1
|
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
|
2
|
Namiot ED, Smirnovová D, Sokolov AV, Chubarev VN, Tarasov VV, Schiöth HB. Depression clinical trials worldwide: a systematic analysis of the ICTRP and comparison with ClinicalTrials.gov. Transl Psychiatry 2024; 14:315. [PMID: 39085220 PMCID: PMC11291508 DOI: 10.1038/s41398-024-03031-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Major depressive disorder (MDD), commonly known as depression, affects over 300 million people worldwide as of 2018 and presents a wide range of clinical symptoms. The international clinical trials registry platform (ICTRP) introduced by WHO includes aggregated data from ClinicalTrials.gov and 17 other national registers, making it the largest clinical trial platform. Here we analysed data in ICTRP with the aim of providing comprehensive insights into clinical trials on depression. Applying a novel hidden duplicate identification method, 10,606 depression trials were identified in ICTRP, with ANZCTR being the largest non- ClinicalTrials.gov database at 1031 trials, followed by IRCT with 576 trials, ISRCTN with 501 trials, CHiCTR with 489 trials, and EUCTR with 351 trials. The top four most studied drugs, ketamine, sertraline, duloxetine, and fluoxetine, were consistent in both groups, but ClinicalTrials.gov had more trials for each drug compared to the non-ClinicalTrials.gov group. Out of 9229 interventional trials, 663 unique agents were identified, including approved drugs (74.5%), investigational drugs (23.2%), withdrawn drugs (1.8%), nutraceuticals (0.3%), and illicit substances (0.2%). Both ClinicalTrials.gov and non-ClinicalTrials.gov databases revealed that the largest categories were antidepressive agents (1172 in ClinicalTrials.gov and 659 in non-ClinicalTrials.gov) and nutrients, amino acids, and chemical elements (250 in ClinicalTrials.gov and 659 in non-ClinicalTrials.gov), indicating a focus on alternative treatments involving dietary supplements and nutrients. Additionally, 26 investigational antidepressive agents targeting 16 different drug targets were identified, with buprenorphine (opioid agonist), saredutant (NK2 antagonist), and seltorexant (OX2 antagonist) being the most frequently studied. This analysis addresses 40 approved drugs for depression treatment including new drug classes like GABA modulators and NMDA antagonists that are offering new prospects for treating MDD, including drug-resistant depression and postpartum depression subtypes.
Collapse
Affiliation(s)
- Eugenia D Namiot
- Department of Surgical Science, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Diana Smirnovová
- Department of Surgical Science, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Aleksandr V Sokolov
- Department of Surgical Science, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Vladimir N Chubarev
- Advanced Molecular Technologies, Limited Liability Company (LLC), Moscow, Russia
| | - Vadim V Tarasov
- Advanced Molecular Technologies, Limited Liability Company (LLC), Moscow, Russia
| | - Helgi B Schiöth
- Department of Surgical Science, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
3
|
Darlami O, Pun R, Ahn SH, Kim SH, Shin D. Macrocyclization strategy for improving candidate profiles in medicinal chemistry. Eur J Med Chem 2024; 272:116501. [PMID: 38754142 DOI: 10.1016/j.ejmech.2024.116501] [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: 04/06/2024] [Revised: 05/12/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
Macrocycles are defined as cyclic compounds with 12 or more members. In medicinal chemistry, they are categorized based on their core chemistry into cyclic peptides and macrocycles. Macrocycles are advantageous because of their structural diversity and ability to achieve high affinity and selectivity towards challenging targets that are often not addressable by conventional small molecules. The potential of macrocyclization to optimize drug-like properties while maintaining adequate bioavailability and permeability has been emphasized as a key innovation in medicinal chemistry. This review provides a detailed case study of the application of macrocyclization over the past 5 years, starting from the initial analysis of acyclic active compounds to optimization of the resulting macrocycles for improved efficacy and drug-like properties. Additionally, it illustrates the strategic value of macrocyclization in contemporary drug discovery efforts.
Collapse
Affiliation(s)
- Om Darlami
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon, 21935, Republic of Korea
| | - Rabin Pun
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon, 21935, Republic of Korea
| | - Sung-Hoon Ahn
- College of Pharmacy, Kangwon National University, Gangwondaehak-gil 1, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Seok-Ho Kim
- College of Pharmacy, Kangwon National University, Gangwondaehak-gil 1, Chuncheon, Gangwon-do, 24341, Republic of Korea.
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon, 21935, Republic of Korea.
| |
Collapse
|
4
|
Nagpal J, Eachus H, Lityagina O, Ryu S. Optogenetic induction of chronic glucocorticoid exposure in early-life leads to blunted stress-response in larval zebrafish. Eur J Neurosci 2024; 59:3134-3146. [PMID: 38602078 DOI: 10.1111/ejn.16301] [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: 04/01/2023] [Revised: 11/29/2023] [Accepted: 02/19/2024] [Indexed: 04/12/2024]
Abstract
Early life stress (ELS) exposure alters stress susceptibility in later life and affects vulnerability to stress-related disorders, but how ELS changes the long-lasting responsiveness of the stress system is not well understood. Zebrafish provides an opportunity to study conserved mechanisms underlying the development and function of the stress response that is regulated largely by the neuroendocrine hypothalamus-pituitary-adrenal/interrenal (HPA/I) axis, with glucocorticoids (GC) as the final effector. In this study, we established a method to chronically elevate endogenous GC levels during early life in larval zebrafish. To this end, we employed an optogenetic actuator, beggiatoa photoactivated adenylyl cyclase, specifically expressed in the interrenal cells of zebrafish and demonstrate that its chronic activation leads to hypercortisolaemia and dampens the acute-stress evoked cortisol levels, across a variety of stressor modalities during early life. This blunting of stress-response was conserved in ontogeny at a later developmental stage. Furthermore, we observe a strong reduction of proopiomelanocortin (pomc)-expression in the pituitary as well as upregulation of fkbp5 gene expression. Going forward, we propose that this model can be leveraged to tease apart the mechanisms underlying developmental programming of the HPA/I axis by early-life GC exposure and its implications for vulnerability and resilience to stress in adulthood.
Collapse
Affiliation(s)
- Jatin Nagpal
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- APC Microbiome Ireland and School of Pharmacy and Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Olga Lityagina
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Soojin Ryu
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 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
|
6
|
Barrios-Palacios D, Organista-Nava J, Balandrán JC, Alarcón-Romero LDC, Zubillaga-Guerrero MI, Illades-Aguiar B, Rivas-Alarcón AA, Diaz-Lucas JJ, Gómez-Gómez Y, Leyva-Vázquez MA. The Role of miRNAs in Childhood Acute Lymphoblastic Leukemia Relapse and the Associated Molecular Mechanisms. Int J Mol Sci 2023; 25:119. [PMID: 38203290 PMCID: PMC10779195 DOI: 10.3390/ijms25010119] [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: 11/28/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children worldwide. Although ALL patients' overall survival rates in wealthy countries currently surpass 80%, 15-20% of patients still experience relapse. The underlying mechanisms of relapse are still not fully understood, and little progress has been made in treating refractory or relapsed disease. Disease relapse and treatment failure are common causes of leukemia-related death. In ALL relapse, several gene signatures have been identified, but it is also important to study miRNAs involved in ALL relapse in an effort to avoid relapse and to achieve better survival rates since miRNAs regulate target genes that participate in signaling pathways involved in relapse, such as those related to drug resistance, survival signals, and antiapoptotic mechanisms. Several miRNAs, such as miR-24, miR-27a, miR-99/100, miR-124, miR-1225b, miR-128b, miR-142-3p, miR-155 and miR-335-3p, are valuable biomarkers for prognosis and treatment response in ALL patients. Thus, this review aimed to analyze the primary miRNAs involved in pediatric ALL relapse and explore the underlying molecular mechanisms in an effort to identify miRNAs that may be potential candidates for anti-ALL therapy soon.
Collapse
Affiliation(s)
- Dalia Barrios-Palacios
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (D.B.-P.); (J.O.-N.); (B.I.-A.); (A.A.R.-A.); (J.J.D.-L.)
| | - Jorge Organista-Nava
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (D.B.-P.); (J.O.-N.); (B.I.-A.); (A.A.R.-A.); (J.J.D.-L.)
| | - Juan Carlos Balandrán
- Department of Pathology and Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA;
| | - Luz del Carmen Alarcón-Romero
- Laboratorio de Citopatología, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (L.d.C.A.-R.); (M.I.Z.-G.)
| | - Ma Isabel Zubillaga-Guerrero
- Laboratorio de Citopatología, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (L.d.C.A.-R.); (M.I.Z.-G.)
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (D.B.-P.); (J.O.-N.); (B.I.-A.); (A.A.R.-A.); (J.J.D.-L.)
| | - Alinne Ayulieth Rivas-Alarcón
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (D.B.-P.); (J.O.-N.); (B.I.-A.); (A.A.R.-A.); (J.J.D.-L.)
| | - Jessica Julieth Diaz-Lucas
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (D.B.-P.); (J.O.-N.); (B.I.-A.); (A.A.R.-A.); (J.J.D.-L.)
| | - Yazmín Gómez-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (D.B.-P.); (J.O.-N.); (B.I.-A.); (A.A.R.-A.); (J.J.D.-L.)
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico; (D.B.-P.); (J.O.-N.); (B.I.-A.); (A.A.R.-A.); (J.J.D.-L.)
| |
Collapse
|
7
|
Wedel S, Hahnefeld L, Schreiber Y, Namendorf C, Heymann T, Uhr M, Schmidt MV, de Bruin N, Hausch F, Thomas D, Geisslinger G, Sisignano M. SAFit2 ameliorates paclitaxel-induced neuropathic pain by reducing spinal gliosis and elevating pro-resolving lipid mediators. J Neuroinflammation 2023; 20:149. [PMID: 37355700 DOI: 10.1186/s12974-023-02835-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Chemotherapy-induced neuropathic pain (CIPN) describes a pathological pain state that occurs dose-dependently as a side effect and can limit or even impede an effective cancer therapy. Unfortunately, current treatment possibilities for CIPN are remarkably confined and mostly inadequate as CIPN therapeutics themselves consist of low effectiveness and may induce severe side effects, pointing out CIPN as pathological entity with an emerging need for novel treatment targets. Here, we investigated whether the novel and highly specific FKBP51 inhibitor SAFit2 reduces paclitaxel-induced neuropathic pain. METHODS In this study, we used a well-established multiple low-dose paclitaxel model to investigate analgesic and anti-inflammatory properties of SAFit2. For this purpose, the behavior of the mice was recorded over 14 days and the mouse tissue was then analyzed using biochemical methods. RESULTS Here, we show that SAFit2 is capable to reduce paclitaxel-induced mechanical hypersensitivity in mice. In addition, we detected that SAFit2 shifts lipid levels in nervous tissue toward an anti-inflammatory and pro-resolving lipid profile that counteracts peripheral sensitization after paclitaxel treatment. Furthermore, SAFit2 reduced the activation of astrocytes and microglia in the spinal cord as well as the levels of pain-mediating chemokines. Its treatment also increased anti-inflammatory cytokines levels in neuronal tissues, ultimately leading to a resolution of neuroinflammation. CONCLUSIONS In summary, SAFit2 shows antihyperalgesic properties as it ameliorates paclitaxel-induced neuropathic pain by reducing peripheral sensitization and resolving neuroinflammation. Therefore, we consider SAFit2 as a potential novel drug candidate for the treatment of paclitaxel-induced neuropathic pain.
