2018. [PMID: 39563809 PMCID: PMC11571008 DOI: 10.1021/acsmedchemlett.4c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024] Open
Abstract
Cyclophilins, especially cyclophilin A, are involved in a variety of diseases, including the life cycle of many viruses. An advanced macrocyclic inhibitor of cyclophilin was reported to bind the catalytic pocket but not the neighboring gatekeeper pocket. Here we describe macrocyclic cyclophilin inhibitors bearing side chains designed to reach out to the gatekeeper pocket. After establishing a suitable synthesis allowing for late-stage modification of the relevant positions, we explored this exit vector. This culminated in a rigid ornithine-resembling analogue as a versatile building block, which was also incorporated into the macrocyclic scaffold. The use of amines as the gatekeeper-engaging modality was invalidated, but the exit vector was successfully established as a promising position for future modifications. Further work is needed to identify suitable motifs to simultaneously engage the catalytic and gatekeeper pockets in this highly developed macrocyclic scaffold.
Collapse
Affiliation(s)
- Johannes K Dreizler
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Christian Meyners
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
| | - Felix Hausch
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg Straße 4, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
3
|
Brudy C, Walz C, Spiske M, Dreizler JK, Hausch F. The Missing Link(er): A Roadmap to Macrocyclization in Drug Discovery. J Med Chem 2024; 67:14768-14785. [PMID: 39171975 DOI: 10.1021/acs.jmedchem.4c01163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Macrocycles are one of nature's preferred choices to generate large but cell-permeable bioactive molecules. Macrocyclization is increasingly prominent in medicinal chemistry beyond natural products, especially for difficult-to-drug targets. However, strategies to best exploit the potential of macrocycles are only beginning to emerge. Here we survey drug discovery campaigns from the past decade that cumulated in advanced macrocyclic drug-like compounds or drug candidates. Most macrocycles were conceived by ring closing based on U- or C-shaped bioactive conformations observed in co-crystal structures. We focus on the key step from linear precursors to the first macrocycle and the follow-up optimization of the resulting macrocyclic scaffold. Conformational control recurrently emerged as a key factor for macrocycle properties and linkers as an opportunity for optimization. With increasingly challenging drug targets, we expect these trends to become more prominent and relevant.
Collapse
Affiliation(s)
- Christian Brudy
- Department of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Carlo Walz
- Department of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Moritz Spiske
- Department of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Johannes K Dreizler
- Department of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Felix Hausch
- Department of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
4
|
Buffa V, Meyners C, Sugiarto WO, Bauder M, Gaali S, Hausch F. 1,4-Pyrazolyl-Containing SAFit-Analogues are Selective FKBP51 Inhibitors With Improved Ligand Efficiency and Drug-Like Profile. ChemMedChem 2024; 19:e202400264. [PMID: 38818693 DOI: 10.1002/cmdc.202400264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
The FK506 binding protein 51 (FKBP51) is an appealing drug target due to its role in several diseases such as depression, anxiety, chronic pain and obesity. Towards this, selectivity versus the close homolog FKBP52 is essential. However, currently available FKBP51-selective ligands such as SAFit2 are too large and lack drug-like properties. Here, we present a structure activity relationship (SAR) analysis of the pipecolic ester moiety of SAFit1 and SAFit2, which culminated in the discovery of the 1,4-pyrazolyl derivative 23 d, displaying a binding affinity of 0.077 μM for FKBP51, reduced molecular weight (541.7 g/mol), lower hydrophobicity (cLogP=3.72) and higher ligand efficiency (LE=0.25). Cocrystal structures revealed the importance of the 1,4- and 1,3,4- substitution patterns of the pyrazole ring versus the 1,4,5 arrangement.
