1
|
Jeanne X, Török Z, Vigh L, Prodromou C. The role of the FKBP51-Hsp90 complex in Alzheimer's disease: An emerging new drug target. Cell Stress Chaperones 2024; 29:792-804. [PMID: 39615785 DOI: 10.1016/j.cstres.2024.11.006] [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: 11/19/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
With increasing age comes the inevitable decline in proteostasis, where chaperone and co-chaperone activity becomes imbalanced. These changes lead to global disturbances and pathogenic rewiring of the chaperone system into epichaperones consisting of protein networks that are ultimately dysfunctional. Such imbalances in proteostasis may favor mechanisms that can lead to neurological diseases, such as Alzheimer's disease (AD). Consequently, there has been an increase in research activity toward finding small molecules that can re-balance the chaperone and co-chaperone machinery to counter the effects of disease resulting from old age. The Hsp90 co-chaperone FKBP51 has recently been identified as a protein whose induction not only increases with age but is elevated further in AD cells. Significantly, FKBP51 plays a role in the Hsp90-dependent isomerization of tau, which in turn influences its phosphorylation and susceptibility to aggregation. We hypothesize that FKBP51 is a major player that is able to elicit tauopathy in response to amyloid-beta senile plaques that damage the brain. We propose that elevated FKBP51 levels result in an abnormal FKBP51-Hsp90 activity that alters the normal processing of tau, which manifests as hyperphosphorylation and oligomerization of tau. Thus, the Hsp90-FKBP51 complex is emerging as a drug target against AD. In support of this idea, the structure of the FKBP51-Hsp90 complex was recently described, and significantly, the small-molecule dihydropyridine LA1011 was shown to be able to disrupt the Hsp90-FKBP51 complex. LA1011 was previously shown to effectively prevent neurodegeneration in the APPxPS1 AD transgenic mouse model. This review looks at the role of Hsp90 and its co-chaperones in AD with a focus on FKBP51.
Collapse
Affiliation(s)
- Xavier Jeanne
- Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, Falmer BN1 9QG, UK
| | - Zsolt Török
- LipidArt Research and Development Ltd, Szeged, Temesvári Street 62, H-6726, Hungary
| | - László Vigh
- LipidArt Research and Development Ltd, Szeged, Temesvári Street 62, H-6726, Hungary
| | - Chrisostomos Prodromou
- Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, Falmer BN1 9QG, UK.
| |
Collapse
|
2
|
DeGiosio RA, Needham PG, Andrews OA, Tristan H, Grubisha MJ, Brodsky JL, Camacho C, Sweet RA. Differential regulation of MAP2 by phosphorylation events in proline-rich versus C-terminal domains. FASEB J 2023; 37:e23194. [PMID: 37702880 PMCID: PMC10539048 DOI: 10.1096/fj.202300486r] [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: 03/13/2023] [Revised: 07/31/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023]
Abstract
MAP2 is a critical cytoskeletal regulator in neurons. The phosphorylation of MAP2 (MAP2-P) is well known to regulate core functions of MAP2, including microtubule (MT)/actin binding and facilitation of tubulin polymerization. However, site-specific studies of MAP2-P function in regions outside of the MT-binding domain (MTBD) are lacking. We previously identified a set of MAP2 phosphopeptides which are differentially expressed and predominantly increased in the cortex of individuals with schizophrenia relative to nonpsychiatric comparison subjects. The phosphopeptides originated not from the MTBD, but from the flanking proline-rich and C-terminal domains of MAP2. We sought to understand the contribution of MAP2-P at these sites on MAP2 function. To this end, we isolated a series of phosphomimetic MAP2C constructs and subjected them to cell-free tubulin polymerization, MT-binding, actin-binding, and actin polymerization assays. A subset of MAP2-P events significantly impaired these functions, with the two domains displaying different patterns of MAP2 regulation: proline-rich domain mutants T293E and T300E impaired MT assembly and actin-binding affinity but did not affect MT-binding, while C-terminal domain mutants S426E and S439D impaired all three functions. S443D also impaired MT assembly with minimal effects on MT- or actin-binding. Using heterologous cells, we also found that S426E but not T293E had a lower capability for process formation than the wild-type protein. These findings demonstrate the functional utility of MAP2-P in the proline-rich and C-terminal domains and point to distinct, domain-dependent regulations of MAP2 function, which can go on to affect cellular morphology.
Collapse
Affiliation(s)
- R A DeGiosio
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - P G Needham
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - O A Andrews
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - H Tristan
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - M J Grubisha
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - J L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - C Camacho
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - R A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Jiang L, Chakraborty P, Zhang L, Wong M, Hill SE, Webber CJ, Libera J, Blair LJ, Wolozin B, Zweckstetter M. Chaperoning of specific tau structure by immunophilin FKBP12 regulates the neuronal resilience to extracellular stress. SCIENCE ADVANCES 2023; 9:eadd9789. [PMID: 36724228 PMCID: PMC9891691 DOI: 10.1126/sciadv.add9789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/05/2023] [Indexed: 06/18/2023]
Abstract
Alzheimer's disease and related tauopathies are characterized by the pathogenic misfolding and aggregation of the microtubule-associated protein tau. Understanding how endogenous chaperones modulate tau misfolding could guide future therapies. Here, we show that the immunophilin FKBP12, the 12-kDa FK506-binding protein (also known as FKBP prolyl isomerase 1A), regulates the neuronal resilience by chaperoning a specific structure in monomeric tau. Using a combination of mouse and cell experiments, in vitro aggregation experiments, nuclear magnetic resonance-based structural analysis of monomeric tau, site-specific phosphorylation and mutation, as well as structure-based analysis using the neural network-based structure prediction program AlphaFold, we define the molecular factors that govern the binding of FKBP12 to tau and its influence on tau-induced neurotoxicity. We further demonstrate that tyrosine phosphorylation of tau blocks the binding of FKBP12 to two highly specific structural motifs in tau. Our data together with previous results demonstrating FKBP12/tau colocalization in neurons and neurofibrillary tangles support a critical role of FKBP12 in regulating tau pathology.