Collapse
Affiliation(s)
- Saskia Wedel
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Christian Namendorf
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Tim Heymann
- Department of Biochemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Manfred Uhr
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Mathias V Schmidt
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Natasja de Bruin
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Felix Hausch
- Department of Biochemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany.
| |
Collapse
|
8
|
Baischew A, Engel S, Geiger TM, Taubert MC, Hausch F. Structural and biochemical insights into FKBP51 as a Hsp90 co-chaperone. J Cell Biochem 2023. [PMID: 36791213 DOI: 10.1002/jcb.30384] [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: 11/04/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
The FK506-binding protein 51 (FKBP51) is a high-molecular-weight immunophilin that emerged as an important drug target for stress-related disorders, chronic pain, and obesity. It has been implicated in a plethora of molecular pathways but remains best characterized as a co-chaperone of Hsp90 in the steroid hormone receptor (SHR) maturation cycle. However, the mechanistic and structural basis for the regulation of SHRs by FKBP51 and the usually antagonistic function compared with its closest homolog FKBP52 remains enigmatic. Here we review recent structural and biochemical studies of FKBPs as regulators in the Hsp90 machinery. These advances provide important insights into the roles of FKBP51 and FKBP52 in SHR regulation.
Collapse
Affiliation(s)
- Asat Baischew
- Department of Chemistry, Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Sarah Engel
- Department of Chemistry, Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Thomas M Geiger
- Department of Chemistry, Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Martha C Taubert
- Department of Chemistry, Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Felix Hausch
- Department of Chemistry, Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| |
Collapse
|
9
|
Fang T, Liu MN, Tian XY, Lu GY, Li F, Zhang X, Liu F, Hao W, Wu N, Li H, Li J. The association of FKBP5 polymorphisms with the severity of depressive disorder in patients with methamphetamine use disorders. Front Psychiatry 2023; 14:1147060. [PMID: 37051166 PMCID: PMC10083280 DOI: 10.3389/fpsyt.2023.1147060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/28/2023] [Indexed: 04/14/2023] Open
Abstract
Background Co-occurring depressive disorder (DD) in patients of methamphetamine use disorder (MAUD) impacts the diagnosis, treatment, and prognosis of the disease. Although FKBP5 has been associated with a variety of psychiatric disorders, whether FKBP5 influences depression susceptibility in MAUD is unknown so far. Methods Here, we sequenced six FKBP5 single-nucleotide polymorphism (SNP) sites (rs4713916, rs6926133, rs9470080, rs737054, rs4713902, and rs9470079) in 282 methamphetamine users. MAUD and DD were evaluated by clinical questionnaires. SPSS was used to analyze the relationship between FKBP5 SNPs and DD in individuals with MAUD. Results Of the 282 methamphetamine users, 161 individuals met the MAUD criteria, and among them, 50 patients (31.1%) had DD co-occurring. Importantly, the incidence of DD in individuals with MAUD was 3.314 times greater than that of the methamphetamine users who did not meet the MAUD criteria (p < 0.001). Although none of the six SNPs of FKBP5 were correlated with the co-occurrence of DD in the population with MAUD, two FKBP5 alleles (rs4713916A and rs6926133A) were substantially associated with the higher DD scores in patients with MAUD (p < 0.05). Moreover, those with the two risk alleles do not have much higher scores than those with a single risk allele, and the strong linkage disequilibrium of the two SNPs may be the underlying cause of this result. Despite having weak linkage disequilibrium with either rs4713916 or rs6926133, FKBP5 rs9470079 became risky when paired with either. Conclusion The results of this study revealed that the FKBP5 risk alleles (rs4713916A and rs6926133A) were associated with a greater probability of severe DD in patients with MAUD. These findings here would help with the development of biological early warning markers and the creation of personalized treatment strategies for MAUD.
Collapse
Affiliation(s)
- Ting Fang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meng-Nan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiao-Yu Tian
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Guan-Yi Lu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Fei Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaojie Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Feng Liu
- Compulsory Detoxification Center of Changsha Public Security Bureau, Changsha, Hunan, China
| | - Wei Hao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ning Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hong Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Hong Li
| | - Jin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Jin Li
| |
Collapse
|
10
|
Codagnone MG, Kara N, Ratsika A, Levone BR, van de Wouw M, Tan LA, Cunningham JI, Sanchez C, Cryan JF, O'Leary OF. Inhibition of FKBP51 induces stress resilience and alters hippocampal neurogenesis. Mol Psychiatry 2022; 27:4928-4938. [PMID: 36104438 PMCID: PMC9763121 DOI: 10.1038/s41380-022-01755-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 07/07/2022] [Accepted: 08/15/2022] [Indexed: 01/19/2023]
Abstract
Stress-related psychiatric disorders such as depression are among the leading causes of morbidity and mortality. Considering that many individuals fail to respond to currently available antidepressant drugs, there is a need for antidepressants with novel mechanisms. Polymorphisms in the gene encoding FK506-binding protein 51 (FKBP51), a co-chaperone of the glucocorticoid receptor, have been linked to susceptibility to stress-related psychiatric disorders. Whether this protein can be targeted for their treatment remains largely unexplored. The aim of this work was to investigate whether inhibition of FKBP51 with SAFit2, a novel selective inhibitor, promotes hippocampal neuron outgrowth and neurogenesis in vitro and stress resilience in vivo in a mouse model of chronic psychosocial stress. Primary hippocampal neuronal cultures or hippocampal neural progenitor cells (NPCs) were treated with SAFit2 and neuronal differentiation and cell proliferation were analyzed. Male C57BL/6 mice were administered SAFit2 while concurrently undergoing a chronic stress paradigm comprising of intermittent social defeat and overcrowding, and anxiety and depressive -related behaviors were evaluated. SAFit2 increased neurite outgrowth and number of branch points to a greater extent than brain derived neurotrophic factor (BDNF) in primary hippocampal neuronal cultures. SAFit2 increased hippocampal NPC neurogenesis and increased neurite complexity and length of these differentiated neurons. In vivo, chronic SAFit2 administration prevented stress-induced social avoidance, decreased anxiety in the novelty-induced hypophagia test, and prevented stress-induced anxiety in the open field but did not alter adult hippocampal neurogenesis in stressed animals. These data warrant further exploration of inhibition of FKBP51 as a strategy to treat stress-related disorders.
Collapse
Affiliation(s)
- Martin G Codagnone
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Instituto de Biología Celular y Neurociencia "de Robertis" IBCN (UBA-CONICET), Buenos Aires, Argentina
| | - Nirit Kara
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Anna Ratsika
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Brunno Rocha Levone
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | | | | | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
11
|
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
|
12
|
Wedel S, Mathoor P, Rauh O, Heymann T, Ciotu CI, Fuhrmann DC, Fischer MJM, Weigert A, de Bruin N, Hausch F, Geisslinger G, Sisignano M. SAFit2 reduces neuroinflammation and ameliorates nerve injury-induced neuropathic pain. J Neuroinflammation 2022; 19:254. [PMID: 36217203 PMCID: PMC9552419 DOI: 10.1186/s12974-022-02615-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/04/2022] [Indexed: 01/17/2024] Open
Abstract
Background Neuropathic pain is experienced worldwide by patients suffering from nerve injuries, infectious or metabolic diseases or chemotherapy. However, the treatment options are still limited because of low efficacy and sometimes severe side effects. Recently, the deficiency of FKBP51 was shown to relieve chronic pain, revealing FKBP51 as a potential therapeutic target. However, a specific and potent FKBP51 inhibitor was not available until recently which hampered targeting of FKBP51. Methods In this study, we used the well-established and robust spared nerve injury model to analyze the effect of SAFit2 on nerve injury-induced neuropathic pain and to elucidate its pharmacodynamics profile. Therefore, the mice were treated with 10 mg/kg SAFit2 after surgery, the mice behavior was assessed over 21 days and biochemical analysis were performed after 14 and 21 days. Furthermore, the impact of SAFit2 on sensory neurons and macrophages was investigated in vitro. Results Here, we show that the FKBP51 inhibitor SAFit2 ameliorates nerve injury-induced neuropathic pain in vivo by reducing neuroinflammation. SAFit2 reduces the infiltration of immune cells into neuronal tissue and counteracts the increased NF-κB pathway activation which leads to reduced cytokine and chemokine levels in the DRGs and spinal cord. In addition, SAFit2 desensitizes the pain-relevant TRPV1 channel and subsequently reduces the release of pro-inflammatory neuropeptides from sensory neurons. Conclusions SAFit2 ameliorates neuroinflammation and counteracts enhanced neuronal activity after nerve injury leading to an amelioration of nerve injury-induced neuropathic pain. Based on these findings, SAFit2 constitutes as a novel and promising drug candidate for the treatment of nerve injury-induced neuropathic pain. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02615-7.
Collapse
Affiliation(s)
- Saskia Wedel
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
| | - Praveen Mathoor
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Oliver Rauh
- Membrane Biophysics, Department of Biology, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Tim Heymann
- Department of Chemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Cosmin I Ciotu
- Center of Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Michael J M Fischer
- Center of Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Natasja de Bruin
- Center of Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Felix Hausch
- Department of Chemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany. .,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany.
| |
Collapse
|
13
|
Häusl AS, Bajaj T, Brix LM, Pöhlmann ML, Hafner K, De Angelis M, Nagler J, Dethloff F, Balsevich G, Schramm KW, Giavalisco P, Chen A, Schmidt MV, Gassen NC. Mediobasal hypothalamic FKBP51 acts as a molecular switch linking autophagy to whole-body metabolism. SCIENCE ADVANCES 2022; 8:eabi4797. [PMID: 35263141 PMCID: PMC8906734 DOI: 10.1126/sciadv.abi4797] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The mediobasal hypothalamus (MBH) is the central region in the physiological response to metabolic stress. The FK506-binding protein 51 (FKBP51) is a major modulator of the stress response and has recently emerged as a scaffolder regulating metabolic and autophagy pathways. However, the detailed protein-protein interactions linking FKBP51 to autophagy upon metabolic challenges remain elusive. We performed mass spectrometry-based metabolomics of FKBP51 knockout (KO) cells revealing an increased amino acid and polyamine metabolism. We identified FKBP51 as a central nexus for the recruitment of the LKB1/AMPK complex to WIPI4 and TSC2 to WIPI3, thereby regulating the balance between autophagy and mTOR signaling in response to metabolic challenges. Furthermore, we demonstrated that MBH FKBP51 deletion strongly induces obesity, while its overexpression protects against high-fat diet (HFD)-induced obesity. Our study provides an important novel regulatory function of MBH FKBP51 within the stress-adapted autophagy response to metabolic challenges.