Collapse
Affiliation(s)
- Vanessa Buffa
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287, Darmstadt, Germany
- Present address Dr. Michael Bauder, InfectoPharm Arzneimittel und Consilium GmbH, Von-Humboldt-Str.1, 64646, Heppenheim, Germany
| | - Christian Meyners
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287, Darmstadt, Germany
- Present address Dr. Michael Bauder, InfectoPharm Arzneimittel und Consilium GmbH, Von-Humboldt-Str.1, 64646, Heppenheim, Germany
| | - Wisely Oki Sugiarto
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287, Darmstadt, Germany
- Present address Dr. Michael Bauder, InfectoPharm Arzneimittel und Consilium GmbH, Von-Humboldt-Str.1, 64646, Heppenheim, Germany
| | - Michael Bauder
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287, Darmstadt, Germany
- Present address Dr. Michael Bauder, InfectoPharm Arzneimittel und Consilium GmbH, Von-Humboldt-Str.1, 64646, Heppenheim, Germany
| | - Steffen Gaali
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- Present address Dr. Steffen Gaali, Roche Diagnostics GmbH, Nonnenwald 2, 82377, Penzberg
| | - Felix Hausch
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287, Darmstadt, Germany
- Present address Dr. Michael Bauder, InfectoPharm Arzneimittel und Consilium GmbH, Von-Humboldt-Str.1, 64646, Heppenheim, Germany
- Center for Synthetic Biology, Technical University Darmstadt, Germany
| |
Collapse
|
5
|
Mareczek L, Mueller LK, Halstenberg L, Geiger TM, Walz M, Zheng M, Hausch F. Use of Poly(vinyl alcohol) in Spray-Dried Dispersions: Enhancing Solubility and Stability of Proteolysis Targeting Chimeras. Pharmaceutics 2024; 16:924. [PMID: 39065621 PMCID: PMC11279962 DOI: 10.3390/pharmaceutics16070924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
PROTACs, proteolysis targeting chimeras, are bifunctional molecules inducing protein degradation through a unique proximity-based mode of action. While offering several advantages unachievable by classical drugs, PROTACs have unfavorable physicochemical properties that pose challenges in application and formulation. In this study, we show the solubility enhancement of two PROTACs, ARV-110 and SelDeg51, using Poly(vinyl alcohol). Hereby, we apply a three-fluid nozzle spray drying set-up to generate an amorphous solid dispersion with a 30% w/w drug loading with the respective PROTACs and the hydrophilic polymer. Dissolution enhancement was achieved and demonstrated for t = 0 and t = 4 weeks at 5 °C using a phosphate buffer with a pH of 6.8. A pH shift study on ARV-110-PVA is shown, covering transfer from simulated gastric fluid (SGF) at pH 2.0 to fasted-state simulated intestinal fluid (FaSSIF) at pH 6.5. Additionally, activity studies and binding assays of the pure SelDeg51 versus the spray-dried SelDeg51-PVA indicate no difference between both samples. Our results show how modern enabling formulation technologies can partially alleviate challenging physicochemical properties, such as the poor solubility of increasingly large 'small' molecules.