Collapse
Affiliation(s)
- Lulu Jiang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Pijush Chakraborty
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Göttingen, Germany
| | - Lushuang Zhang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Melissa Wong
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Shannon E. Hill
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Chelsea Joy Webber
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jenna Libera
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Laura J. Blair
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33612, USA
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Neurophotonics, Boston University, Boston, MA 02215, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Göttingen, Germany
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Faßberg 11, 37077 Göttingen, Germany
| |
Collapse
|
4
|
Ortiz NR, Guy N, Garcia YA, Sivils JC, Galigniana MD, Cox MB. Functions of the Hsp90-Binding FKBP Immunophilins. Subcell Biochem 2023; 101:41-80. [PMID: 36520303 DOI: 10.1007/978-3-031-14740-1_2] [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] [Indexed: 12/23/2022]
Abstract
The Hsp90 chaperone is known to interact with a diverse array of client proteins. However, in every case examined, Hsp90 is also accompanied by a single or several co-chaperone proteins. One class of co-chaperone contains a tetratricopeptide repeat (TPR) domain that targets the co-chaperone to the C-terminal region of Hsp90. Within this class are Hsp90-binding peptidylprolyl isomerases, most of which belong to the FK506-binding protein (FKBP) family. Despite the common association of FKBP co-chaperones with Hsp90, it is abundantly clear that the client protein influences, and is often influenced by, the particular FKBP bound to Hsp90. Examples include Xap2 in aryl hydrocarbon receptor complexes and FKBP52 in steroid receptor complexes. In this chapter, we discuss the known functional roles played by FKBP co-chaperones and, where possible, relate distinctive functions to structural differences between FKBP members.
Collapse
Affiliation(s)
- Nina R Ortiz
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Naihsuan Guy
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Yenni A Garcia
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Jeffrey C Sivils
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Mario D Galigniana
- Departamento de Química Biológica/IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires, Argentina
| | - Marc B Cox
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, USA.
| |
Collapse
|
5
|
Barge S, Jade D, Ayyamperumal S, Manna P, Borah J, Nanjan CMJ, Nanjan MJ, Talukdar NC. Potential inhibitors for FKBP51: an in silico study using virtual screening, molecular docking and molecular dynamics simulation. J Biomol Struct Dyn 2022; 40:13799-13811. [PMID: 34709133 DOI: 10.1080/07391102.2021.1994877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Over the years, FK506-binding proteins have been targeted for different pharmaceutical interests. The FK506-binding protein, encoded by the FKBP5 gene, is responsible for stress and metabolic-related disorders, including cancer. In addition, the FKBD-I domain of the protein is a potential target for endocrine-related physiological diseases. In the present study, a set of natural compounds from the ZINC database was screened against FKBP51 protein using in silico strategy, namely pharmacophore modeling, molecular docking, and molecular dynamic simulation. A protein-ligand-based pharmacophore model workflow was employed to identify small molecules. The resultant compounds were then assessed for their toxicity using ADMET prediction. Based on ADMET prediction, 4768 compounds were selected for molecular docking to elucidate their binding mode. Based on the binding energy, 857 compounds were selected, and their Similarity Tanimoto coefficient was calculated, followed by clustering according to Jarvis-Patrick clustering methods (Jarp). The clustered singletons resulted in 14 hit compounds. The top 05 hit compounds and 05 known compounds were then subjected to 100 ns MD simulation to check the stability of complexes. The study revealed that the selected complexes are stable throughout the 100 ns simulation; for FKBD-I (4TW6), crystal structure compared with FKBP-51 (1KT0) crystal structure. Finally, the binding free energies of the hit complexes were calculated using molecular mechanics energies combined with Poisson-Boltzmann. The data reveal that all the complexes show negative BFEs, indicating a good affinity of the hit compounds to the protein. The top five compounds are, therefore, potential inhibitors for FKBP51. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sagar Barge
- Biochemistry and Drug Discovery Lab, Institute of Advanced Study in Science and Technology, Assam, India.,Department of Molecular Biology and Biotechnology, Cotton University, Panbazar, Assam, India
| | - Dhananjay Jade
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty, Tamil Nadu, India
| | - Selvaraj Ayyamperumal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty, Tamil Nadu, India
| | - Prasenjit Manna
- Biochemistry and Drug Discovery Lab, Institute of Advanced Study in Science and Technology, Assam, India
| | - Jagat Borah
- Biochemistry and Drug Discovery Lab, Institute of Advanced Study in Science and Technology, Assam, India
| | | | | | - Narayan Chandra Talukdar
- Biochemistry and Drug Discovery Lab, Institute of Advanced Study in Science and Technology, Assam, India.,Assam Down Town University, Panikhaiti, Guwahati, Assam, India
| |
Collapse
|
6
|
Sinsky J, Pichlerova K, Hanes J. Tau Protein Interaction Partners and Their Roles in Alzheimer's Disease and Other Tauopathies. Int J Mol Sci 2021; 22:9207. [PMID: 34502116 PMCID: PMC8431036 DOI: 10.3390/ijms22179207] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Tau protein plays a critical role in the assembly, stabilization, and modulation of microtubules, which are important for the normal function of neurons and the brain. In diseased conditions, several pathological modifications of tau protein manifest. These changes lead to tau protein aggregation and the formation of paired helical filaments (PHF) and neurofibrillary tangles (NFT), which are common hallmarks of Alzheimer's disease and other tauopathies. The accumulation of PHFs and NFTs results in impairment of physiological functions, apoptosis, and neuronal loss, which is reflected as cognitive impairment, and in the late stages of the disease, leads to death. The causes of this pathological transformation of tau protein haven't been fully understood yet. In both physiological and pathological conditions, tau interacts with several proteins which maintain their proper function or can participate in their pathological modifications. Interaction partners of tau protein and associated molecular pathways can either initiate and drive the tau pathology or can act neuroprotective, by reducing pathological tau proteins or inflammation. In this review, we focus on the tau as a multifunctional protein and its known interacting partners active in regulations of different processes and the roles of these proteins in Alzheimer's disease and tauopathies.