Collapse
Affiliation(s)
- Alexander S. Häusl
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Thomas Bajaj
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, 53127 Bonn, Germany
| | - Lea M. Brix
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Max L. Pöhlmann
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Meri De Angelis
- Helmholtz Center Munich Germany Research Center for Environmental Health, Molecular EXposomics, Neuherberg, Germany
| | - Joachim Nagler
- Helmholtz Center Munich Germany Research Center for Environmental Health, Molecular EXposomics, Neuherberg, Germany
| | | | - Georgia Balsevich
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Karl-Werner Schramm
- Helmholtz Center Munich Germany Research Center for Environmental Health, Molecular EXposomics, Neuherberg, Germany
| | | | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Mathias V. Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Corresponding author. (M.V.S.); (N.C.G.)
| | - Nils C. Gassen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, 53127 Bonn, Germany
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Corresponding author. (M.V.S.); (N.C.G.)
| |
Collapse
|
14
|
Qiu B, Zhong Z, Righter S, Xu Y, Wang J, Deng R, Wang C, Williams KE, Ma YY, Tsechpenakis G, Liang T, Yong W. FKBP51 modulates hippocampal size and function in post-translational regulation of Parkin. Cell Mol Life Sci 2022; 79:175. [PMID: 35244772 PMCID: PMC11072506 DOI: 10.1007/s00018-022-04167-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 11/29/2022]
Abstract
FK506-binding protein 51 (encoded by Fkpb51, also known as Fkbp5) has been associated with stress-related mental illness. To investigate its function, we studied the morphological consequences of Fkbp51 deletion. Artificial Intelligence-assisted morphological analysis revealed that male Fkbp51 knock-out (KO) mice possess more elongated dentate gyrus (DG) but shorter hippocampal height in coronal sections when compared to WT. Primary cultured Fkbp51 KO hippocampal neurons were shown to exhibit larger dendritic outgrowth than wild-type (WT) controls and pharmacological manipulation experiments suggest that this may occur through the regulation of microtubule-associated protein. Both in vitro primary culture and in vivo labeling support a role for FKBP51 in the regulation of microtubule-associated protein expression. Furthermore, Fkbp51 KO hippocampi exhibited decreases in βIII-tubulin, MAP2, and Tau protein levels, but a greater than 2.5-fold increase in Parkin protein. Overexpression and knock-down FKBP51 demonstrated that FKBP51 negatively regulates Parkin in a dose-dependent and ubiquitin-mediated manner. These results indicate a potential novel post-translational regulatory mechanism of Parkin by FKBP51 and the significance of their interaction on disease onset. KO has more flattened hippocampus using AI-assisted measurement Both pyramidal cell layer (PCL) of CA and granular cell layer (GCL) of DG distinguishable as two layers: deep cell layer and superficial layer. Distinct MAP2 expression between deep and superficial layer between KO and WT, Higher Parkin expression in KO brain Mechanism of FKBP51 inhibition resulting in Parkin, MAP2, Tau, and Tubulin expression differences between KO and WT mice, and resulting neurite outgrowth differences.
Collapse
Affiliation(s)
- Bin Qiu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Zhaohui Zhong
- Department of General Surgery, Peking University People's Hospital, Beijing, 100032, China
| | - Shawn Righter
- Department of Computer and Information Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Yuxue Xu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jun Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ran Deng
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chao Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kent E Williams
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yao-Ying Ma
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Gavriil Tsechpenakis
- Department of Computer and Information Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Tiebing Liang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Weidong Yong
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
15
|
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: 16] [Impact Index Per Article: 5.3] [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
|
16
|
Voll AM, Meyners C, Taubert MC, Bajaj T, Heymann T, Merz S, Charalampidou A, Kolos J, Purder PL, Geiger TM, Wessig P, Gassen NC, Bracher A, Hausch F. Makrozyklische FKBP51‐Liganden enthüllen einen transienten Bindungsmodus mit erhöhter Selektivität. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202017352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Andreas M. Voll
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Christian Meyners
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Martha C. Taubert
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Thomas Bajaj
- Research Group Neurohomeostasis Department of Psychiatry and Psychotherapy University of Bonn Venusberg Campus 1 53127 Bonn Deutschland
| | - Tim Heymann
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Stephanie Merz
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Anna Charalampidou
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Jürgen Kolos
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Patrick L. Purder
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Thomas M. Geiger
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| | - Pablo Wessig
- Universität Potsdam Institut für Chemie Karl-Liebknecht-Straße 24–25 14476 Potsdam Deutschland
| | - Nils C. Gassen
- Research Group Neurohomeostasis Department of Psychiatry and Psychotherapy University of Bonn Venusberg Campus 1 53127 Bonn Deutschland
| | - Andreas Bracher
- Max-Planck-Institute of Biochemistry Am Klopferspitz 18 82152 Martinsried Deutschland
| | - Felix Hausch
- Department Chemistry and Biochemistry Clemens-Schöpf-Institute Technical University Darmstadt Alarich-Weiss Straße 4 64287 Darmstadt Deutschland
| |
Collapse
|
17
|
Voll AM, Meyners C, Taubert MC, Bajaj T, Heymann T, Merz S, Charalampidou A, Kolos J, Purder PL, Geiger TM, Wessig P, Gassen NC, Bracher A, Hausch F. Macrocyclic FKBP51 Ligands Define a Transient Binding Mode with Enhanced Selectivity. Angew Chem Int Ed Engl 2021; 60:13257-13263. [PMID: 33843131 PMCID: PMC8252719 DOI: 10.1002/anie.202017352] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/14/2021] [Indexed: 12/28/2022]
Abstract
Subtype selectivity represents a challenge in many drug discovery campaigns. A typical example is the FK506 binding protein 51 (FKBP51), which has emerged as an attractive drug target. The most advanced FKBP51 ligands of the SAFit class are highly selective vs. FKBP52 but poorly discriminate against the homologs and off-targets FKBP12 and FKBP12.6. During a macrocyclization pilot study, we observed that many of these macrocyclic analogs have unanticipated and unprecedented preference for FKBP51 over FKBP12 and FKBP12.6. Structural studies revealed that these macrocycles bind with a new binding mode featuring a transient conformation, which is disfavored for the small FKBPs. Using a conformation-sensitive assay we show that this binding mode occurs in solution and is characteristic for this new class of compounds. The discovered macrocycles are non-immunosuppressive, engage FKBP51 in cells, and block the cellular effect of FKBP51 on IKKα. Our findings provide a new chemical scaffold for improved FKBP51 ligands and the structural basis for enhanced selectivity.
Collapse
Affiliation(s)
- Andreas M. Voll
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Christian Meyners
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Martha C. Taubert
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Thomas Bajaj
- Research Group NeurohomeostasisDepartment of Psychiatry and PsychotherapyUniversity of BonnVenusberg Campus 153127BonnGermany
| | - Tim Heymann
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Stephanie Merz
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Anna Charalampidou
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Jürgen Kolos
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Patrick L. Purder
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Thomas M. Geiger
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| | - Pablo Wessig
- Universität PotsdamInstitut für ChemieKarl-Liebknecht-Strasse 24–2514476PotsdamGermany
| | - Nils C. Gassen
- Research Group NeurohomeostasisDepartment of Psychiatry and PsychotherapyUniversity of BonnVenusberg Campus 153127BonnGermany
| | - Andreas Bracher
- Max-Planck-Institute of BiochemistryAm Klopferspitz 1882152MartinsriedGermany
| | - Felix Hausch
- Department Chemistry and BiochemistryClemens-Schöpf-InstituteTechnical University DarmstadtAlarich-Weiss Strasse 464287DarmstadtGermany
| |
Collapse
|
18
|
The Impact of the FKBP5 Gene Polymorphisms on the Relationship between Traumatic Life Events and Psychotic-Like Experiences in Non-Clinical Adults. Brain Sci 2021; 11:brainsci11050561. [PMID: 33925151 PMCID: PMC8144983 DOI: 10.3390/brainsci11050561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/24/2021] [Indexed: 01/30/2023] Open
Abstract
Common variations of the FKBP5 gene are implicated in psychotic disorders, by modulating the hypothalamic–pituitary–adrenal axis reactivity to stress. It has been demonstrated that some of them might moderate the effects of childhood trauma on psychosis proneness. However, these associations have not been investigated with respect to traumatic life events (TLEs). Therefore, we aimed to explore whether the FKBP5 polymorphisms moderate the effects of TLEs on the level of psychotic-like experiences (PLEs). A total of 535 non-clinical adults were approached for participation, and genotyping of six FKBP5 polymorphisms (rs3800373, rs9470080, rs4713902, rs737054, rs1360780 and rs9296158) was performed. The Prodromal Questionnaire-16 (PQ-16) and the Traumatic Events Checklist (TEC) were administered to assess PLEs and TLEs, respectively. Among the rs1360780 CC homozygotes, a history of physical abuse was associated with significantly higher PQ-16 scores. This difference was not significant in the rs1360780 T allele carriers. Similarly, a history of physical abuse was associated with significantly higher PQ-16 scores in the rs9296158 GG homozygotes but not in the rs9296158 A allele carriers. Finally, emotional neglect was related to significantly higher PQ-16 scores in the rs737054 T allele carriers but not in the rs737054 CC homozygotes. The present study indicates that variation in the FKBP5 gene might moderate the effects of lifetime traumatic events on psychosis proneness.
Collapse
|
19
|
Decidual cell FKBP51-progesterone receptor binding mediates maternal stress-induced preterm birth. Proc Natl Acad Sci U S A 2021; 118:2010282118. [PMID: 33836562 PMCID: PMC7980401 DOI: 10.1073/pnas.2010282118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Depression and posttraumatic stress disorder increase the risk of idiopathic preterm birth (iPTB); however, the exact molecular mechanism is unknown. Depression and stress-related disorders are linked to increased FK506-binding protein 51 (FKBP51) expression levels in the brain and/or FKBP5 gene polymorphisms. Fkbp5-deficient (Fkbp5 -/-) mice resist stress-induced depressive and anxiety-like behaviors. FKBP51 binding to progesterone (P4) receptors (PRs) inhibits PR function. Moreover, reduced PR activity and/or expression stimulates human labor. We report enhanced in situ FKBP51 expression and increased nuclear FKBP51-PR binding in decidual cells of women with iPTB versus gestational age-matched controls. In Fkbp5 +/+ mice, maternal restraint stress did not accelerate systemic P4 withdrawal but increased Fkbp5, decreased PR, and elevated AKR1C18 expression in uteri at E17.25 followed by reduced P4 levels and increased oxytocin receptor (Oxtr) expression at 18.25 in uteri resulting in PTB. These changes correlate with inhibition of uterine PR function by maternal stress-induced FKBP51. In contrast, Fkbp5 -/- mice exhibit prolonged gestation and are completely resistant to maternal stress-induced PTB and labor-inducing uterine changes detected in stressed Fkbp5 +/+ mice. Collectively, these results uncover a functional P4 withdrawal mechanism mediated by maternal stress-induced enhanced uterine FKBP51 expression and FKPB51-PR binding, resulting in iPTB.
Collapse
|
20
|
Bauder M, Meyners C, Purder PL, Merz S, Sugiarto WO, Voll AM, Heymann T, Hausch F. Structure-Based Design of High-Affinity Macrocyclic FKBP51 Inhibitors. J Med Chem 2021; 64:3320-3349. [PMID: 33666419 DOI: 10.1021/acs.jmedchem.0c02195] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The FK506-binding protein 51 (FKBP51) emerged as a key player in several diseases like stress-related disorders, chronic pain, and obesity. Linear analogues of FK506 called SAFit were shown to be highly selective for FKBP51 over its closest homologue FKBP52, allowing the proof-of-concept studies in animal models. Here, we designed and synthesized the first macrocyclic FKBP51-selective ligands to stabilize the active conformation. All macrocycles retained full FKBP51 affinity and selectivity over FKBP52 and the incorporation of polar functionalities further enhanced affinity. Six high-resolution crystal structures of macrocyclic inhibitors in complex with FKBP51 confirmed the desired selectivity-enabling binding mode. Our results show that macrocyclization is a viable strategy to target the shallow FKBP51 binding site selectively.