Collapse
Affiliation(s)
| | | | | | - Thomas M. Geiger
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Michael Walz
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Min Zheng
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Felix Hausch
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
6
|
Agam G, Atawna B, Damri O, Azab AN. The Role of FKBPs in Complex Disorders: Neuropsychiatric Diseases, Cancer, and Type 2 Diabetes Mellitus. Cells 2024; 13:801. [PMID: 38786025 PMCID: PMC11119362 DOI: 10.3390/cells13100801] [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: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Stress is a common denominator of complex disorders and the FK-506 binding protein (FKBP)51 plays a central role in stress. Hence, it is not surprising that multiple studies imply the involvement of the FKBP51 protein and/or its coding gene, FKBP5, in complex disorders. This review summarizes such reports concentrating on three disorder clusters-neuropsychiatric, cancer, and type 2 diabetes mellitus (T2DM). We also attempt to point to potential mechanisms suggested to mediate the effect of FKBP5/FKBP51 on these disorders. Neuropsychiatric diseases considered in this paper include (i) Huntington's disease for which increased autophagic cellular clearance mechanisms related to decreased FKBP51 protein levels or activity is discussed, Alzheimer's disease for which increased FKBP51 activity has been shown to induce Tau phosphorylation and aggregation, and Parkinson's disease in the context of which FKBP12 is mentioned; and (ii) mental disorders, for which significant association with the single nucleotide polymorphism (SNP) rs1360780 of FKBP5 intron 7 along with decreased DNA methylation were revealed. Since cancer is a large group of diseases that can start in almost any organ or tissue of the body, FKBP51's role depends on the tissue type and differences among pathways expressed in those tumors. The FKBP51-heat-shock protein-(Hsp)90-p23 super-chaperone complex might function as an oncogene or as a tumor suppressor by downregulating the serine/threonine protein kinase (AKt) pathway. In T2DM, two potential pathways for the involvement of FKBP51 are highlighted as affecting the pathogenesis of the disease-the peroxisome proliferator-activated receptor-γ (PPARγ) and AKt.
Collapse
Affiliation(s)
- Galila Agam
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience and Zelman Center—The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (B.A.); (O.D.)
| | - Bayan Atawna
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience and Zelman Center—The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (B.A.); (O.D.)
| | - Odeya Damri
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience and Zelman Center—The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (B.A.); (O.D.)
| | - Abed N. Azab
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience and Zelman Center—The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (B.A.); (O.D.)
- Department of Nursing, School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
7
|
Geiger TM, Walz M, Meyners C, Kuehn A, Dreizler JK, Sugiarto WO, Maciel EVS, Zheng M, Lermyte F, Hausch F. Discovery of a Potent Proteolysis Targeting Chimera Enables Targeting the Scaffolding Functions of FK506-Binding Protein 51 (FKBP51). Angew Chem Int Ed Engl 2024; 63:e202309706. [PMID: 37942685 DOI: 10.1002/anie.202309706] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
The FK506-binding protein 51 (FKBP51) is a promising target in a variety of disorders including depression, chronic pain, and obesity. Previous FKBP51-targeting strategies were restricted to occupation of the FK506-binding site, which does not affect core functions of FKBP51. Here, we report the discovery of the first FKBP51 proteolysis targeting chimera (PROTAC) that enables degradation of FKBP51 abolishing its scaffolding function. Initial synthesis of 220 FKBP-focused PROTACs yielded a plethora of active PROTACs for FKBP12, six for FKBP51, and none for FKBP52. Structural analysis of a binary FKBP12:PROTAC complex revealed the molecular basis for negative cooperativity. Linker-based optimization of first generation FKBP51 PROTACs led to the PROTAC SelDeg51 with improved cellular activity, selectivity, and high cooperativity. The structure of the ternary FKBP51:SelDeg51:VCB complex revealed how SelDeg51 establishes cooperativity by dimerizing FKBP51 and the von Hippel-Lindau protein (VHL) in a glue-like fashion. SelDeg51 efficiently depletes FKBP51 and reactivates glucocorticoid receptor (GR)-signalling, highlighting the enhanced efficacy of full protein degradation compared to classical FKBP51 binding.