Collapse
Affiliation(s)
| | | | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia; (J.S.); (K.P.)
| |
Collapse
|
7
|
Gong LB, Zhang C, Yu RX, Li C, Fan YB, Liu YP, Qu XJ. FKBP10 Acts as a New Biomarker for Prognosis and Lymph Node Metastasis of Gastric Cancer by Bioinformatics Analysis and in Vitro Experiments. Onco Targets Ther 2020; 13:7399-7409. [PMID: 32801763 PMCID: PMC7395699 DOI: 10.2147/ott.s253154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose To explore the role of FKBP prolyl isomerase 10 (FKBP10) protein in the progression of gastric cancer. Methods Four independent gastric cancer databases (GSE27342, GSE29272, GSE54129 and TCGA-STAD) were used to identify differentially expressed genes (DEGs). Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was used to identify the abnormally active pathways in patients with gastric cancer. Univariate Cox regression analysis was used to identify genes with stable prognostic value in gastric cancer patients based on three independent gastric cancer databases (GSE15459, GSE62254, TCGA-STAD). Gene set enrichment analysis (GSEA) was used to explore the possible pathways related to FKBP10. The reverse transcription-polymerase chain reaction (RT-PCR) was employed to determine the expression of FKBP10 mRNA in the HGC-27 and MKN-7 cell lines. Adhesion assay was used to detect changes in cell adhesion ability. FKBP10, ITGA1, ITGA2, ITGA5, ITGAV, ITGA6, P-AKT473, P-AKT308, AKT, and β-actin were evaluated by Western blot (WB). Results We first performed differential expression genes (DEGs) screening of four independent GC databases (GSE27342, GSE29272, GSE54129 and TCGA-STAD). Eighty-nine genes showed consistent up-regulation in GC, the results of pathway analysis showed that they were related to “Focal adhesion”. The prognostic value of these 89 genes was tested in three independent GC databases GSE15459, GSE62254 and TCGA-STAD cohort. Finally, 12 genes, in which the expression of FKBP10 was prominently increased in patients with lymph node metastasis (LNM), showed stable prognostic value. The following gene set enrichment analysis (GSEA) also showed that FKBP10 is mainly involved in cell adhesion process, while adhesion experiments confirmed that cell adhesion was down-regulated after silencing FKBP10 in GC cells, and adhesion-related molecules integrin αV and α6 were down-regulated. Conclusion FKBP10 may be used as a marker for lymph node metastasis of GC and could be used as a potential target for future treatment of GC.
Collapse
Affiliation(s)
- Li-Bao Gong
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Chuang Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Ruo-Xi Yu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yi-Bo Fan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yun-Peng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Xiu-Juan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
8
|
Post-translational modifications and stress adaptation: the paradigm of FKBP51. Biochem Soc Trans 2020; 48:441-449. [PMID: 32318709 PMCID: PMC7200631 DOI: 10.1042/bst20190332] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 01/22/2023]
Abstract
Adaptation to stress is a fundamental requirement to cope with changing environmental conditions that pose a threat to the homeostasis of cells and organisms. Post-translational modifications (PTMs) of proteins represent a possibility to quickly produce proteins with new features demanding relatively little cellular resources. FK506 binding protein (FKBP) 51 is a pivotal stress protein that is involved in the regulation of several executers of PTMs. This mini-review discusses the role of FKBP51 in the function of proteins responsible for setting the phosphorylation, ubiquitination and lipidation of other proteins. Examples include the kinases Akt1, CDK5 and GSK3β, the phosphatases calcineurin, PP2A and PHLPP, and the ubiquitin E3-ligase SKP2. The impact of FKBP51 on PTMs of signal transduction proteins significantly extends the functional versatility of this protein. As a stress-induced protein, FKBP51 uses re-setting of PTMs to relay the effect of stress on various signaling pathways.
Collapse
|
9
|
Ulagesan S, Choi JW, Nam TJ, Choi YH. Peptidyl-prolyl isomerase and the biological activities of recombinant protein cyclophilin from Pyropia yezoensis (PyCyp). Protein Expr Purif 2020; 172:105636. [PMID: 32272150 DOI: 10.1016/j.pep.2020.105636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023]
Abstract
Cyclophilins are highly conserved proteins associated with peptidyl-prolyl cis-trans isomerase activity (PPIase). The present study was designed to analyze the biological activity of recombinant cyclophilin from the marine red algae Pyropia yezoensis (PyCyp). The cyclophilin gene from P. yezoensis was cloned into the pPROEX-HTA expression vector. The plasmid was transformed into BL21 Escherichia coli by high efficiency transformation. Recombinant protein was expressed using 0.1 mM IPTG and the fusion protein was purified by affinity column chromatography. The His-tag was removed by TEV protease. The recombinant protein was further purified on a HiPrep Sephacryl S-200 HR column and by reversed-phase high performance liquid chromatography with a Sep-pak plus C18 column. Purified cyclophilin was characterized by a variety of analytical methods and analyzed for its peptidyl-prolyl isomerase activity. Our recombinant PyCyp was shown to catalyze cis-trans isomerization. PyCyp was also evaluated for antimicrobial activity against both Gram-positive and Gram-negative bacteria cultures and showed significant antibacterial activity against tested pathogens. PyCyp was shown to permeabilize bacterial membranes as evidenced by increased fluorescence intensity in SYTOX Green uptake assays with Staphylococcus aureus. The radical scavenging activity of PyCyp increased in a dose-dependent manner, indicating significant antioxidant activity. This study provides information for the development of therapeutic proteins from marine algae.