Collapse
Affiliation(s)
- Michael Bauder
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Christian Meyners
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Patrick L Purder
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Stephanie Merz
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Wisely Oki Sugiarto
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Andreas M Voll
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Tim Heymann
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Felix Hausch
- Department Chemistry and Biochemistry, Clemens-Schöpf-Institute, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| |
Collapse
|
21
|
Sannes AC, Christensen JO, Nielsen MB, Gjerstad J. The influence of age, gender and the FKBP5 genotype on subjective health complaints in the Norwegian working population. J Psychosom Res 2020; 139:110264. [PMID: 33038818 DOI: 10.1016/j.jpsychores.2020.110264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 01/30/2023]
Abstract
OBJECTIVE In the present study we examined the potential association between age, gender, the genetic variant FKBP5 rs9470080 and subjective health complaints. METHODS The data were collected through a three-wave nationally representative survey of 1060 Norwegian employees drawn from the Norwegian Central Employee Register by Statistics Norway. The follow-up period was six months. Subjective health complaints were scored by eight items reflecting; headache, neck pain, mid- and low back pain, stomach cramps or intestinal discomfort, problems with sleep onset or maintenance and early morning awakening. Genotyping with regard to FKBP5 rs9470080, previously linked to cortisol sensitivity, was carried out using Taqman assay. RESULTS The baseline data showed that in women, levels of subjective health complaints were highest among those above 50 years of age. Moreover, in women the rs9470080 CC variant at baseline was associated with higher levels of subjective health complaints. However, the effect size was not large, and no relationships were demonstrated between age or genotype and subjective health complaints in men. Also, the study variables were not related to any changes in the levels of subjective health complaints during the follow-up period. CONCLUSION We conclude that our biology such as age and sex, but also single genetic variants found in non-sex chromosomes, may be important in understanding the mechanisms underlying subjective health complaints in the general working population.
Collapse
|
22
|
Zhang J, Wu D, He Y, Li L, Liu S, Lu J, Gui H, Wang Y, Tao Y, Wang H, Kaushik D, Rodriguez R, Wang Z. Rapamycin inhibits AR signaling pathway in prostate cancer by interacting with the FK1 domain of FKBP51. Biochem Biophys Rep 2020; 23:100778. [PMID: 32695889 PMCID: PMC7365970 DOI: 10.1016/j.bbrep.2020.100778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/31/2020] [Accepted: 07/01/2020] [Indexed: 01/08/2023] Open
Abstract
Reactivation of the androgen receptor signaling pathway in the emasculated environment is the main reason for the occurrence of castration-resistant prostate cancer (CRPC). The immunophilin FKBP51, as a co-chaperone protein, together with Hsp90 help the correct folding of AR. Rapamycin is a known small-molecule inhibitor of FKBP51, but its effect on the FKBP51/AR signaling pathway is not clear. In this study, the interaction mechanism between FKBP51 and rapamycin was investigated using steady-state fluorescence quenching, X-ray crystallization, MTT assay, and qRT-PCR. Steady-state fluorescence quenching assay showed that rapamycin could interact with FKBP51. The crystal of the rapamycin-FKBP51 complex indicated that rapamycin occupies the hydrophobic binding pocket of FK1 domain which is vital for AR activity. The residues involving rapamycin binding are mainly hydrophobic and may overlap with the AR interaction site. Further assays showed that rapamycin could inhibit the androgen-dependent growth of human prostate cancer cells by down-regulating the expression levels of AR activated downstream genes. Taken together, our study demonstrates that rapamycin suppresses AR signaling pathway by interfering with the interaction between AR and FKBP51. The results of this study not only can provide useful information about the interaction mechanism between rapamycin and FKBP51, but also can provide new clues for the treatment of prostate cancer and castration-resistant prostate cancer. Rapamycin occupies the hydrophobic binding pocket of FK1 domain of FKBP51. Rapamycin suppresses the AR signaling pathway by interacting with FKBP51. Rapamycin inhibits the growth of prostate cancer cells via the AR signaling pathway.
Collapse
Affiliation(s)
- Jing Zhang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| | - Dan Wu
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China.,School of Life Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Yongxing He
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Lanlan Li
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| | - Shanhui Liu
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| | - Jianzhong Lu
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| | - Huiming Gui
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| | - Yuhan Wang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| | - Yan Tao
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| | - Hanzhang Wang
- Department of Urology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, USA
| | - Dharam Kaushik
- Department of Urology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, USA
| | - Ronald Rodriguez
- Department of Urology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, USA
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Lanzhou, Gansu, 730000, PR China
| |
Collapse
|
23
|
Jung JA, Yoon YJ. Development of Non-Immunosuppressive FK506 Derivatives as Antifungal and Neurotrophic Agents. J Microbiol Biotechnol 2020; 30:1-10. [PMID: 31752059 PMCID: PMC9728173 DOI: 10.4014/jmb.1911.11008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
FK506, also known as tacrolimus, is a clinically important immunosuppressant drug and has promising therapeutic potentials owing to its antifungal, neuroprotective, and neuroregenerative activities. To generate various FK506 derivatives, the structure of FK506 has been modified by chemical methods or biosynthetic pathway engineering. Herein, we describe the mode of the antifungal action of FK506 and the structure-activity relationship of FK506 derivatives in the context of immunosuppressive and antifungal activities. In addition, we discuss the neurotrophic mechanism of FK506 known to date, along with the neurotrophic FK506 derivatives with significantly reduced immunosuppressive activity. This review suggests the possibility to generate novel FK506 derivatives as antifungal as well as neuroregenerative/neuroprotective agents.
Collapse
Affiliation(s)
- Jin A Jung
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yeo Joon Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea,Corresponding author Phone: +82-2-3277-4082 Fax: +82-2-3277-3419 E-mail:
| |
Collapse
|
24
|
Feng X, Sippel C, Knaup FH, Bracher A, Staibano S, Romano MF, Hausch F. A Novel Decalin-Based Bicyclic Scaffold for FKBP51-Selective Ligands. J Med Chem 2019; 63:231-240. [PMID: 31800244 DOI: 10.1021/acs.jmedchem.9b01157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Selective inhibition of FKBP51 has emerged as possible novel treatment for diseases like major depressive disorder, obesity, chronic pain, and certain cancers. The current FKBP51 inhibitors are rather large, flexible, and have to be further optimized. By using a structure-based rigidification strategy, we hereby report the design and synthesis of a novel promising bicyclic scaffold for FKBP51 ligands. The structure-activity analysis revealed the decalin scaffold as the best moiety for the selectivity-enabling subpocket of FBKP51. The resulting compounds retain high potency for FKBP51 and excellent selectivity over the close homologue FKBP52. With the cocrystal structure of an advanced ligand in this novel series, we show how the decalin locks the key selectivity-inducing cyclohexyl moiety of the ligand in a conformation typical for FKBP51-selective binding. The best compound 29 produces cell death in a HeLa-derived KB cell line, a cellular model of cervical adenocarcinoma, where FKBP51 is highly overexpressed. Our results show how FKBP51 inhibitors can be rigidified and extended while preserving FKBP51 selectivity. Such inhibitors might be novel tools in the treatment of human cancers with deregulated FKBP51.
Collapse
Affiliation(s)
- Xixi Feng
- Department of Translational Research in Psychiatry , Max Planck Institute of Psychiatry , Kraepelinstrasse 2 , 80804 Munich , Germany
| | - Claudia Sippel
- Department of Translational Research in Psychiatry , Max Planck Institute of Psychiatry , Kraepelinstrasse 2 , 80804 Munich , Germany
| | - Fabian H Knaup
- Institute for Organic Chemistry and Biochemistry , Technische Universität Darmstadt , Alarich-Weiss-Strasse 4 , D-64287 Darmstadt , Germany
| | - Andreas Bracher
- Max Planck Institute of Biochemistry , Am Klopferspitz 18 , 82152 Martinsried , Germany
| | - Stefania Staibano
- Department of Advanced Biomedical Sciences , Federico II University of Naples , 80131 Naples , Italy
| | - Maria F Romano
- Department of Molecular Medicine and Medical Biotechnologies , Federico II University , 80131 Naples , Italy
| | - Felix Hausch
- Institute for Organic Chemistry and Biochemistry , Technische Universität Darmstadt , Alarich-Weiss-Strasse 4 , D-64287 Darmstadt , Germany
| |
Collapse
|
25
|
Häusl AS, Balsevich G, Gassen NC, Schmidt MV. Focus on FKBP51: A molecular link between stress and metabolic disorders. Mol Metab 2019; 29:170-181. [PMID: 31668388 PMCID: PMC6812026 DOI: 10.1016/j.molmet.2019.09.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Obesity, Type 2 diabetes (T2D) as well as stress-related disorders are rising public health threats and major burdens for modern society. Chronic stress and depression are highly associated with symptoms of the metabolic syndrome, but the molecular link is still not fully understood. Furthermore, therapies tackling these biological disorders are still lacking. The identification of shared molecular targets underlying both pathophysiologies may lead to the development of new treatments. The FK506 binding protein 51 (FKBP51) has recently been identified as a promising therapeutic target for stress-related psychiatric disorders and obesity-related metabolic outcomes. SCOPE OF THE REVIEW The aim of this review is to summarize current evidence of in vitro, preclinical, and human studies on the stress responsive protein FKBP51, focusing on its newly discovered role in metabolism. Also, we highlight the therapeutic potential of FKBP51 as a new treatment target for symptoms of the metabolic syndrome. MAJOR CONCLUSIONS We conclude the review by emphasizing missing knowledge gaps that remain and future research opportunities needed to implement FKBP51 as a drug target for stress-related obesity or T2D.
Collapse
Affiliation(s)
- Alexander S Häusl
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| | - Georgia Balsevich
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Ab T2N 4N1, Canada
| | - Nils C Gassen
- Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, 53127, Bonn, Germany; Department of Translational Research in Psychiatry, 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.
| |
Collapse
|
26
|
Jagtap PKA, Asami S, Sippel C, Kaila VRI, Hausch F, Sattler M. Selective Inhibitors of FKBP51 Employ Conformational Selection of Dynamic Invisible States. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201902994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Pravin Kumar Ankush Jagtap
- Lehrstuhl für Biomolekulare NMR-SpektroskopieTechnische Universität München Lichtenbergstr. 4 85747 Garching Germany
| | - Sam Asami
- Lehrstuhl für Biomolekulare NMR-SpektroskopieTechnische Universität München Lichtenbergstr. 4 85747 Garching Germany
| | - Claudia Sippel
- Max Planck Institute of Psychiatry Kraepelinstr. 2–10 80804 Munich Germany
| | - Ville R. I. Kaila
- Department ChemieTechnische Universität München Lichtenbergstr. 4 85747 Garching Germany
| | - Felix Hausch
- Max Planck Institute of Psychiatry Kraepelinstr. 2–10 80804 Munich Germany
- Present address: Structure-Based Drug ResearchTechnische Universität Darmstadt Alarich-Weiss-Str. 4 64287 Darmstadt Germany
| | - Michael Sattler
- Lehrstuhl für Biomolekulare NMR-SpektroskopieTechnische Universität München Lichtenbergstr. 4 85747 Garching Germany
| |
Collapse
|
27
|
chen J, Yin B, Pang L, Wang W, Zhang JZH, Zhu T. Binding modes and conformational changes of FK506-binding protein 51 induced by inhibitor bindings: insight into molecular mechanisms based on multiple simulation technologies. J Biomol Struct Dyn 2019; 38:2141-2155. [DOI: 10.1080/07391102.2019.1624616] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jianzhong chen
- School of Science, Shandong Jiaotong University, Jinan, China
| | - Baohua Yin
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Laixue Pang
- School of Science, Shandong Jiaotong University, Jinan, China
| | - Wei Wang
- School of Science, Shandong Jiaotong University, Jinan, China
| | - John Z. H. Zhang
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, China
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Tong Zhu
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, China
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| |
Collapse
|
28
|
Jagtap PKA, Asami S, Sippel C, Kaila VRI, Hausch F, Sattler M. Selective Inhibitors of FKBP51 Employ Conformational Selection of Dynamic Invisible States. Angew Chem Int Ed Engl 2019; 58:9429-9433. [PMID: 31100184 DOI: 10.1002/anie.201902994] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/24/2019] [Indexed: 12/26/2022]
Abstract
The recently discovered SAFit class of inhibitors against the Hsp90 co-chaperone FKBP51 show greater than 10 000-fold selectivity over its closely related paralogue FKBP52. However, the mechanism underlying this selectivity remained unknown. By combining NMR spectroscopy, biophysical and computational methods with mutational analysis, we show that the SAFit molecules bind to a transient pocket in FKBP51. This represents a weakly populated conformation resembling the inhibitor-bound state of FKBP51, suggesting conformational selection rather than induced fit as the major binding mechanism. The inhibitor-bound conformation of FKBP51 is stabilized by an allosteric network of residues located away from the inhibitor-binding site. These residues stabilize the Phe67 side chain in a dynamic outward conformation and are distinct in FKBP52, thus rationalizing the basis for the selectivity of SAFit inhibitors. Our results represent a paradigm for the selective inhibition of transient binding pockets.
