1
|
García-Rojo G, Valenzuela Martínez I, Aguayo F, Muñoz-Llanos M, Ramírez D, Fiedler JL. The ROCK Inhibitor Fasudil and Sertraline Share Morphological and Molecular Effects in the Hippocampus of Chronically Stressed Rats: Exploring Common Antidepressant Pathways by Network Pharmacology. ACS Pharmacol Transl Sci 2025; 8:1292-1312. [PMID: 40370991 PMCID: PMC12070322 DOI: 10.1021/acsptsci.4c00680] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/26/2025] [Accepted: 03/27/2025] [Indexed: 05/16/2025]
Abstract
Despite the widespread use of selective serotonin reuptake inhibitors like sertraline, the intricate molecular mechanisms underlying major depression and the therapeutic efficacy of these treatments remain not fully elucidated. Building on our preliminary findings, this study investigates the antidepressant effects of fasudil, a Rho-associated protein kinase (ROCK) inhibitor typically utilized as a vasodilator and antispasmodic, and compares its effects with those of sertraline using a chronic restraint stress model in rats. Specifically, we examined the effects of chronic administration on dendritic spine density, key molecular survival pathways, and miRNA levels in the hippocampus. Adult male Sprague-Dawley rats were administered sertraline, fasudil (10 mg/kg/day), or saline over 14 days, with a subset experiencing daily restraint stress. Our findings demonstrate that both sertraline and fasudil effectively prevented stress-induced reductions in dendritic spine density and miR-138 levels in the rat hippocampus. Additionally, by employing a network pharmacology approach, we explored the converging molecular pathways influenced by both drugs, facilitating the identification of novel molecular targets and pathways implicated in the pathophysiology of depression and its treatment. Pharmacoinformatic analysis revealed common signaling cascades and critical proteins that may potentially underlie the observed pharmacological effects, contributing to a paradigm shift in understanding depression by integrating drug repurposing and network pharmacology, offering valuable insights into the underlying mechanisms of depression and the antidepressant effect from a new network-based paradigm rather than focusing solely on a single protein target.
Collapse
Affiliation(s)
- Gonzalo García-Rojo
- Laboratory
of Neuroplasticity and Neurogenetics, Department of Biochemistry and
Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, Chile
- Departamento
de Química, Facultad de Ciencias, Universidad de La Serena, La Serena 1700000, Chile
| | - Ignacio Valenzuela Martínez
- Departamento
de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile
- Doctorado
en Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile
| | - Felipe Aguayo
- Laboratory
of Neuroplasticity and Neurogenetics, Department of Biochemistry and
Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, Chile
| | - Mauricio Muñoz-Llanos
- Laboratory
of Neuroplasticity and Neurogenetics, Department of Biochemistry and
Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, Chile
| | - David Ramírez
- Departamento
de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile
| | - Jenny L. Fiedler
- Laboratory
of Neuroplasticity and Neurogenetics, Department of Biochemistry and
Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, Chile
| |
Collapse
|
2
|
Gu J, He Y, He C, Zhang Q, Huang Q, Bai S, Wang R, You Q, Wang L. Advances in the structures, mechanisms and targeting of molecular chaperones. Signal Transduct Target Ther 2025; 10:84. [PMID: 40069202 PMCID: PMC11897415 DOI: 10.1038/s41392-025-02166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Molecular chaperones, a class of complex client regulatory systems, play significant roles in the prevention of protein misfolding and abnormal aggregation, the modulation of protein homeostasis, and the protection of cells from damage under constantly changing environmental conditions. As the understanding of the biological mechanisms of molecular chaperones has increased, their link with the occurrence and progression of disease has suggested that these proteins are promising targets for therapeutic intervention, drawing intensive interest. Here, we review recent advances in determining the structures of molecular chaperones and heat shock protein 90 (HSP90) chaperone system complexes. We also describe the features of molecular chaperones and shed light on the complicated regulatory mechanism that operates through interactions with various co-chaperones in molecular chaperone cycles. In addition, how molecular chaperones affect diseases by regulating pathogenic proteins has been thoroughly analyzed. Furthermore, we focus on molecular chaperones to systematically discuss recent clinical advances and various drug design strategies in the preclinical stage. Recent studies have identified a variety of novel regulatory strategies targeting molecular chaperone systems with compounds that act through different mechanisms from those of traditional inhibitors. Therefore, as more novel design strategies are developed, targeting molecular chaperones will significantly contribute to the discovery of new potential drugs.
Collapse
Affiliation(s)
- Jinying Gu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yanyi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chenxi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qifei Huang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shangjun Bai
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Provincial TCM Engineering Technology Research Center of Highly Efficient Drug Delivery Systems (DDSs), Nanjing, China.