Collapse
Affiliation(s)
- Selvakumari Ulagesan
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041, Republic of Korea
| | - Jeong-Wook Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041, Republic of Korea
| | - Taek-Jeong Nam
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041, Republic of Korea
| | - Youn-Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041, Republic of Korea; Department of Marine Bio-materials & Aquaculture, Pukyong National University, 45, Yongso-ro, Nam-Gu, Busan, 48513, Republic of Korea.
| |
Collapse
|
10
|
Sabbagh JJ, Cordova RA, Zheng D, Criado-Marrero M, Lemus A, Li P, Baker JD, Nordhues BA, Darling AL, Martinez-Licha C, Rutz DA, Patel S, Buchner J, Leahy JW, Koren J, Dickey CA, Blair LJ. Targeting the FKBP51/GR/Hsp90 Complex to Identify Functionally Relevant Treatments for Depression and PTSD. ACS Chem Biol 2018; 13:2288-2299. [PMID: 29893552 DOI: 10.1021/acschembio.8b00454] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Genetic and epigenetic alterations in FK506-binding protein 5 ( FKBP5) have been associated with increased risk for psychiatric disorders, including post-traumatic stress disorder (PTSD). Some of these common variants can increase the expression of FKBP5, the gene that encodes FKBP51. Excess FKBP51 promotes hypothalamic-pituitary-adrenal (HPA) axis dysregulation through altered glucocorticoid receptor (GR) signaling. Thus, we hypothesized that GR activity could be restored by perturbing FKBP51. Here, we screened 1280 pharmacologically active compounds and identified three compounds that rescued FKBP51-mediated suppression of GR activity without directly activating GR. One of the three compounds, benztropine mesylate, disrupted the association of FKBP51 with the GR/Hsp90 complex in vitro. Moreover, we show that removal of FKBP51 from this complex by benztropine restored GR localization in ex vivo brain slices and primary neurons from mice. In conclusion, we have identified a novel disruptor of the FKBP51/GR/Hsp90 complex. Targeting this complex may be a viable approach to developing treatments for disorders related to aberrant FKBP51 expression.
Collapse
Affiliation(s)
- Jonathan J. Sabbagh
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Ricardo A. Cordova
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Dali Zheng
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Marangelie Criado-Marrero
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Andrea Lemus
- Department of Chemistry, University of South Florida, Tampa, Florida, United States of America
| | - Pengfei Li
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Jeremy D. Baker
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Bryce A. Nordhues
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - April L. Darling
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Carlos Martinez-Licha
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Daniel A. Rutz
- Department Chemie, Technische Universität München, 85748 Munich, Germany
| | - Shreya Patel
- Department of Chemistry, University of South Florida, Tampa, Florida, United States of America
| | - Johannes Buchner
- Department Chemie, Technische Universität München, 85748 Munich, Germany
| | - James W. Leahy
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- Department of Chemistry, University of South Florida, Tampa, Florida, United States of America
- Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida, United States of America
| | - John Koren
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Chad A. Dickey
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| | - Laura J. Blair
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, United States of America
- USF Health Byrd Institute, University of South Florida, Tampa, Florida, United States of America
| |
Collapse
|
11
|
Ghartey-Kwansah G, Li Z, Feng R, Wang L, Zhou X, Chen FZ, Xu MM, Jones O, Mu Y, Chen S, Bryant J, Isaacs WB, Ma J, Xu X. Comparative analysis of FKBP family protein: evaluation, structure, and function in mammals and Drosophila melanogaster. BMC DEVELOPMENTAL BIOLOGY 2018; 18:7. [PMID: 29587629 PMCID: PMC5870485 DOI: 10.1186/s12861-018-0167-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 03/12/2018] [Indexed: 12/19/2022]
Abstract
Background FK506-binding proteins (FKBPs) have become the subject of considerable interest in several fields, leading to the identification of several cellular and molecular pathways in which FKBPs impact prenatal development and pathogenesis of many human diseases. Main body This analysis revealed differences between how mammalian and Drosophila FKBPs mechanisms function in relation to the immunosuppressant drugs, FK506 and rapamycin. Differences that could be used to design insect-specific pesticides. (1) Molecular phylogenetic analysis of FKBP family proteins revealed that the eight known Drosophila FKBPs share homology with the human FKBP12. This indicates a close evolutionary relationship, and possible origination from a common ancestor. (2) The known FKBPs contain FK domains, that is, a prolyl cis/trans isomerase (PPIase) domain that mediates immune suppression through inhibition of calcineurin. The dFKBP59, CG4735/Shutdown, CG1847, and CG5482 have a Tetratricopeptide receptor domain at the C-terminus, which regulates transcription and protein transportation. (3) FKBP51 and FKBP52 (dFKBP59), along with Cyclophilin 40 and protein phosphatase 5, function as Hsp90 immunophilin co-chaperones within steroid receptor-Hsp90 heterocomplexes. These immunophilins are potential drug targets in pathways associated with normal physiology and may be used to treat a variety of steroid-based diseases by targeting exocytic/endocytic cycling and vesicular trafficking. (4) By associating with presinilin, a critical component of the Notch signaling pathway, FKBP14 is a downstream effector of Notch activation at the membrane. Meanwhile, Shutdown associates with transposons in the PIWI-interacting RNA pathway, playing a crucial role in both germ cells and ovarian somas. Mutations in or silencing of dFKBPs lead to early embryonic lethality in Drosophila. Therefore, further understanding the mechanisms of FK506 and rapamycin binding to immunophilin FKBPs in endocrine, cardiovascular, and neurological function in both mammals and Drosophila would provide prospects in generating unique, insect specific therapeutics targeting the above cellular signaling pathways. Conclusion This review will evaluate the functional roles of FKBP family proteins, and systematically summarize the similarities and differences between FKBP proteins in Drosophila and Mammals. Specific therapeutics targeting cellular signaling pathways will also be discussed.