Collapse
Affiliation(s)
- Thomas M Geiger
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Michael Walz
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Christian Meyners
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Angela Kuehn
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Johannes K Dreizler
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Wisely O Sugiarto
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Edvaldo V S Maciel
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Min Zheng
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Frederik Lermyte
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Felix Hausch
- Department of Chemistry and Biochemistry Clemens-Schöpf-Institute, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64283, Darmstadt, Germany
| |
Collapse
|
8
|
Deutscher RCE, Safa Karagöz M, Purder PL, Kolos JM, Meyners C, Oki Sugiarto W, Krajczy P, Tebbe F, Geiger TM, Ünal C, Hellmich UA, Steinert M, Hausch F. [4.3.1]Bicyclic FKBP Ligands Inhibit Legionella Pneumophila Infection by LpMip-Dependent and LpMip-Independent Mechanisms. Chembiochem 2023; 24:e202300442. [PMID: 37489700 DOI: 10.1002/cbic.202300442] [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: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 07/26/2023]
Abstract
Legionella pneumophila is the causative agent of Legionnaires' disease, a serious form of pneumonia. Its macrophage infectivity potentiator (Mip), a member of a highly conserved family of FK506-binding proteins (FKBPs), plays a major role in the proliferation of the gram-negative bacterium in host organisms. In this work, we test our library of >1000 FKBP-focused ligands for inhibition of LpMip. The [4.3.1]-bicyclic sulfonamide turned out as a highly preferred scaffold and provided the most potent LpMip inhibitors known so far. Selected compounds were non-toxic to human cells, displayed antibacterial activity and block bacterial proliferation in cellular infection-assays as well as infectivity in human lung tissue explants. The results confirm [4.3.1]-bicyclic sulfonamides as anti-legionellal agents, although their anti-infective properties cannot be explained by inhibition of LpMip alone.
Collapse
Affiliation(s)
- Robin C E Deutscher
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - M Safa Karagöz
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Patrick L Purder
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Jürgen M Kolos
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Christian Meyners
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Wisely Oki Sugiarto
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Patryk Krajczy
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Frederike Tebbe
- Institute of Organic Chemistry & Macromolecular Chemistry (IOMC), Friedrich Schiller University Germany, Humboldtstraße 10, 07743, Jena, Germany
| | - Thomas M Geiger
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
| | - Can Ünal
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Ute A Hellmich
- Institute of Organic Chemistry & Macromolecular Chemistry (IOMC), Friedrich Schiller University Germany, Humboldtstraße 10, 07743, Jena, Germany
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Max-von-Laue-Str. 9, 60438, Frankurt/Main, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Michael Steinert
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
- Helmholtz Centre for Infection Research, 38106, Braunschweig, Germany
| | - Felix Hausch
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64287, Darmstadt, Germany
| |
Collapse
|
9
|
Buffa V, Knaup FH, Heymann T, Springer M, Schmidt MV, Hausch F. Analysis of the Selective Antagonist SAFit2 as a Chemical Probe for the FK506-Binding Protein 51. ACS Pharmacol Transl Sci 2023; 6:361-371. [PMID: 36926456 PMCID: PMC10012253 DOI: 10.1021/acsptsci.2c00234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Indexed: 02/16/2023]
Abstract
The FK506-binding protein 51 (FKBP51) has emerged as an important regulator of the mammalian stress response and is involved in persistent pain states and metabolic pathways. The FK506 analog SAFit2 (short for selective antagonist of FKBP51 by induced fit) was the first potent and selective FKBP51 ligand with an acceptable pharmacokinetic profile. At present, SAFit2 represents the gold standard for FKBP51 pharmacology and has been extensively used in numerous biological studies. Here we review the current knowledge on SAFit2 as well as guidelines for its use.