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
3
|
Gebru NT, Beaulieu-Abdelahad D, Gulick D, Blair LJ. FKBP51 overexpression in the corticolimbic system stabilizes circadian rhythms. Cell Stress Chaperones 2025; 30:22-32. [PMID: 39674313 PMCID: PMC11750455 DOI: 10.1016/j.cstres.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
Circadian rhythm disruptions have been associated with a wide range of health issues and complications, including an increased risk of circadian rhythm sleep disorders (CRSDs). CRSDs are common among individuals who have been through a traumatic event, particularly in those who have post-traumatic stress disorder (PTSD). Allelic variations in the gene encoding for FK506-binding protein 51 (FKBP51) can increase the susceptibility for PTSD and other stress-related disorders following trauma. At least one of these variants increases the levels of FKBP51 following stress through a glucocorticoid receptor-mediated process. Here, we used a mouse model that overexpresses human FKBP51 throughout the forebrain, rTgFKBP5, to investigate if elevated FKBP51 contributes to circadian rhythm disruption. Surprisingly, our findings indicate a greater rhythm amplitude and decreased rhythm fragmentation in rTgFKBP5 mice, particularly females, compared to controls. Female rTgFKBP5 mice also showed higher corticosterone levels basally and following stress exposure. Overall, this study associates FKBP51 overexpression with beneficial circadian rhythm outcomes.
Collapse
Affiliation(s)
- Niat T Gebru
- Byrd Alzheimer's Center and Research Institute, Tampa, FL 33613, USA; Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| | - David Beaulieu-Abdelahad
- Byrd Alzheimer's Center and Research Institute, Tampa, FL 33613, USA; Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Danielle Gulick
- Byrd Alzheimer's Center and Research Institute, Tampa, FL 33613, USA; Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Laura J Blair
- Byrd Alzheimer's Center and Research Institute, Tampa, FL 33613, USA; Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA; Research and Development, James A. Haley Veterans Hospital, Tampa, FL 33612, USA.
| |
Collapse
|
4
|
Fan X, Sun L, Qin Y, Liu Y, Wu S, Du L. The Role of HSP90 Molecular Chaperones in Depression: Potential Mechanisms. Mol Neurobiol 2025; 62:708-717. [PMID: 38896156 DOI: 10.1007/s12035-024-04284-4] [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: 04/28/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024]
Abstract
Major depressive disorder (MDD) is characterized by high rates of disability and death and has become a public health problem that threatens human life and health worldwide. HPA axis disorder and neuroinflammation are two common biological abnormalities in MDD patients. Hsp90 is an important molecular chaperone that is widely distributed in the organism. Hsp90 binds to the co-chaperone and goes through a molecular chaperone cycle to complete its regulation of the client protein. Numerous studies have demonstrated that Hsp90 regulates how the HPA axis reacts to stress and how GR, the HPA axis' responsive substrate, matures. In addition, Hsp90 exhibits pro-inflammatory effects that are closely related to neuroinflammation in MDD. Currently, Hsp90 inhibitors have made some progress in the treatment of a variety of human diseases, but they still need to be improved. Further insight into the role of Hsp90 in MDD provides new ideas for the development of new antidepressant drugs targeting Hsp90.
Collapse
Affiliation(s)
- Xuyuan Fan
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Lei Sun
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Ye Qin
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Yuan Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Shusheng Wu
- Department of the Central Laboratory, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| | - Longfei Du
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| |
Collapse
|
5
|
Gebru NT, Hill SE, Blair LJ. Genetically engineered mouse models of FK506-binding protein 5. J Cell Biochem 2024; 125:e30374. [PMID: 36780339 PMCID: PMC10423308 DOI: 10.1002/jcb.30374] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/25/2022] [Accepted: 01/15/2023] [Indexed: 02/14/2023]
Abstract
FK506 binding protein 51 (FKBP51) is a molecular chaperone that influences stress response. In addition to having an integral role in the regulation of steroid hormone receptors, including glucocorticoid receptor, FKBP51 has been linked with several biological processes including metabolism and neuronal health. Genetic and epigenetic alterations in the gene that encodes FKBP51, FKBP5, are associated with increased susceptibility to multiple neuropsychiatric disorders, which has fueled much of the research on this protein. Because of the complexity of these processes, animal models have been important in understanding the role of FKBP51. This review examines each of the current mouse models of FKBP5, which include whole animal knockout, conditional knockout, overexpression, and humanized mouse models. The generation of each model and observational details are discussed, including behavioral phenotypes, molecular changes, and electrophysiological alterations basally and following various challenges. While much has been learned through these models, there are still many aspects of FKBP51 biology that remain opaque and future studies are needed to help illuminate these current gaps in knowledge. Overall, FKBP5 continues to be an exciting potential target for stress-related disorders.