Collapse
Affiliation(s)
- George Ghartey-Kwansah
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, Xi'an, 710062, China.,Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences, Xi'an, 710062, China
| | - Zhongguang Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, Xi'an, 710062, China.,Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences, Xi'an, 710062, China
| | - Rui Feng
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, Xi'an, 710062, China.,Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences, Xi'an, 710062, China
| | - Liyang Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, Xi'an, 710062, China.,Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences, Xi'an, 710062, China
| | - Xin Zhou
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, Xi'an, 710062, China.,Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences, Xi'an, 710062, China.,Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Meng Meng Xu
- Department of Pharmacology, Duke University Medical Center, Durham, NC, USA
| | - Odell Jones
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yulian Mu
- State Key Laboratory for Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | | | - Joseph Bryant
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Jianjie Ma
- Ohio State University College of Medicine, Columbus, OH, USA
| | - Xuehong Xu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, Xi'an, 710062, China. .,Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences, Xi'an, 710062, China.
| |
Collapse
|
12
|
Dilworth D, Gudavicius G, Xu X, Boyce AKJ, O’Sullivan C, Serpa JJ, Bilenky M, Petrochenko EV, Borchers CH, Hirst M, Swayne LA, Howard P, Nelson CJ. The prolyl isomerase FKBP25 regulates microtubule polymerization impacting cell cycle progression and genomic stability. Nucleic Acids Res 2018; 46:2459-2478. [PMID: 29361176 PMCID: PMC5861405 DOI: 10.1093/nar/gky008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/14/2017] [Accepted: 01/12/2018] [Indexed: 12/25/2022] Open
Abstract
FK506 binding proteins (FKBPs) catalyze the interconversion of cis-trans proline conformers in proteins. Importantly, FK506 drugs have anti-cancer and neuroprotective properties, but the effectors and mechanisms underpinning these properties are not well understood because the cellular function(s) of most FKBP proteins are unclear. FKBP25 is a nuclear prolyl isomerase that interacts directly with nucleic acids and is associated with several DNA/RNA binding proteins. Here, we show the catalytic FKBP domain binds microtubules (MTs) directly to promote their polymerization and stabilize the MT network. Furthermore, FKBP25 associates with the mitotic spindle and regulates entry into mitosis. This interaction is important for mitotic spindle dynamics, as we observe increased chromosome instability in FKBP25 knockdown cells. Finally, we provide evidence that FKBP25 association with chromatin is cell-cycle regulated by Protein Kinase C phosphorylation. This disrupts FKBP25-DNA contacts during mitosis while maintaining its interaction with the spindle apparatus. Collectively, these data support a model where FKBP25 association with chromatin and MTs is carefully choreographed to ensure faithful genome duplication. Additionally, they highlight that FKBP25 is a MT-associated FK506 receptor and potential therapeutic target in MT-associated diseases.
Collapse
Affiliation(s)
- David Dilworth
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Geoff Gudavicius
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Xiaoxue Xu
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Andrew K J Boyce
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Connor O’Sullivan
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Jason J Serpa
- University of Victoria Genome BC Proteomics Centre, Vancouver Island Technology Park, Victoria, BC, V8Z 7X8, Canada
| | - Misha Bilenky
- BC Cancer Agency Genome Sciences Centre and the Department of Microbiology & Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Evgeniy V Petrochenko
- University of Victoria Genome BC Proteomics Centre, Vancouver Island Technology Park, Victoria, BC, V8Z 7X8, Canada
| | - Christoph H Borchers
- University of Victoria Genome BC Proteomics Centre, Vancouver Island Technology Park, Victoria, BC, V8Z 7X8, Canada
| | - Martin Hirst
- BC Cancer Agency Genome Sciences Centre and the Department of Microbiology & Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Perry Howard
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Christopher J Nelson
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| |
Collapse
|
13
|
Duan AR, Jonasson EM, Alberico EO, Li C, Scripture JP, Miller RA, Alber MS, Goodson HV. Interactions between Tau and Different Conformations of Tubulin: Implications for Tau Function and Mechanism. J Mol Biol 2017; 429:1424-1438. [PMID: 28322917 DOI: 10.1016/j.jmb.2017.03.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/22/2017] [Accepted: 03/12/2017] [Indexed: 11/16/2022]
Abstract
Tau is a multifaceted neuronal protein that stabilizes microtubules (MTs), but the mechanism of this activity remains poorly understood. Questions include whether Tau binds MTs laterally or longitudinally and whether Tau's binding affinity depends on the nucleotide state of tubulin. We observed that Tau binds tightly to Dolastatin-10 tubulin rings and promotes the formation of Dolastatin-10 ring stacks, implying that Tau can crosslink MT protofilaments laterally. In addition, we found that Tau prefers GDP-like tubulin conformations, which implies that Tau binding to the MT surface is biased away from the dynamic GTP-rich MT tip. To investigate the potential impact of these Tau activities on MT stabilization, we incorporated them into our previously developed dimer-scale computational model of MT dynamics. We found that lateral crosslinking activities have a much greater effect on MT stability than do longitudinal crosslinking activities, and that introducing a bias toward GDP tubulin has little impact on the observed MT stabilization. To address the question of why Tau is GDP-tubulin-biased, we tested whether Tau might affect MT binding of the +TIP EB1. We confirmed recent reports that Tau binds directly to EB1 and that Tau competes with EB1 for MT binding. Our results lead to a conceptual model where Tau stabilizes the MT lattice by strengthening lateral interactions between protofilaments. We propose that Tau's GDP preference allows the cell to independently regulate the dynamics of the MT tip and the stability of the lattice.