Collapse
Affiliation(s)
- Pravin Kumar Ankush Jagtap
- Lehrstuhl für Biomolekulare NMR-Spektroskopie, Technische Universität München, Lichtenbergstr. 4, 85747, Garching, Germany
| | - Sam Asami
- Lehrstuhl für Biomolekulare NMR-Spektroskopie, Technische Universität München, Lichtenbergstr. 4, 85747, Garching, Germany
| | - Claudia Sippel
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Ville R I Kaila
- Department Chemie, Technische Universität München, Lichtenbergstr. 4, 85747, Garching, Germany
| | - Felix Hausch
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany.,Present address: Structure-Based Drug Research, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, 64287, Darmstadt, Germany
| | - Michael Sattler
- Lehrstuhl für Biomolekulare NMR-Spektroskopie, Technische Universität München, Lichtenbergstr. 4, 85747, Garching, Germany
| |
Collapse
|
29
|
Balsevich G, Abizaid A, Chen A, Karatsoreos IN, Schmidt MV. Stress and glucocorticoid modulation of feeding and metabolism. Neurobiol Stress 2019; 11:100171. [PMID: 31193462 PMCID: PMC6529856 DOI: 10.1016/j.ynstr.2019.100171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 12/04/2022] Open
Abstract
This perspective highlights research presented as part of the symposium entitled, “Stress and Glucocorticoid Modulation of Feeding and Metabolism” at the 2018 Neurobiology of Stress Workshop held in Banff, AB, Canada. The symposium comprised five researchers at different career stages who each study different aspects of the interaction between the stress response and metabolic control. Their collective results reveal the complexity of this relationship in terms of behavioural and physiological outcomes. Their work emphasizes the need to consider the level of interaction (cellular, tissue, systems) as well as the timing and context in which the interaction is studied. Rather than a comprehensive review on the work presented at the Symposium, here we discuss recurring themes that emerged at the biennial workshop, which address new avenues of research that will drive the field forward.
Collapse
Affiliation(s)
- G Balsevich
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - A Abizaid
- Institute of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - A Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstrasse 2 - 10, Munich, 80804, Germany
| | - I N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, 1815 Ferdinand's Lane, Pullman, WA, 99164, United States
| | - M V Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstrasse 2 - 10, Munich, 80804, Germany
| |
Collapse
|
30
|
Snijders C, de Nijs L, Baker DG, Hauger RL, van den Hove D, Kenis G, Nievergelt CM, Boks MP, Vermetten E, Gage FH, Rutten BPF. MicroRNAs in Post-traumatic Stress Disorder. Curr Top Behav Neurosci 2019; 38:23-46. [PMID: 29063484 DOI: 10.1007/7854_2017_32] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disorder that can develop following exposure to or witnessing of a (potentially) threatening event. A critical issue is to pinpoint the (neuro)biological mechanisms underlying the susceptibility to stress-related disorder such as PTSD, which develops in the minority of ~15% of individuals exposed to trauma. Over the last few years, a first wave of epigenetic studies has been performed in an attempt to identify the molecular underpinnings of the long-lasting behavioral and mental effects of trauma exposure. The potential roles of non-coding RNAs (ncRNAs) such as microRNAs (miRNAs) in moderating or mediating the impact of severe stress and trauma are increasingly gaining attention. To date, most studies focusing on the roles of miRNAs in PTSD have, however, been completed in animals, using cross-sectional study designs and focusing almost exclusively on subjects with susceptible phenotypes. Therefore, there is a strong need for new research comprising translational and cross-species approaches that use longitudinal designs for studying trajectories of change contrasting susceptible and resilient subjects. The present review offers a comprehensive overview of available studies of miRNAs in PTSD and discusses the current challenges, pitfalls, and future perspectives of this field.
Collapse
Affiliation(s)
- Clara Snijders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, European Graduate School of Neuroscience, (EURON), Maastricht, 6200 MD, The Netherlands
| | - Laurence de Nijs
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, European Graduate School of Neuroscience, (EURON), Maastricht, 6200 MD, The Netherlands
| | - Dewleen G Baker
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, 92037, USA
- VA Center of Excellence for Stress and Mental Health, San Diego, La Jolla, CA, 92037, USA
- VA San Diego Healthcare System, San Diego, La Jolla, CA, 92037, USA
| | - Richard L Hauger
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, 92037, USA
- VA Center of Excellence for Stress and Mental Health, San Diego, La Jolla, CA, 92037, USA
- VA San Diego Healthcare System, San Diego, La Jolla, CA, 92037, USA
| | - Daniel van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, European Graduate School of Neuroscience, (EURON), Maastricht, 6200 MD, The Netherlands
- Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, 97080, Germany
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, European Graduate School of Neuroscience, (EURON), Maastricht, 6200 MD, The Netherlands
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, 92037, USA
- VA Center of Excellence for Stress and Mental Health, San Diego, La Jolla, CA, 92037, USA
| | - Marco P Boks
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, 3584 CG, The Netherlands
| | - Eric Vermetten
- Military Mental Health Research Center, Ministry of Defense, P.O. Box 90000, Utrecht, 3509 AA, The Netherlands
- Department of Psychiatry, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- Arq Psychotrauma Research Group, Diemen, 1112 XE, The Netherlands
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, European Graduate School of Neuroscience, (EURON), Maastricht, 6200 MD, The Netherlands.
| |
Collapse
|
31
|
Boonying W, Joselin A, Huang E, Qu D, Safarpour F, Iyirhiaro GO, Gonzalez YR, Callaghan SM, Slack RS, Figeys D, Chung YH, Park DS. Pink1 regulates FKBP5 interaction with AKT/PHLPP and protects neurons from neurotoxin stress induced by MPP .. J Neurochem 2019; 150:312-329. [PMID: 30734931 DOI: 10.1111/jnc.14683] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 12/14/2022]
Abstract
Loss of function mutations in the PTEN-induced putative kinase 1 (Pink1) gene have been linked with an autosomal recessive familial form of early onset Parkinson's disease (PD). However, the underlying mechanism(s) responsible for degeneration remains elusive. Presently, using co-immunoprecipitation in HEK (Human embryonic kidney) 293 cells, we show that Pink1 endogenously interacts with FK506-binding protein 51 (FKBP51 or FKBP5), FKBP5 and directly phosphorylates FKBP5 at Serine in an in vitro kinase assay. Both FKBP5 and Pink1 have been previously associated with protein kinase B (AKT) regulation. We provide evidence using primary cortical cultured neurons from Pink1-deficient mice that Pink1 increases AKT phosphorylation at Serine 473 (Ser473) challenged by 1-methyl-4-phenylpyridinium (MPP+ ) and that over-expression of FKBP5 using an adeno-associated virus delivery system negatively regulates AKT phosphorylation at Ser473 in murine-cultured cortical neurons. Interestingly, FKBP5 over-expression promotes death in response to MPP+ in the absence of Pink1. Conversely, shRNA-mediated knockdown of FKBP5 in cultured cortical neurons is protective and this effect is reversed with inhibition of AKT signaling. In addition, shRNA down-regulation of PH domain leucine-rich repeat protein phosphatase (PHLPP) in Pink1 WT neurons increases neuronal survival, while down-regulation of PHLPP in Pink1 KO rescues neuronal death in response to MPP+ . Finally, using co-immunoprecipitation, we show that FKBP5 interacts with the kinase AKT and phosphatase PHLPP. This interaction is increased in the absence of Pink1, both in Mouse Embryonic Fibroblasts (MEF) and in mouse brain tissue. Expression of kinase dead Pink1 (K219M) enhances FKBP5 interaction with both AKT and PHLPP. Overall, our results suggest a testable model by which Pink1 could regulate AKT through phosphorylation of FKBP5 and interaction of AKT with PHLPP. Our results suggest a potential mechanism by which PINK1-FKBP5 pathway contributes to neuronal death in PD. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Wassamon Boonying
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, South Korea.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Alvin Joselin
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - En Huang
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Dianbo Qu
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Farzaneh Safarpour
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Grace O Iyirhiaro
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Yasmilde Rodriguez Gonzalez
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Steve M Callaghan
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Daniel Figeys
- Department of Biochemistry, Microbiology and Immunology, Department of Chemistry and Biomolecular Sciences, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Young-Hwa Chung
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, South Korea
| | - David S Park
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, South Korea.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
32
|
Richter-Levin G, Stork O, Schmidt MV. Animal models of PTSD: a challenge to be met. Mol Psychiatry 2019; 24:1135-1156. [PMID: 30816289 PMCID: PMC6756084 DOI: 10.1038/s41380-018-0272-5] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 08/13/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
Abstract
Recent years have seen increased interest in psychopathologies related to trauma exposure. Specifically, there has been a growing awareness to posttraumatic stress disorder (PTSD) in part due to terrorism, climate change-associated natural disasters, the global refugee crisis, and increased violence in overpopulated urban areas. However, notwithstanding the increased awareness to the disorder, the increasing number of patients, and the devastating impact on the lives of patients and their families, the efficacy of available treatments remains limited and highly unsatisfactory. A major scientific effort is therefore devoted to unravel the neural mechanisms underlying PTSD with the aim of paving the way to developing novel or improved treatment approaches and drugs to treat PTSD. One of the major scientific tools used to gain insight into understanding physiological and neuronal mechanisms underlying diseases and for treatment development is the use of animal models of human diseases. While much progress has been made using these models in understanding mechanisms of conditioned fear and fear memory, the gained knowledge has not yet led to better treatment options for PTSD patients. This poor translational outcome has already led some scientists and pharmaceutical companies, who do not in general hold opinions against animal models, to propose that those models should be abandoned. Here, we critically examine aspects of animal models of PTSD that may have contributed to the relative lack of translatability, including the focus on the exposure to trauma, overlooking individual and sex differences, and the contribution of risk factors. Based on findings from recent years, we propose research-based modifications that we believe are required in order to overcome some of the shortcomings of previous practice. These modifications include the usage of animal models of PTSD which incorporate risk factors and of the behavioral profiling analysis of individuals in a sample. These modifications are aimed to address factors such as individual predisposition and resilience, thus taking into consideration the fact that only a fraction of individuals exposed to trauma develop PTSD. We suggest that with an appropriate shift of practice, animal models are not only a valuable tool to enhance our understanding of fear and memory processes, but could serve as effective platforms for understanding PTSD, for PTSD drug development and drug testing.