Collapse
Affiliation(s)
- Vanessa Buffa
- Department
of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287 Darmstadt, Germany
| | - Fabian H. Knaup
- Department
of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287 Darmstadt, Germany
| | - Tim Heymann
- Department
of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287 Darmstadt, Germany
| | - Margherita Springer
- Research
Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Mathias V. Schmidt
- Research
Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Felix Hausch
- Department
of Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss Straße 4, 64287 Darmstadt, Germany
| |
Collapse
|
10
|
Quist-Løkken I, Andersson-Rusch C, Kastnes MH, Kolos JM, Jatzlau J, Hella H, Olsen OE, Sundan A, Knaus P, Hausch F, Holien T. FKBP12 is a major regulator of ALK2 activity in multiple myeloma cells. Cell Commun Signal 2023; 21:25. [PMID: 36717825 PMCID: PMC9885706 DOI: 10.1186/s12964-022-01033-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/28/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The immunophilin FKBP12 binds to TGF-β family type I receptors, including the BMP type I receptor ALK2. FKBP12 keeps the type I receptor in an inactive state and controls signaling activity. Removal of FKBP12 with drugs such as the FKBP-ligand FK506 enhances BMP activity in various cell types. In multiple myeloma cells, activation of SMAD1/5/8 leads to apoptosis. We hypothesized that removing FKBP12 from ALK2 in myeloma cells would potentiate BMP-induced ALK2-SMAD1/5/8 activity and in consequence cell death. METHODS Multiple myeloma cell lines were treated with FK506, or other FKBP-binding compounds, combined with different BMPs before analyzing SMAD1/5/8 activity and cell viability. SMAD1/5/8 activity was also investigated using a reporter cell line, INA-6 BRE-luc. To characterize the functional signaling receptor complex, we genetically manipulated receptor expression by siRNA, shRNA and CRISPR/Cas9 technology. RESULTS FK506 potentiated BMP-induced SMAD1/5/8 activation and apoptosis in multiple myeloma cell lines. By using FKBP-binding compounds with different affinity profiles, and siRNA targeting FKBP12, we show that the FK506 effect is mediated by binding to FKBP12. Ligands that typically signal via ALK3 in myeloma cells, BMP2, BMP4, and BMP10, did not induce apoptosis in cells lacking ALK3. Notably, BMP10 competed with BMP6 and BMP9 and antagonized their activity via ALK2. However, upon addition of FK506, we saw a surprising shift in specificity, as the ALK3 ligands gained the ability to signal via ALK2 and induce apoptosis. This indicates that the receptor complex can switch from an inactive non-signaling complex (NSC) to an active one by adding FK506. This gain of activity was also seen in other cell types, indicating that the observed effects have broader relevance. BMP2, BMP4 and BMP10 depended on BMPR2 as type II receptor to signal, which contrasts with BMP6 and BMP9, that activate ALK2 more potently when BMPR2 is knocked down. CONCLUSIONS In summary, our data suggest that FKBP12 is a major regulator of ALK2 activity in multiple myeloma cells, partly by switching an NSC into an active signaling complex. FKBP12 targeting compounds devoid of immunosuppressing activity could have potential in novel treatment strategies aiming at reducing multiple myeloma tumor load. Video Abstract.
Collapse
Affiliation(s)
- Ingrid Quist-Løkken
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Clara Andersson-Rusch
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Martin Haugrud Kastnes
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Centre of Molecular Inflammation Research, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Jürgen Markus Kolos
- grid.6546.10000 0001 0940 1669Department of Chemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jerome Jatzlau
- grid.14095.390000 0000 9116 4836Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Hanne Hella
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Oddrun Elise Olsen
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Anders Sundan
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Centre of Molecular Inflammation Research, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Hematology, St. Olav’s University Hospital, Trondheim, Norway
| | - Petra Knaus
- grid.14095.390000 0000 9116 4836Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Felix Hausch
- grid.6546.10000 0001 0940 1669Department of Chemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Toril Holien
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Immunology and Transfusion Medicine, St. Olav’s University Hospital, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Hematology, St. Olav’s University Hospital, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Department of Biomedical Laboratory Science, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| |
Collapse
|
11
|
Lerma Romero JA, Meyners C, Rupp N, Hausch F, Kolmar H. A protein engineering approach toward understanding FKBP51 conformational dynamics and mechanisms of ligand binding. Protein Eng Des Sel 2023; 36:gzad014. [PMID: 37903068 DOI: 10.1093/protein/gzad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/15/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
Most proteins are flexible molecules that coexist in an ensemble of several conformations. Point mutations in the amino acid sequence of a protein can trigger structural changes that drive the protein population to a conformation distinct from the native state. Here, we report a protein engineering approach to better understand protein dynamics and ligand binding of the FK506-binding protein 51 (FKBP51), a prospective target for stress-related diseases, metabolic disorders, some types of cancers and chronic pain. By randomizing selected regions of its ligand-binding domain and sorting yeast display libraries expressing these variants, mutants with high affinity to conformation-specific FKBP51 selective ligands were identified. These improved mutants are valuable tools for the discovery of novel selective ligands that preferentially and specifically bind the FKBP51 active site in its open conformation state. Moreover, they will help us understand the conformational dynamics and ligand binding mechanics of the FKBP51 binding pocket.