Collapse
Affiliation(s)
- Niat T. Gebru
- USF Health Byrd Alzheimer’s Institute, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Department of Molecular Medicine, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
| | - Shannon E. Hill
- USF Health Byrd Alzheimer’s Institute, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Department of Molecular Medicine, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
| | - Laura J. Blair
- USF Health Byrd Alzheimer’s Institute, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Department of Molecular Medicine, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Research Service, James A. Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, United States
| |
Collapse
|
6
|
Soto OB, Ramirez CS, Koyani R, Rodriguez-Palomares IA, Dirmeyer JR, Grajeda B, Roy S, Cox MB. Structure and function of the TPR-domain immunophilins FKBP51 and FKBP52 in normal physiology and disease. J Cell Biochem 2024; 125:e30406. [PMID: 37087733 PMCID: PMC10903107 DOI: 10.1002/jcb.30406] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/22/2023] [Accepted: 04/04/2023] [Indexed: 04/24/2023]
Abstract
Coordinated cochaperone interactions with Hsp90 and associated client proteins are crucial for a multitude of signaling pathways in normal physiology, as well as in disease settings. Research on the molecular mechanisms regulated by the Hsp90 multiprotein complexes has demonstrated increasingly diverse roles for cochaperones throughout Hsp90-regulated signaling pathways. Thus, the Hsp90-associated cochaperones have emerged as attractive therapeutic targets in a wide variety of disease settings. The tetratricopeptide repeat (TPR)-domain immunophilins FKBP51 and FKBP52 are of special interest among the Hsp90-associated cochaperones given their Hsp90 client protein specificity, ubiquitous expression across tissues, and their increasingly important roles in neuronal signaling, intracellular calcium release, peptide bond isomerization, viral replication, steroid hormone receptor function, and cell proliferation to name a few. This review summarizes the current knowledge of the structure and molecular functions of TPR-domain immunophilins FKBP51 and FKBP52, recent findings implicating these immunophilins in disease, and the therapeutic potential of targeting FKBP51 and FKBP52 for the treatment of disease.
Collapse
Affiliation(s)
- Olga B. Soto
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Christian S. Ramirez
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Rina Koyani
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Isela A. Rodriguez-Palomares
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Jessica R. Dirmeyer
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Brian Grajeda
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Sourav Roy
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
| | - Marc B. Cox
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968
| |
Collapse
|
7
|
González-Castro TB, Rodríguez-Fuentes I, Tovilla-Zárate CA, Juárez-Rojop IE, Hernández-Díaz Y, López-Narváez ML, Uresti-Rivera EE, Hernández-Vicencio JL. The role of SKA2 on affective disorder, post-traumatic stress disorder and suicide behavior: systematic review and in silico analysis. Metab Brain Dis 2024; 39:1005-1014. [PMID: 38722562 DOI: 10.1007/s11011-024-01346-3] [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: 08/28/2023] [Accepted: 05/04/2024] [Indexed: 07/10/2024]
Abstract
Genes involved in the hypothalamic-pituitary-adrenal axis may be a robust biomarker of psychiatric disorders. Genetic polymorphisms of the SKA2 gene are associated with several behavioral disorders. In this study, we embarked on a systematic search of all possible reports of genetic association with SKA2 and affective disorder, post-traumatic stress disorder, and suicide behavior; the functional consequences of nsSNPs were explored through computational tools with an in silico analysis. Eight eligible articles were included. Our study identified that SKA2 did not show association with risk of Major Depression Disorder. Epigenetic variation at SKA2 mediates vulnerability to Post-Traumatic Stress Disorder. Studies provide strong preliminary evidence that alterations at the SKA2 gene covary with types of suicide behavior, including suicidal ideation, attempts, and completions. Results from in silico analysis predicted that I22S, I22G, I78T, A15L, D18R, R25L, N42I, Y21S, K14I, K14L, and L60R were the most structurally and functionally significant nsSNPs in SKA2. Amino acid conservation analysis revealed that the amino acids were highly conserved and some dissimilarities of mutant type amino acids from wild-type amino acids such as charge, size, and hydrophobicity were observed. In the future, SKA2 gene have the potential to be evaluated as prognostic biomarkers for diagnosis and research.