Collapse
Affiliation(s)
- Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel. .,The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa, Israel. .,Psychology Department, University of Haifa, Haifa, Israel.
| | - Oliver Stork
- 0000 0001 1018 4307grid.5807.aDepartment of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany ,grid.452320.2Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Mathias V. Schmidt
- 0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
33
|
Menke A. Is the HPA Axis as Target for Depression Outdated, or Is There a New Hope? Front Psychiatry 2019; 10:101. [PMID: 30890970 PMCID: PMC6413696 DOI: 10.3389/fpsyt.2019.00101] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/11/2019] [Indexed: 12/14/2022] Open
Abstract
Major depressive disorder (MDD) is a very common stress-related mental disorder that carries a huge burden for affected patients and the society. It is associated with a high mortality that derives from suicidality and the development of serious medical conditions such as heart diseases, diabetes, and stroke. Although a range of effective antidepressants are available, more than 50% of the patients do not respond to the first treatment they are prescribed and around 30% fail to respond even after several treatment attempts. The heterogeneous condition of MDD, the lack of biomarkers matching patients with the right treatments and the situation that almost all available drugs are only targeting the serotonin, norepinephrine, or dopamine signaling, without regulating other potentially dysregulated systems may explain the insufficient treatment status. The hypothalamic-pituitary-adrenal (HPA) axis is one of these other systems, there is numerous and robust evidence that it is implicated in MDD and other stress-related conditions, but up to date there is no specific drug targeting HPA axis components that is approved and no test that is routinely used in the clinical setting identifying patients for such a specific treatment. Is there still hope after these many years for a breakthrough of agents targeting the HPA axis? This review will cover tests detecting altered HPA axis function and the specific treatment options such as glucocorticoid receptor (GR) antagonists, corticotropin-releasing hormone 1 (CRH1) receptor antagonists, tryptophan 2,3-dioxygenase (TDO) inhibitors and FK506 binding protein 5 (FKBP5) receptor antagonists.
Collapse
Affiliation(s)
- Andreas Menke
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
34
|
Kolos JM, Voll AM, Bauder M, Hausch F. FKBP Ligands-Where We Are and Where to Go? Front Pharmacol 2018; 9:1425. [PMID: 30568592 PMCID: PMC6290070 DOI: 10.3389/fphar.2018.01425] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
In recent years, many members of the FK506-binding protein (FKBP) family were increasingly linked to various diseases. The binding domain of FKBPs differs only in a few amino acid residues, but their biological roles are versatile. High-affinity ligands with selectivity between close homologs are scarce. This review will give an overview of the most prominent ligands developed for FKBPs and highlight a perspective for future developments. More precisely, human FKBPs and correlated diseases will be discussed as well as microbial FKBPs in the context of anti-bacterial and anti-fungal therapeutics. The last section gives insights into high-affinity ligands as chemical tools and dimerizers.
Collapse
Affiliation(s)
| | | | | | - Felix Hausch
- Department of Chemistry, Institute of Chemistry and Biochemistry, Darmstadt University of Technology, Darmstadt, Germany
| |
Collapse
|
35
|
Alterations in Systemic and Cognitive Glucocorticoid Sensitivity in Depression. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 4:310-320. [PMID: 30686583 DOI: 10.1016/j.bpsc.2018.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/21/2018] [Accepted: 11/27/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Decades of research point to cortisol insensitivity as a biomarker of depression. Despite a vast literature on cortisol's effects on memory, the role of cortisol insensitivity in core psychological features of depression, such as emotional memory biases, is unknown. METHODS Sixty-five premenopausal women with varying levels of depression completed this study involving an at-home low-dose dexamethasone suppression test and four experimental sessions (i.e., two visits for memory encoding of emotionally arousing pictures, each of which was followed 48 hours later by a recall test). Participants received 20 mg of oral cortisol (CORT) or placebo prior to encoding. We tested whether systemic cortisol insensitivity measured with the dexamethasone suppression test predicted cognitive sensitivity to CORT, which was operationalized as the change in negatively biased memory formation for pictures encoded following CORT versus placebo administration. RESULTS Cortisol insensitivity was associated with more severe depression and flatter diurnal cortisol levels. Cortisol insensitivity predicted negative memory bias for pictures encoded during the placebo session and reduction in negative memory bias for pictures encoded during the CORT (compared with placebo) session, even after accounting for psychiatric symptomatology. CONCLUSIONS Our findings replicate research showing that cortisol insensitivity predicts depression severity and flatter diurnal cortisol levels. The results further suggest that systemic cortisol insensitivity is related to negative memory bias and its alleviation by cortisol administration. These novel cognitive findings tie together knowledge regarding endocrine and psychological dysfunction in depression and suggest that boosting cortisol signal may cognitively benefit individuals with cortisol insensitivity.
Collapse
|
36
|
Pomplun S, Sippel C, Hähle A, Tay D, Shima K, Klages A, Ünal CM, Rieß B, Toh HT, Hansen G, Yoon HS, Bracher A, Preiser P, Rupp J, Steinert M, Hausch F. Chemogenomic Profiling of Human and Microbial FK506-Binding Proteins. J Med Chem 2018; 61:3660-3673. [PMID: 29578710 DOI: 10.1021/acs.jmedchem.8b00137] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
FK506-binding proteins (FKBPs) are evolutionarily conserved proteins that display peptidyl-prolyl isomerase activities and act as coreceptors for immunosuppressants. Microbial macrophage-infectivity-potentiator (Mip)-type FKBPs can enhance infectivity. However, developing druglike ligands for FKBPs or Mips has proven difficult, and many FKBPs and Mips still lack biologically useful ligands. To explore the scope and potential of C5-substituted [4.3.1]-aza-bicyclic sulfonamides as a broadly applicable class of FKBP inhibitors, we developed a new synthesis method for the bicyclic core scaffold and used it to prepare an FKBP- and Mip-focused library. This allowed us to perform a systematic structure-activity-relationship analysis across key human FKBPs and microbial Mips, yielding highly improved inhibitors for all the FKBPs studied. A cocrystal structure confirmed the molecular-binding mode of the core structure and explained the affinity gained as a result of the preferred substituents. The best FKBP and Mip ligands showed promising antimalarial, antileginonellal, and antichlamydial properties in cellular models of infectivity, suggesting that substituted [4.3.1]-aza-bicyclic sulfonamides could be a novel class of anti-infectives.
Collapse
Affiliation(s)
- Sebastian Pomplun
- Department of Translational Research in Psychiatry , Max Planck Institute of Psychiatry , 80804 Munich , Germany
| | - Claudia Sippel
- Department of Translational Research in Psychiatry , Max Planck Institute of Psychiatry , 80804 Munich , Germany
| | - Andreas Hähle
- Department of Translational Research in Psychiatry , Max Planck Institute of Psychiatry , 80804 Munich , Germany.,Technical University Darmstadt , Alarich-Weiss-Straße 4 , 64287 Darmstadt , Germany
| | - Donald Tay
- School of Biological Sciences , Nanyang Technological University , 639798 Singapore
| | - Kensuke Shima
- Department of Infectious Diseases and Microbiology , University of Lübeck , 23562 Lübeck , Germany
| | - Alina Klages
- Technische Universität Braunschweig , 38106 Braunschweig , Germany
| | - Can Murat Ünal
- Technische Universität Braunschweig , 38106 Braunschweig , Germany
| | - Benedikt Rieß
- Department of Translational Research in Psychiatry , Max Planck Institute of Psychiatry , 80804 Munich , Germany
| | - Hui Ting Toh
- School of Biological Sciences , Nanyang Technological University , 639798 Singapore
| | | | - Ho Sup Yoon
- School of Biological Sciences , Nanyang Technological University , 639798 Singapore
| | - Andreas Bracher
- Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
| | - Peter Preiser
- School of Biological Sciences , Nanyang Technological University , 639798 Singapore
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology , University of Lübeck , 23562 Lübeck , Germany
| | - Michael Steinert
- Department of Infectious Diseases and Microbiology , University of Lübeck , 23562 Lübeck , Germany.,Helmholtz Centre for Infection Research , 38124 Braunschweig , Germany
| | - Felix Hausch
- Department of Translational Research in Psychiatry , Max Planck Institute of Psychiatry , 80804 Munich , Germany.,Technical University Darmstadt , Alarich-Weiss-Straße 4 , 64287 Darmstadt , Germany
| |
Collapse
|
37
|
Shi D, Bai Q, Zhou S, Liu X, Liu H, Yao X. Molecular dynamics simulation, binding free energy calculation and unbinding pathway analysis on selectivity difference between FKBP51 and FKBP52: Insight into the molecular mechanism of isoform selectivity. Proteins 2017; 86:43-56. [PMID: 29023988 DOI: 10.1002/prot.25401] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 09/13/2017] [Accepted: 10/11/2017] [Indexed: 12/12/2022]
Abstract
As co-chaperones of the 90-kDa heat shock protein(HSP90), FK506 binding protein 51 (FKBP51) and FK506 binding protein 52 (FKBP52) modulate the maturation of steroid hormone receptor through their specific FK1 domains (FKBP12-like domain 1). The inhibitors targeting FK1 domains are potential therapies for endocrine-related physiological disorders. However, the structural conservation of the FK1 domains between FKBP51 and FKBP52 make it difficult to obtain satisfactory selectivity in FK506-based drug design. Fortunately, a series of iFit ligands synthesized by Hausch et al exhibited excellent selectivity for FKBP51, providing new opportunity for design selective inhibitors. We performed molecular dynamics simulation, binding free energy calculation and unbinding pathway analysis to reveal selective mechanism for the inhibitor iFit4 binding with FKBP51 and FKBP52. The conformational stability evaluation of the "Phe67-in" and "Phe67-out" states implies that FKBP51 and FKBP52 have different preferences for "Phe67-in" and "Phe67-out" states, which we suggest as the determinant factor for the selectivity for FKBP51. The binding free energy calculations demonstrate that nonpolar interaction is favorable for the inhibitors binding, while the polar interaction and entropy contribution are adverse for the inhibitors binding. According to the results from binding free energy decomposition, the electrostatic difference of residue 85 causes the most significant thermodynamics effects on the binding of iFit4 to FKBP51 and FKBP52. Furthermore, the importance of substructure units on iFit4 were further evaluated by unbinding pathway analysis and residue-residue contact analysis between iFit4 and the proteins. The results will provide new clues for the design of selective inhibitors for FKBP51.