Collapse
Affiliation(s)
- Aranda R Duan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Erin M Jonasson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Emily O Alberico
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Chunlei Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jared P Scripture
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel A Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Mark S Alber
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Mathematics, University of California, Riverside, CA 92521, USA
| | - Holly V Goodson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
14
|
Inda C, Bolaender A, Wang T, Gandu SR, Koren J. Stressing Out Hsp90 in Neurotoxic Proteinopathies. Curr Top Med Chem 2017; 16:2829-38. [PMID: 27072699 DOI: 10.2174/1568026616666160413141350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/05/2016] [Accepted: 01/17/2016] [Indexed: 12/12/2022]
Abstract
A toxic accumulation of proteins is the hallmark pathology of several neurodegenerative disorders. Protein accumulation is regularly prevented by the network of molecular chaperone proteins, including and especially Hsp90. For reasons not yet elucidated, Hsp90 and the molecular chaperones interact with, but do not degrade, these toxic proteins resulting in the pathogenic accumulation of proteins such as tau, in Alzheimer's Disease, and α-synuclein, in Parkinson's Disease. In this review, we describe the associations between Hsp90 and the pathogenic and driver proteins of several neurodegenerative disorders. We additionally describe how the inhibition of Hsp90 promotes the degradation of both mutant and pathogenic protein species in models of neurodegenerative diseases. We also examine the current state of Hsp90 inhibitors capable of crossing the blood-brain barrier; compounds which may be capable of slowing, preventing, and possible reversing neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | - John Koren
- Program in Chemical Biology, Memorial Sloan-Kettering Cancer Center, New York, USA.
| |
Collapse
|
15
|
Dunyak BM, Gestwicki JE. Peptidyl-Proline Isomerases (PPIases): Targets for Natural Products and Natural Product-Inspired Compounds. J Med Chem 2016; 59:9622-9644. [PMID: 27409354 DOI: 10.1021/acs.jmedchem.6b00411] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptidyl-proline isomerases (PPIases) are a chaperone superfamily comprising the FK506-binding proteins (FKBPs), cyclophilins, and parvulins. PPIases catalyze the cis/trans isomerization of proline, acting as a regulatory switch during folding, activation, and/or degradation of many proteins. These "clients" include proteins with key roles in cancer, neurodegeneration, and psychiatric disorders, suggesting that PPIase inhibitors could be important therapeutics. However, the active site of PPIases is shallow, solvent-exposed, and well conserved between family members, making selective inhibitor design challenging. Despite these hurdles, macrocyclic natural products, including FK506, rapamycin, and cyclosporin, bind PPIases with nanomolar or better affinity. De novo attempts to derive new classes of inhibitors have been somewhat less successful, often showcasing the "undruggable" features of PPIases. Interestingly, the most potent of these next-generation molecules tend to integrate features of the natural products, including macrocyclization or proline mimicry strategies. Here, we review recent developments and ongoing challenges in the inhibition of PPIases, with a focus on how natural products might inform the creation of potent and selective inhibitors.
Collapse
Affiliation(s)
- Bryan M Dunyak
- Department of Biological Chemistry, University of Michigan Medical School , 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States.,Department of Pharmaceutical Chemistry, University of California at San Francisco , 675 Nelson Rising Lane, San Francisco, California 94158, United States
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California at San Francisco , 675 Nelson Rising Lane, San Francisco, California 94158, United States
| |
Collapse
|
16
|
Young KA, Thompson PM, Cruz DA, Williamson DE, Selemon LD. BA11 FKBP5 expression levels correlate with dendritic spine density in postmortem PTSD and controls. Neurobiol Stress 2015; 2:67-72. [PMID: 26844242 PMCID: PMC4721476 DOI: 10.1016/j.ynstr.2015.07.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/21/2015] [Indexed: 01/07/2023] Open
Abstract
Genetic variants of the immunophilin FKBP5 have been implicated in susceptibility to post-traumatic stress disorder (PTSD) and other stress-related disorders. We examined the relationship between mushroom, stubby, thin and filopodial spine densities measured with Golgi staining and FKBP5 gene expression in the medial orbitofrontal cortex (BA11) in individuals diagnosed with PTSD and normal controls (n = 8/8). ANCOVA revealed PTSD cases had a significantly elevated density of stubby spines (29%, P < 0.037) and a trend for a reduction in mushroom spine density (25%, p < 0.082). Levels of FKBP5 mRNA were marginally elevated in the PTSD cases (z = 1.94, p = 0.053) and levels correlated inversely with mushroom (Spearman's rho = −0.83, p < 0.001) and overall spine density (rho = −0.75, p < 0.002) and directly with stubby spine density (rho = 0.55, p < 0.027). These data suggest that FKBP5 may participate in a cellular pathway modulating neuronal spine density changes in the brain, and that this pathway may be dysregulated in PTSD. The present study is one of the first human post-mortem PTSD studies to date. Extreme stress has robust repercussions on glucocorticoids and dendritic spine morphology in animal models. FKBP5, involved in glucocorticoid signaling, was inversely associated with mushroom spine density in frontal cortex. These findings are consistent with alterations in glucocorticoid signaling in PTSD affecting synaptic plasticity.