Collapse
Affiliation(s)
- Jorge A Lerma Romero
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt 64287, Germany
| | - Christian Meyners
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt 64287, Germany
| | - Nicole Rupp
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt 64287, Germany
| | - Felix Hausch
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt 64287, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt 64287, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt 64287, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt 64287, Germany
| |
Collapse
|
12
|
Lerma Romero JA, Meyners C, Christmann A, Reinbold LM, Charalampidou A, Hausch F, Kolmar H. Binding pocket stabilization by high-throughput screening of yeast display libraries. Front Mol Biosci 2022; 9:1023131. [DOI: 10.3389/fmolb.2022.1023131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
Protein dynamics have a great influence on the binding pockets of some therapeutic targets. Flexible protein binding sites can result in transient binding pocket formation which might have a negative impact on drug screening efforts. Here, we describe a protein engineering strategy with FK506-binding protein 51 (FKBP51) as a model protein, which is a promising target for stress-related disorders. High-throughput screening of yeast display libraries of FKBP51 resulted in the identification of variants exhibiting higher affinity binding of conformation-specific FKBP51 selective inhibitors. The gene libraries of a random mutagenesis and site saturation mutagenesis of the FK1 domain of FKBP51 encoding sequence were used to create a yeast surface display library. Fluorescence-activated cell sorting for FKBP51 variants that bind conformation-specific fluorescently labeled ligands with high affinity allowed for the identification of 15 different protein variants with improved binding to either, or both FKBP51-specific ligands used in the screening, with improved affinities up to 34-fold compared to the wild type. These variants will pave the way to a better understanding of the conformational flexibility of the FKBP51 binding pocket and may enable the isolation of new selective ligands that preferably and selectively bind the active site of the protein in its open conformation state.
Collapse
|
13
|
Diamanti E, Méndez M, Ross T, Kuttruff CA, Lefranc J, Klingler FM, von Nussbaum F, Jung M, Gehringer M. Frontiers in Medicinal Chemistry 2022 Goes Virtual. ChemMedChem 2022; 17:e202200419. [PMID: 36198574 DOI: 10.1002/cmdc.202200419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Indexed: 11/09/2022]
Abstract
The Frontiers in Medicinal Chemistry (FiMC) meeting, which represents the largest international medicinal chemistry conference in Germany, took place from March 14th to 16th 2022 in a fully virtual format. Organized by the Division of Medicinal Chemistry of the German Chemical Society (GDCh) together with the Division of Pharmaceutical & Medicinal Chemistry of the German Pharmaceutical Society (DPhG) and a "local" organization committee from the University of Freiburg headed by Manfred Jung, the meeting brought together 271 participants from around 20 countries. The program included 33 lectures by leading scientists from industry and academia as well as early career investigators. 67 posters were presented in two poster sessions and with over 20.000 poster abstract downloads. The general organization and the time-shift function were very much appreciated as demonstrated by almost 600 on-demand contents retrieved. The online format fitted perfectly to bring together medicinal chemists from academia and industry across the globe.