Collapse
Affiliation(s)
- Danfeng Shi
- Department of Chemistry, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, China
| | - Qifeng Bai
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Shuangyan Zhou
- Department of Chemistry, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, China.,School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xuewei Liu
- Department of Chemistry, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, China
| | - Huanxiang Liu
- Department of Chemistry, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, China.,School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xiaojun Yao
- Department of Chemistry, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| |
Collapse
|
38
|
Maiarù M, Tochiki KK, Cox MB, Annan LV, Bell CG, Feng X, Hausch F, Géranton SM. The stress regulator FKBP51 drives chronic pain by modulating spinal glucocorticoid signaling. Sci Transl Med 2016; 8:325ra19. [PMID: 26865567 DOI: 10.1126/scitranslmed.aab3376] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Polymorphisms in FKBP51 are associated with stress-related psychiatric disorders and influence the severity of pain symptoms experienced after trauma. We report that FKBP51 (FK506 binding protein 51) is crucial for the full development and maintenance of long-term pain states. Indeed, FKBP51 knockout mice, as well as mice in which silencing of FKBP51 is restricted to the spinal cord, showed reduced hypersensitivity in several persistent pain models in rodents. FKBP51 deletion did not compromise the detection of acute painful stimuli, a critical protective mechanism. Moreover, the intrathecal administration of the specific FKBP51 inhibitor SAFit2 reduced the severity of an established pain state, confirming the crucial role of spinal FKBP51 in nociceptive processing. Finally, glucocorticoid signaling, which is known to modulate persistent pain states in rodents, was impaired in FKBP51 knockout mice. This finding suggested that FKBP51 regulates chronic pain by modulation of glucocorticoid signaling. Thus, FKBP51 is a central mediator of chronic pain, likely in humans as well as rodents, and is a new pharmacologically tractable target for the treatment of long-term pain states.
Collapse
Affiliation(s)
- Maria Maiarù
- Cell and Developmental Biology, University College London (UCL), London WC1E 6BT, UK
| | - Keri K Tochiki
- Cell and Developmental Biology, University College London (UCL), London WC1E 6BT, UK
| | - Marc B Cox
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Leonette V Annan
- Cell and Developmental Biology, University College London (UCL), London WC1E 6BT, UK
| | - Christopher G Bell
- Epigenomic Medicine, Centre for Biological Sciences, Institute of Developmental Sciences, S017 1BJ, and MRC (Medical Research Council) Lifecourse Epidemiology Unit, S016 6YD, University of Southampton, Southampton, UK
| | - Xixi Feng
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Felix Hausch
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Sandrine M Géranton
- Cell and Developmental Biology, University College London (UCL), London WC1E 6BT, UK.
| |
Collapse
|
39
|
LeMaster DM, Hernandez G. Conformational Dynamics in FKBP Domains: Relevance to Molecular Signaling and Drug Design. Curr Mol Pharmacol 2016; 9:5-26. [PMID: 25986571 DOI: 10.2174/1874467208666150519113146] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 02/25/2015] [Accepted: 05/17/2015] [Indexed: 01/05/2023]
Abstract
Among the 22 FKBP domains in the human genome, FKBP12.6 and the first FKBP domains (FK1) of FKBP51 and FKBP52 are evolutionarily and structurally most similar to the archetypical FKBP12. As such, the development of inhibitors with selectivity among these four FKBP domains poses a significant challenge for structure-based design. The pleiotropic effects of these FKBP domains in a range of signaling processes such as the regulation of ryanodine receptor calcium channels by FKBP12 and FKBP12.6 and steroid receptor regulation by the FK1 domains of FKBP51 and FKBP52 amply justify the efforts to develop selective therapies. In contrast to their close structural similarities, these four FKBP domains exhibit a substantial diversity in their conformational flexibility. A number of distinct conformational transitions have been characterized for FKBP12 spanning timeframes from 20 s to 10 ns and in each case these dynamics have been shown to markedly differ from the conformational behavior for one or more of the other three FKBP domains. Protein flexibilitybased inhibitor design could draw upon the transitions that are significantly populated in only one of the targeted proteins. Both the similarities and differences among these four proteins valuably inform the understanding of how dynamical effects propagate across the FKBP domains as well as potentially how such intramolecular transitions might couple to the larger scale transitions that are central to the signaling complexes in which these FKBP domains function.
Collapse
Affiliation(s)
| | - Griselda Hernandez
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, New York, 12201, USA; Department of Biomedical Sciences, School of Public Health, University at Albany - SUNY, Empire State Plaza, Albany, New York, 12201, USA.
| |
Collapse
|
40
|
Nylander I, Todkar A, Granholm L, Vrettou M, Bendre M, Boon W, Andershed H, Tuvblad C, Nilsson KW, Comasco E. Evidence for a Link Between Fkbp5/FKBP5, Early Life Social Relations and Alcohol Drinking in Young Adult Rats and Humans. Mol Neurobiol 2016; 54:6225-6234. [PMID: 27709495 PMCID: PMC5583263 DOI: 10.1007/s12035-016-0157-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/22/2016] [Indexed: 01/10/2023]
Abstract
Alcohol misuse has been linked to dysregulation of stress, emotion, and reward brain circuitries. A candidate key mediator of this association is the FK506-binding protein (FKBP5), a negative regulator of the glucocorticoid receptor. The aim of the present study was to further understand the Fkbp5/FKBP5-related genetic underpinnings underlying the relationship between early life social relations and alcohol drinking. The effect of maternal separation and voluntary alcohol drinking on Fkbp5 expression was investigated in the brain of young adult rats, whereas the interaction effect of the functional FKBP5 single nucleotide polymorphism rs1360780 genotype and parent-child relationship on problematic drinking was examined in young adult humans. In rats, Fkbp5 expression in the nucleus accumbens and ventral tegmental area, core regions of the reward system, was affected in a region-dependent manner and in opposite direction by maternal separation and alcohol drinking. Fkbp5 expression in the cingulate cortex was affected by the combined effect of maternal separation and alcohol drinking. In humans, the TT genotype, in the presence of a poor relationship between the child and parents, was associated with problematic drinking behavior. The present findings suggest that Fkbp5 expression in mesocorticolimbic dopaminergic regions associates with early life stress-mediated sensitivity to alcohol drinking and that FKBP5 genotype interacts with parent-child relationship to influence alcohol drinking. These findings are the first to point to a role of FKBP5 in propensity to alcohol misuse and call for studies of the underlying molecular mechanisms to identify potential drug targets.
Collapse
Affiliation(s)
- Ingrid Nylander
- Department of Pharmaceutical Bioscience, Uppsala University, Box 591, SE-75124, Uppsala, Sweden
| | - Aniruddha Todkar
- Department of Neuroscience, Uppsala University, Box 593, SE-75124, Uppsala, Sweden
| | - Linnea Granholm
- Department of Pharmaceutical Bioscience, Uppsala University, Box 591, SE-75124, Uppsala, Sweden
| | - Maria Vrettou
- Department of Neuroscience, Uppsala University, Box 593, SE-75124, Uppsala, Sweden
| | - Megha Bendre
- Department of Neuroscience, Uppsala University, Box 593, SE-75124, Uppsala, Sweden
| | - Wout Boon
- Department of Neuroscience, Uppsala University, Box 593, SE-75124, Uppsala, Sweden
| | - Henrik Andershed
- School of Law, Psychology and Social Work, Örebro University, SE-70182, Örebro, Sweden
| | - Catherine Tuvblad
- School of Law, Psychology and Social Work, Örebro University, SE-70182, Örebro, Sweden
- Department of Psychology, University of Southern California, 3620 S. McClintock Ave, Los Angeles, CA, 90089-1061, USA
| | - Kent W Nilsson
- Centre for Clinical Research, Västerås Central Hospital, SE-72189, Västerås, Sweden
| | - Erika Comasco
- Department of Neuroscience, Uppsala University, Box 593, SE-75124, Uppsala, Sweden.
| |
Collapse
|
41
|
Stechschulte LA, Qiu B, Warrier M, Hinds TD, Zhang M, Gu H, Xu Y, Khuder SS, Russo L, Najjar SM, Lecka-Czernik B, Yong W, Sanchez ER. FKBP51 Null Mice Are Resistant to Diet-Induced Obesity and the PPARγ Agonist Rosiglitazone. Endocrinology 2016; 157:3888-3900. [PMID: 27442117 PMCID: PMC5045506 DOI: 10.1210/en.2015-1996] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
FK506-binding protein-51 (FKBP51) is a molecular cochaperone recently shown to be a positive regulator of peroxisome proliferator-activated receptor (PPAR)γ, the master regulator of adipocyte differentiation and function. In cellular models of adipogenesis, loss of FKBP51 not only reduced PPARγ activity but also reduced lipid accumulation, suggesting that FKBP51 knock-out (KO) mice might have insufficient development of adipose tissue and lipid storage ability. This model was tested by examining wild-type (WT) and FKBP51-KO mice under regular and high-fat diet conditions. Under both diets, FKBP51-KO mice were resistant to weight gain, hepatic steatosis, and had greatly reduced white adipose tissue (WAT) but higher amounts of brown adipose tissue. Under high-fat diet, KO mice were highly resistant to adiposity and exhibited reduced plasma lipids and elevated glucose and insulin tolerance. Profiling of perigonadal and sc WAT revealed elevated expression of brown adipose tissue lineage genes in KO mice that correlated increased energy expenditure and a shift of substrate oxidation to carbohydrates, as measured by indirect calorimetry. To directly test PPARγ involvement, WT and KO mice were fed rosiglitazone agonist. In WT mice, rosiglitazone induced whole-body weight gain, increased WAT mass, a shift of substrate oxidation to lipids, and elevated expression of PPARγ-regulated lipogenic genes in WAT. In contrast, KO mice had reduced rosiglitazone responses for these parameters. Our results identify FKBP51 as an important regulator of PPARγ in WAT and as a potential new target in the treatment of obesity and diabetes.
Collapse
|
42
|
Zheng D, Sabbagh JJ, Blair LJ, Darling AL, Wen X, Dickey CA. MicroRNA-511 Binds to FKBP5 mRNA, Which Encodes a Chaperone Protein, and Regulates Neuronal Differentiation. J Biol Chem 2016; 291:17897-906. [PMID: 27334923 DOI: 10.1074/jbc.m116.727941] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Indexed: 12/11/2022] Open
Abstract
Single nucleotide polymorphisms in the FKBP5 gene increase the expression of the FKBP51 protein and have been associated with increased risk for neuropsychiatric disorders such as major depression and post-traumatic stress disorder. Moreover, levels of FKBP51 are increased with aging and in Alzheimer disease, potentially contributing to disease pathogenesis. However, aside from its glucocorticoid responsiveness, little is known about what regulates FKBP5 In recent years, non-coding RNAs, and in particular microRNAs, have been shown to modulate disease-related genes and processes. The current study sought to investigate which miRNAs could target and functionally regulate FKBP5 Following in silico data mining and initial target expression validation, miR-511 was found to suppress FKBP5 mRNA and protein levels. Using luciferase p-miR-Report constructs and RNA pulldown assays, we confirmed that miR-511 bound directly to the 3'-UTR of FKBP5, validating the predicted gene-microRNA interaction. miR-511 suppressed glucocorticoid-induced up-regulation of FKBP51 in cells and primary neurons, demonstrating functional, disease-relevant control of the protein. Consistent with a regulator of FKBP5, miR-511 expression in the mouse brain decreased with age but increased following chronic glucocorticoid treatment. Analysis of the predicted target genes of miR-511 revealed that neurogenesis, neuronal development, and neuronal differentiation were likely controlled by these genes. Accordingly, miR-511 increased neuronal differentiation in cells and enhanced neuronal development in primary neurons. Collectively, these findings show that miR-511 is a functional regulator of FKBP5 and can contribute to neuronal differentiation.