Collapse
Affiliation(s)
- Keith A Young
- Central Texas Veterans Health Care System, Temple, TX, USA; Department of Veterans Affairs, VISN 17 Center of Excellence for Research on Returning War Veterans, Waco, TX, USA; Department of Psychiatry and Behavioral Science, Texas A&M Health Science Center, Temple, TX, USA
| | - Peter M Thompson
- Department of Psychiatry and Southwest Brain Bank, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Dianne A Cruz
- Department of Psychiatry and Southwest Brain Bank, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Douglas E Williamson
- Department of Psychiatry and Southwest Brain Bank, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Lynn D Selemon
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
17
|
Guy NC, Garcia YA, Sivils JC, Galigniana MD, Cox MB. Functions of the Hsp90-binding FKBP immunophilins. Subcell Biochem 2015; 78:35-68. [PMID: 25487015 DOI: 10.1007/978-3-319-11731-7_2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hsp90 functionally interacts with a broad array of client proteins, but in every case examined Hsp90 is accompanied by one or more co-chaperones. One class of co-chaperone contains a tetratricopeptide repeat domain that targets the co-chaperone to the C-terminal region of Hsp90. Within this class are Hsp90-binding peptidylprolyl isomerases, most of which belong to the FK506-binding protein (FKBP) family. Despite the common association of FKBP co-chaperones with Hsp90, it is now clear that the client protein influences, and is influenced by, the particular FKBP bound to Hsp90. Examples include Xap2 in aryl hydrocarbon receptor complexes and FKBP52 in steroid receptor complexes. In this chapter, we discuss the known functional roles played by FKBP co-chaperones and, where possible, relate distinctive functions to structural differences between FKBP members.
Collapse
Affiliation(s)
- Naihsuan C Guy
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 79968, El Paso, TX, USA,
| | | | | | | | | |
Collapse
|
18
|
Kitagishi Y, Nakanishi A, Ogura Y, Matsuda S. Dietary regulation of PI3K/AKT/GSK-3β pathway in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2014; 6:35. [PMID: 25031641 PMCID: PMC4075129 DOI: 10.1186/alzrt265] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alzheimer’s disease (AD) is characterized by the formation of senile plaques and neurofibrillary tangles composed of phosphorylated Tau. Several findings suggest that correcting signal dysregulation for Tau phosphorylation in AD may offer a potential therapeutic approach. The PI3K/AKT/GSK-3β pathway has been shown to play a pivotal role in neuroprotection, enhancing cell survival by stimulating cell proliferation and inhibiting apoptosis. This pathway appears to be crucial in AD because it promotes protein hyper-phosphorylation in Tau. Understanding those regulations may provide a better efficacy of new therapeutic approaches. In this review, we summarize advances in the involvement of the PI3K/AKT/GSK-3β pathways in cell signaling of neuronal cells. We also review recent studies on the features of several diets and the signaling pathway involved in AD.
Collapse
Affiliation(s)
- Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - Atsuko Nakanishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - Yasunori Ogura
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| |
Collapse
|
19
|
Hung TC, Chang TT, Fan MJ, Lee CC, Chen CYC. In Silico Insight into Potent of Anthocyanin Regulation of FKBP52 to Prevent Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2014; 2014:450592. [PMID: 24899909 PMCID: PMC4036721 DOI: 10.1155/2014/450592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/03/2014] [Accepted: 01/03/2014] [Indexed: 02/01/2023]
Abstract
Alzheimer's disease (AD) is caused by the hyperphosphorylation of Tau protein aggregation. FKBP52 (FK506 binding protein 52) has been found to inhibit Tau protein aggregation. This study found six different kinds of anthocyanins that have high binding potential. After analyzing the docking positions, hydrophobic interactions, and hydrogen bond interactions, several amino acids were identified that play important roles in protein and ligand interaction. The proteins' variation is described using eigenvectors and the distance between the amino acids during a molecular dynamics simulation (MD). This study investigates the three loops based around Glu85, Tyr113, and Lys121-all of which are important in inducing FKBP52 activation. By performing a molecular dynamic simulation process between unbound proteins and the protein complex with FK506, it was found that ligand targets that docked onto the FK1 domain will decrease the distance between Glu85/Tyr113 and Glu85/Lys121. The FKBP52 structure variation may induce FKBP52 activation and inhibit Tau protein aggregation. The results indicate that anthocyanins might change the conformation of FKBP52 during binding. In addition, the purple anthocyanins, such as cyanidin-3-glucoside and malvidin-3-glucoside, might be better than FK506 in regulating FKBP52 and treating Alzheimer's disease.