Collapse
Affiliation(s)
- Eleonora Diamanti
- HIPS - Helmholtz-Institut für Pharmazeutische Forschung Saarland, Campus E8 1, 66123, Saarbrücken, Germany
| | - María Méndez
- Sanofi R&D, Integrated Drug Discovery, Industriepark Höchst, Blg. G838, 65926, Frankfurt am Main, Germany
| | - Tatjana Ross
- Merck Healthcare KGaA, Frankfurter Straße 250, 64293, Darmstadt, Germany
| | - Christian A Kuttruff
- Boehringer Ingelheim International GmbH, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Julien Lefranc
- Merck Healthcare KGaA, Frankfurter Straße 250, 64293, Darmstadt, Germany
| | | | - Franz von Nussbaum
- NUVISAN Innovation Campus Berlin, NUVISAN ICB GmbH, Muellerstr. 178, 13353, Berlin, Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104, Freiburg im Breisgau, Germany
| | - Matthias Gehringer
- University of Tübingen, Institute of Pharmaceutical Sciences, Pharmaceutical/Medicinal Chemistry Department, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| |
Collapse
|
14
|
Kolos JM, Pomplun S, Jung S, Rieß B, Purder PL, Voll AM, Merz S, Gnatzy M, Geiger TM, Quist-Løkken I, Jatzlau J, Knaus P, Holien T, Bracher A, Meyners C, Czodrowski P, Krewald V, Hausch F. Picomolar FKBP inhibitors enabled by a single water-displacing methyl group in bicyclic [4.3.1] aza-amides. Chem Sci 2021; 12:14758-14765. [PMID: 34820091 PMCID: PMC8597852 DOI: 10.1039/d1sc04638a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/22/2021] [Indexed: 01/30/2023] Open
Abstract
Methyl groups can have profound effects in drug discovery but the underlying mechanisms are diverse and incompletely understood. Here we report the stereospecific effect of a single, solvent-exposed methyl group in bicyclic [4.3.1] aza-amides, robustly leading to a 2 to 10-fold increase in binding affinity for FK506-binding proteins (FKBPs). This resulted in the most potent and efficient FKBP ligands known to date. By a combination of co-crystal structures, isothermal titration calorimetry (ITC), density-functional theory (DFT), and 3D reference interaction site model (3D-RISM) calculations we elucidated the origin of the observed affinity boost, which was purely entropically driven and relied on the displacement of a water molecule at the protein-ligand-bulk solvent interface. The best compounds potently occupied FKBPs in cells and enhanced bone morphogenic protein (BMP) signaling. Our results show how subtle manipulation of the solvent network can be used to design atom-efficient ligands for difficult, solvent-exposed binding pockets.
Collapse
Affiliation(s)
- Jürgen M Kolos
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany .,Max Planck Institute of Psychiatry Kraepelinstr. 2-10 80804 München Germany
| | - Sebastian Pomplun
- Max Planck Institute of Psychiatry Kraepelinstr. 2-10 80804 München Germany
| | - Sascha Jung
- Technische Universität Dortmund, Fakultät für Chemie und Chemische Biologie Otto-Hahn-Straße 6 44227 Dortmund Germany
| | - Benedikt Rieß
- Max Planck Institute of Psychiatry Kraepelinstr. 2-10 80804 München Germany
| | - Patrick L Purder
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| | - Andreas M Voll
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| | - Stephanie Merz
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| | - Monika Gnatzy
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| | - Thomas M Geiger
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| | - Ingrid Quist-Løkken
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology 7491 Trondheim Norway.,Department of Immunology and Transfusion Medicine, St. Olav's University Hospital 7030 Trondheim Norway.,Department of Hematology, St. Olav's University Hospital 7030 Trondheim Norway
| | - Jerome Jatzlau
- Institute for Chemistry and Biochemistry, Freie Universität Berlin 14195 Berlin Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin 14195 Berlin Germany
| | - Toril Holien
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology 7491 Trondheim Norway.,Department of Immunology and Transfusion Medicine, St. Olav's University Hospital 7030 Trondheim Norway.,Department of Hematology, St. Olav's University Hospital 7030 Trondheim Norway
| | - Andreas Bracher
- Research Department Cellular Biochemistry, Max Planck Institute of Biochemistry Am Klopferspitz 18, 82152 Planegg Germany