Collapse
Affiliation(s)
- Dali Zheng
- From the Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida 33613
| | - Jonathan J Sabbagh
- From the Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida 33613
| | - Laura J Blair
- From the Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida 33613
| | - April L Darling
- From the Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida 33613
| | - Xiaoqi Wen
- From the Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida 33613
| | - Chad A Dickey
- From the Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida 33613
| |
Collapse
|
43
|
Bekiesch P, Basitta P, Apel AK. Challenges in the Heterologous Production of Antibiotics inStreptomyces. Arch Pharm (Weinheim) 2016; 349:594-601. [DOI: 10.1002/ardp.201600058] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/27/2016] [Accepted: 05/06/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Paulina Bekiesch
- Pharmaceutical Biology; Pharmaceutical Institute; Eberhard-Karls-Universität Tübingen; Tübingen Germany
- German Centre for Infection Research (DZIF); Partner Site Tübingen; Tübingen Germany
| | - Patrick Basitta
- Pharmaceutical Biology; Pharmaceutical Institute; Eberhard-Karls-Universität Tübingen; Tübingen Germany
- German Centre for Infection Research (DZIF); Partner Site Tübingen; Tübingen Germany
| | - Alexander K. Apel
- Pharmaceutical Biology; Pharmaceutical Institute; Eberhard-Karls-Universität Tübingen; Tübingen Germany
- German Centre for Infection Research (DZIF); Partner Site Tübingen; Tübingen Germany
| |
Collapse
|
44
|
Gaali S, Feng X, Hähle A, Sippel C, Bracher A, Hausch F. Rapid, Structure-Based Exploration of Pipecolic Acid Amides as Novel Selective Antagonists of the FK506-Binding Protein 51. J Med Chem 2016; 59:2410-22. [PMID: 26954324 DOI: 10.1021/acs.jmedchem.5b01355] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The FK506-binding protein 51 (FKBP51) is a key regulator of stress hormone receptors and an established risk factor for stress-related disorders. Drug development for FKBP51 has been impaired by the structurally similar but functionally opposing homologue FKBP52. High selectivity between FKBP51 and FKBP52 can be achieved by ligands that stabilize a recently discovered FKBP51-favoring conformation. However, drug-like parameters for these ligands remained unfavorable. In the present study, we replaced the potentially labile pipecolic ester group of previous FKBP51 ligands by various low molecular weight amides. This resulted in the first series of pipecolic acid amides, which had much lower molecular weights without affecting FKBP51 selectivity. We discovered a geminally substituted cyclopentyl amide as a preferred FKBP51-binding motif and elucidated its binding mode to provide a new lead structure for future drug optimization.
Collapse
Affiliation(s)
- Steffen Gaali
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry , Kraepelinstrasse 2, 80804 Munich, Germany
| | - Xixi Feng
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry , Kraepelinstrasse 2, 80804 Munich, Germany
| | - Andreas Hähle
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry , Kraepelinstrasse 2, 80804 Munich, Germany
| | - Claudia Sippel
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry , Kraepelinstrasse 2, 80804 Munich, Germany
| | - Andreas Bracher
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry , Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Felix Hausch
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry , Kraepelinstrasse 2, 80804 Munich, Germany
| |
Collapse
|
45
|
Gene-Stress-Epigenetic Regulation of FKBP5: Clinical and Translational Implications. Neuropsychopharmacology 2016; 41:261-74. [PMID: 26250598 PMCID: PMC4677131 DOI: 10.1038/npp.2015.235] [Citation(s) in RCA: 352] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/13/2022]
Abstract
Stress responses and related outcomes vary markedly across individuals. Elucidating the molecular underpinnings of this variability is of great relevance for developing individualized prevention strategies and treatments for stress-related disorders. An important modulator of stress responses is the FK506-binding protein 51 (FKBP5/FKBP51). FKBP5 acts as a co-chaperone that modulates not only glucocorticoid receptor activity in response to stressors but also a multitude of other cellular processes in both the brain and periphery. Notably, the FKBP5 gene is regulated via complex interactions among environmental stressors, FKBP5 genetic variants, and epigenetic modifications of glucocorticoid-responsive genomic sites. These interactions can result in FKBP5 disinhibition that has been shown to contribute to a number of aberrant phenotypes in both rodents and humans. Consequently, FKBP5 blockade may hold promise as treatment intervention for stress-related disorders, and recently developed selective FKBP5 blockers show encouraging results in vitro and in rodent models. Although risk for stress-related disorders is conferred by multiple environmental and genetic factors, the findings related to FKBP5 illustrate how a deeper understanding of the molecular and systemic mechanisms underlying specific gene-environment interactions may provide insights into the pathogenesis of stress-related disorders.
Collapse
|
46
|
Kitraki E, Nalvarte I, Alavian-Ghavanini A, Rüegg J. Developmental exposure to bisphenol A alters expression and DNA methylation of Fkbp5, an important regulator of the stress response. Mol Cell Endocrinol 2015; 417:191-9. [PMID: 26427651 DOI: 10.1016/j.mce.2015.09.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/09/2015] [Accepted: 09/24/2015] [Indexed: 11/20/2022]
Abstract
Bisphenol A (BPA), an abundant endocrine disruptor, affects stress-responsiveness and related behaviors in children. In rats, perinatal BPA exposure modifies stress response in pubertal offspring via unknown mechanisms. Here we examined possible epigenetic modifications in the glucocorticoid receptor gene and its regulator Fkbp5 in hypothalamus and hippocampus of exposed offspring. We found increased DNA methylation of Fkbp5 and reduced protein levels in the hippocampus of exposed male rats. Similar effects were obtained in a male hippocampal cell line when exposed to BPA during differentiation. The estrogen receptor (ER) antagonist ICI 182,780 or ERβ knock-down affected Fkbp5 expression and methylation similarly to BPA. Further, BPA's effect on Fkbp5 was abolished upon knock-down of ERβ, suggesting a role for this receptor in mediating BPA's effects on Fkbp5. These data demonstrate that developmental BPA exposure modifies Fkbp5 methylation and expression in male rats, which may be related to its impact on stress responsiveness.
Collapse
Affiliation(s)
- Efthymia Kitraki
- School of Health Sciences, National and Kapodistrian University of Athens, Thivon 2str, Athens 11527, Greece.
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden.
| | - Ali Alavian-Ghavanini
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine (CMM) L8:00, 171 76 Stockholm, Sweden; Swedish Toxicology Science Research Center (Swetox), Forskargatan 20, 15136 Södertälje, Sweden.
| | - Joëlle Rüegg
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine (CMM) L8:00, 171 76 Stockholm, Sweden; Swedish Toxicology Science Research Center (Swetox), Forskargatan 20, 15136 Södertälje, Sweden.
| |
Collapse
|
47
|
Feng X, Sippel C, Bracher A, Hausch F. Structure–Affinity Relationship Analysis of Selective FKBP51 Ligands. J Med Chem 2015; 58:7796-806. [DOI: 10.1021/acs.jmedchem.5b00785] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Xixi Feng
- Department
of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstrasse 2, 80804 Munich, Germany
| | - Claudia Sippel
- Department
of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstrasse 2, 80804 Munich, Germany
| | - Andreas Bracher
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Felix Hausch
- Department
of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstrasse 2, 80804 Munich, Germany
| |
Collapse
|
48
|
Abstract
Anxiety-related psychiatric disorders represent one of the largest health burdens worldwide. Single nucleotide polymorphisms of the FK506 binding protein 51 (FKBP51) gene have been repeatedly associated with anxiety-related disorders and stress sensitivity. Given the intimate relationship of stress and anxiety, we hypothesized that amygdala FKBP51 may mediate anxiety-related behaviors. Mimicking the stress effect by specifically overexpressing FKBP51 in the basolateral amygdala (BLA) or central amygdala resulted in increased anxiety-related behavior, respectively. In contrast, application of a highly selective FKBP51 point mutant antagonist, following FKBP51(mut) BLA-overexpression, reduced the anxiogenic phenotype. We subsequently tested a novel FKBP51 antagonist, SAFit2, in wild-type mice via BLA microinjections, which reduced anxiety-related behavior. Remarkably, the same effect was observed following peripheral administration of SAFit2. To our knowledge, this is the first in vivo study using a specific FKBP51 antagonist, thereby unraveling the role of FKBP51 and its potential as a novel drug target for the improved treatment of anxiety-related disorders.
Collapse
|
49
|
Enhanced Human Decidual Cell-Expressed FKBP51 May Promote Labor-Related Functional Progesterone Withdrawal. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [PMID: 26207680 DOI: 10.1016/j.ajpath.2015.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Sustained plasma progesterone (P4) levels suggest initiation of human term labor by functional P4 withdrawal, reflecting reduced progesterone receptor (PR) and/or glucocorticoid receptor (GR) expression or activity. The steroid-induced immunophilin cochaperone FKBP51 inhibits PR- and GR-mediated transcription, suggesting a labor-initiating role. Gestational age-matched decidual sections were immunostained for FKBP51 and decidual cell (DC) and interstitial trophoblast (IT) markers, vimentin and cytokeratin, respectively. Term DC cultures were incubated with vehicle (control), estradiol (E2) with or without medroxyprogesterone acetate, dexamethasone (Dex), or Organon 2058. FKBP51 histologic scoring was significantly higher in DC nuclei during labor versus prelabor decidua, whereas FKBP51 immunostaining was undetected in interstitial trophoblasts (P < 0.05). In term DC cultures, E2 + medroxyprogesterone acetate or E2 + Dex enhanced FKBP51 expression (P < 0.01), whereas E2 + Organon 2058 inhibited PR expression (P < 0.05), and E2 + Dex inhibited GR expression (P < 0.05). Unlike term DCs, FKBP51 was undetected in control or Dex-treated cultured third-trimester trophoblasts. Electrophoretic mobility shift assays revealed that FKPB51 overexpression or silencing in cultured DCs altered PR-DNA binding. Increased FKBP51 levels in term DCs during labor complement our prior in situ observations of significantly lower PR in labor versus prelabor DCs. In a milieu of sustained plasma P4 levels, these reciprocal changes will amplify functional P4 withdrawal in DCs via FKBP51-mediated PR resistance coupled with declining PR levels, whereas the lack of FKBP51 expression in interstitial trophoblasts suggests unopposed constitutive GR action.
Collapse
|
50
|
Jovicic MJ, Lukic I, Radojcic M, Adzic M, Maric NP. Modulation of c-Jun N-terminal kinase signaling and specific glucocorticoid receptor phosphorylation in the treatment of major depression. Med Hypotheses 2015; 85:291-4. [PMID: 26052031 DOI: 10.1016/j.mehy.2015.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 05/15/2015] [Accepted: 05/27/2015] [Indexed: 12/28/2022]
Abstract
Glucocorticoid resistance is a common finding in major depressive disorder. Increased glucocorticoid receptor (GR) phosphorylation at serine 226 is associated with increased glucocorticoid resistance. Previously we have demonstrated that depressed patients exhibit higher levels of GR phosphorylated at serine 226 compared to healthy controls. The enzyme that is involved in this specific GR phosphorylation is c-Jun N-terminal kinase (JNK). We propose that modulation of glucocorticoid phosphorylation at serine 226, by targeting JNK signaling pathway, could be a potential strategy for antidepressant treatment. We base this assumption on the results of previous research that examined GR phosphorylation and JNK signaling in animal models and human studies. We also discuss the potential challenges in targeting JNK signaling pathway in depression.
Collapse
Affiliation(s)
| | - Iva Lukic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Marija Radojcic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Nadja P Maric
- School of Medicine, University of Belgrade, Serbia; Clinic for Psychiatry, Clinical Center of Serbia, Belgrade, Serbia.
| |
Collapse
|