Collapse
Affiliation(s)
- Tzu-Chieh Hung
- Department of Biomedical Informatics, Asia University, Taichung 41354, Taiwan
| | - Tung-Ti Chang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Ming-Jen Fan
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
| | - Cheng-Chun Lee
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Calvin Yu-Chian Chen
- Department of Biomedical Informatics, Asia University, Taichung 41354, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
20
|
Chaperone-interacting TPR proteins in Caenorhabditis elegans. J Mol Biol 2013; 425:2922-39. [PMID: 23727266 DOI: 10.1016/j.jmb.2013.05.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/30/2013] [Accepted: 05/22/2013] [Indexed: 11/21/2022]
Abstract
The ATP-hydrolyzing molecular chaperones Hsc70/Hsp70 and Hsp90 bind a diverse set of tetratricopeptide repeat (TPR)-containing cofactors via their C-terminal peptide motifs IEEVD and MEEVD. These cochaperones contribute to substrate turnover and confer specific activities to the chaperones. Higher eukaryotic genomes encode a large number of TPR-domain-containing proteins. The human proteome contains more than 200 TPR proteins, and that of Caenorhabditis elegans, about 80. It is unknown how many of them interact with Hsc70 or Hsp90. We systematically screened the C. elegans proteome for TPR-domain-containing proteins that likely interact with Hsc70 and Hsp90 and ranked them due to their similarity with known chaperone-interacting TPRs. We find C. elegans to encode many TPR proteins, which are not present in yeast. All of these have homologs in fruit fly or humans. Highly ranking uncharacterized open reading frames C33H5.8, C34B2.5 and ZK370.8 may encode weakly conserved homologs of the human proteins RPAP3, TTC1 and TOM70. C34B2.5 and ZK370.8 bind both Hsc70 and Hsp90 with low micromolar affinities. Mutation of amino acids involved in EEVD binding disrupts the interaction. In vivo, ZK370.8 is localized to mitochondria in tissues with known chaperone requirements, while C34B2.5 colocalizes with Hsc70 in intestinal cells. The highest-ranking open reading frame with non-conserved EEVD-interacting residues, F52H3.5, did not show any binding to Hsc70 or Hsp90, suggesting that only about 15 of the TPR-domain-containing proteins in C. elegans interact with chaperones, while the many others may have evolved to bind other ligands.
Collapse
|
21
|
Li XH, Chen C, Tu Y, Sun HT, Zhao ML, Cheng SX, Qu Y, Zhang S. Sirt1 Promotes Axonogenesis by Deacetylation of Akt and Inactivation of GSK3. Mol Neurobiol 2013; 48:490-9. [DOI: 10.1007/s12035-013-8437-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
|
22
|
Carman A, Kishinevsky S, Koren J, Lou W, Chiosis G. Chaperone-dependent Neurodegeneration: A Molecular Perspective on Therapeutic Intervention. ACTA ACUST UNITED AC 2013; 2013. [PMID: 25258700 PMCID: PMC4172285 DOI: 10.4172/2161-0460.s10-007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Maintenance of cellular homeostasis is regulated by the molecular chaperones. Under pathogenic conditions, aberrant proteins are triaged by the chaperone network. These aberrant proteins, known as "clients," have major roles in the pathogenesis of numerous neurological disorders, including tau in Alzheimer's disease, α-synuclein and LRRK2 in Parkinson's disease, SOD-1, TDP-43 and FUS in amyotrophic lateral sclerosis, and polyQ-expanded proteins such as huntingtin in Huntington's disease. Recent work has demonstrated that the use of chemical compounds which inhibit the activity of molecular chaperones subsequently alter the fate of aberrant clients. Inhibition of Hsp90 and Hsc70, two major molecular chaperones, has led to a greater understanding of how chaperone triage decisions are made and how perturbing the chaperone system can promote clearance of these pathogenic clients. Described here are major pathways and components of several prominent neurological disorders. Also discussed is how treatment with chaperone inhibitors, predominately Hsp90 inhibitors which are selective for a diseased state, can relieve the burden of aberrant client signaling in these neurological disorders.
Collapse
Affiliation(s)
- Aaron Carman
- Department of Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Centre, New York, NY, USA
| | - Sarah Kishinevsky
- Department of Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Centre, New York, NY, USA
| | - John Koren
- Department of Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Centre, New York, NY, USA
| | - Wenjie Lou
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY, USA
| | - Gabriela Chiosis
- Department of Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Centre, New York, NY, USA
| |
Collapse
|
23
|
Jasnovidova O, Stefl R. The CTD code of RNA polymerase II: a structural view. WILEY INTERDISCIPLINARY REVIEWS-RNA 2012; 4:1-16. [DOI: 10.1002/wrna.1138] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
24
|
Martić S, Beheshti S, Kraatz HB, Litchfield DW. Electrochemical Investigations of Tau Protein Phosphorylations and Interactions with Pin1. Chem Biodivers 2012; 9:1693-702. [DOI: 10.1002/cbdv.201100418] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
25
|
Abstract
Hsp90 is a highly abundant and ubiquitous molecular chaperone which plays an essential role in many cellular processes including cell cycle control, cell survival, hormone and other signalling pathways. It is important for the cell's response to stress and is a key player in maintaining cellular homeostasis. In the last ten years, it has become a major therapeutic target for cancer, and there has also been increasing interest in it as a therapeutic target in neurodegenerative disorders, and in the development of anti-virals and anti-protozoan infections. The focus of this review is the structural and mechanistic studies which have been performed in order to understand how this important chaperone acts on a wide variety of different proteins (its client proteins) and cellular processes. As with many of the other classes of molecular chaperone, Hsp90 has a critical ATPase activity, and ATP binding and hydrolysis known to modulate the conformational dynamics of the protein. It also uses a host of cochaperones which not only regulate the ATPase activity and conformational dynamics but which also mediate interactions with Hsp90 client proteins. The system is also regulated by post-translational modifications including phosphorylation and acetylation. This review discusses all these aspects of Hsp90 structure and function.
Collapse
|