| | - Christian Meyners
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| | - Paul Czodrowski
- Technische Universität Dortmund, Fakultät für Chemie und Chemische Biologie Otto-Hahn-Straße 6 44227 Dortmund Germany
| | - Vera Krewald
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| | - Felix Hausch
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Straße 4 64293 Darmstadt Germany
| |
Collapse
|
15
|
Fenton-Chemistry-Based Oxidative Modification of Proteins Reflects Their Conformation. Int J Mol Sci 2021; 22:ijms22189927. [PMID: 34576105 PMCID: PMC8469487 DOI: 10.3390/ijms22189927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 11/25/2022] Open
Abstract
In order to understand protein structure to a sufficient extent for, e.g., drug discovery, no single technique can provide satisfactory information on both the lowest-energy conformation and on dynamic changes over time (the ‘four-dimensional’ protein structure). Instead, a combination of complementary techniques is required. Mass spectrometry methods have shown promise in addressing protein dynamics, but often rely on the use of high-end commercial or custom instruments. Here, we apply well-established chemistry to conformation-sensitive oxidative protein labelling on a timescale of a few seconds, followed by analysis through a routine protein analysis workflow. For a set of model proteins, we show that site selectivity of labelling can indeed be rationalised in terms of known structural information, and that conformational changes induced by ligand binding are reflected in the modification pattern. In addition to conventional bottom-up analysis, further insights are obtained from intact mass measurement and native mass spectrometry. We believe that this method will provide a valuable and robust addition to the ‘toolbox’ of mass spectrometry researchers studying higher-order protein structure.
Collapse
|
16
|
Gnatzy MT, Geiger TM, Kuehn A, Gutfreund N, Walz M, Kolos JM, Hausch F. Development of NanoBRET-Binding Assays for FKBP-Ligand Profiling in Living Cells. Chembiochem 2021; 22:2257-2261. [PMID: 33887102 PMCID: PMC8360185 DOI: 10.1002/cbic.202100113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/20/2021] [Indexed: 11/11/2022]
Abstract
FK506-binding proteins (FKBPs) are promising targets for a variety of disorders and infectious diseases. High FKBP occupancy is thought to be necessary for ligands to effectively compete with the endogenous intracellular functions of FKBPs. Here, we report the development of NanoBRET assays for the most prominent cytosolic FKBPs, FKBP12, 12.6, 51 and 52. These assays allowed rapid profiling of FKBP ligands for target engagement and selectivity in living cells. These assays confirmed the selectivity of SAFit-type ligands for FKBP51 over FKBP52 but revealed a substantial offset for the intracellular activity of these ligands compared to bicyclic ligands or natural products. Our results stress the importance to control for intracellular FKBP occupancy and provide the assays to guide further FKBP ligand optimization.
Collapse
Affiliation(s)
- Monika T. Gnatzy
- Department Chemistry and Biochemistry Clemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Straße 464287DarmstadtGermany
| | - Thomas M. Geiger
- Department Chemistry and Biochemistry Clemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Straße 464287DarmstadtGermany
| | - Angela Kuehn
- Department Chemistry and Biochemistry Clemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Straße 464287DarmstadtGermany
| | - Niklas Gutfreund
- Institute of Biophysical ChemistryCenter for Biomolecular Magnetic Resonance andCluster of Excellence Macromolecular Complexes (CEF)Goethe University FrankfurtMax-von-Laue-Straße 960438Frankfurt am MainGermany
| | - Michael Walz
- Department Chemistry and Biochemistry Clemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Straße 464287DarmstadtGermany
| | - Jürgen M. Kolos
- Department Chemistry and Biochemistry Clemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Straße 464287DarmstadtGermany
| | - Felix Hausch
- Department Chemistry and Biochemistry Clemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Straße 464287DarmstadtGermany
| |
Collapse
|