Collapse
Affiliation(s)
- Thelma Beatriz González-Castro
- División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Méndez, Tabasco, México
| | - Itzel Rodríguez-Fuentes
- División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Méndez, Tabasco, México
| | - Carlos Alfonso Tovilla-Zárate
- División Académica Multidisciplinaria de Comalcalco, Universidad Juárez Autónoma de Tabasco, Comalcalco, Tabasco, México
| | - Isela Esther Juárez-Rojop
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, México
| | - Yazmín Hernández-Díaz
- División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Méndez, Tabasco, México.
| | | | - Edith Elena Uresti-Rivera
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, México
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, México
| | | |
Collapse
|
8
|
Wang L, Kumar R, Winblad B, Pavlov PF. Structure-based discovery of small molecule inhibitors of FKBP51-Hsp90 protein-protein interaction. Eur J Med Chem 2024; 270:116356. [PMID: 38579621 DOI: 10.1016/j.ejmech.2024.116356] [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: 02/06/2024] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/07/2024]
Abstract
The heat shock protein 90 kDa (Hsp90) molecular chaperone machinery is responsible for the folding and activation of hundreds of important clients such as kinases, steroid hormone receptors, transcription factors, etc. This process is dynamically regulated in an ATP-dependent manner by Hsp90 co-chaperones including a group of tetratricopeptide (TPR) motif proteins that bind to the C-terminus of Hsp90. Among these TPR containing co-chaperones, FK506-binding protein 51 kDa (FKBP51) is reported to play an important role in stress-related pathologies, psychiatric disorders, Alzheimer's disease, and cancer, making FKBP51-Hsp90 interaction a potential therapeutic target. In this study, we report identification of potent and selective inhibitors of FKBP51-Hsp90 protein-protein interaction using a structure-based virtual screening approach. Upon in vitro evaluation, the identified hits show a considerable degree of selectivity towards FKBP51 over other TPR proteins, particularly for highly homologous FKBP52. Tyr355 of FKBP51 emerged as an important contributor to inhibitor's specificity. Additionally, we demonstrate the impact of these inhibitors on cellular energy metabolism, and neurite outgrowth, which are subjects of FKBP51 regulation. Overall, the results from this study highlight a novel pharmacological approach towards regulation of FKBP51 function and more generally, Hsp90 function via its interaction with TPR co-chaperones.
Collapse
Affiliation(s)
- Lisha Wang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden.
| | - Rajnish Kumar
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), 221005, Varanasi, India.
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Theme Inflammation and Aging, Karolinska University Hospital, 14186, Huddinge, Sweden
| | - Pavel F Pavlov
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
| |
Collapse
|
9
|
Wang L, Wojcieszak J, Kumar R, Zhao Z, Sun X, Xie S, Winblad B, Pavlov PF. FKBP51-Hsp90 Interaction-Deficient Mice Exhibit Altered Endocrine Stress Response and Sex Differences Under High-Fat Diet. Mol Neurobiol 2024; 61:1479-1494. [PMID: 37726498 PMCID: PMC10896785 DOI: 10.1007/s12035-023-03627-x] [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: 04/11/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
FK506-binding protein 51 kDa (FKBP51), encoded by Fkbp5 gene, gained considerable attention as an important regulator of several aspects of human biology including stress response, metabolic dysfunction, inflammation, and age-dependent neurodegeneration. Its catalytic peptidyl-prolyl isomerase (PPIase) activity is mediated by the N-terminal FK506-binding (FK1) domain, whereas the C-terminal tetratricopeptide motif (TPR) domain is responsible for FKBP51 interaction with molecular chaperone heat shock protein 90 (Hsp90). To understand FKBP51-related biology, several mouse models have been created. These include Fkbp5 complete and conditional knockouts, overexpression, and humanized models. To dissect the role of FKBP51-Hsp90 interaction in FKBP51 biology, we have created an interaction-deficient mouse (Fkbp5TPRmut) by introducing two-point mutations in the TPR domain of FKBP51. FKBP51-Hsp90 interaction-deficient mice are viable, fertile and show Mendelian inheritance. Intracellular association of FKBP51 with Hsp90 is significantly reduced in homozygous mutants compared to wild-type animals. No behavioral differences between genotypes were seen at 2 months of age, however, sex-dependent differences were detected in Y-maze and fear conditioning tests at the age of 12 months. Moreover, we have found a significant reduction in plasma levels of corticosterone and adrenocorticotropic hormone in Fkbp5TPRmut mice after acute stress. In contrast to Fkbp5 knockout mice, females of Fkbp5TPRmut showed increased body weight gain under high-fat diet treatment. Our data confirm the importance of FKBP51-Hsp90 interactions for stress-related endocrine signaling. Also, Fkbp5TPRmut mice can serve as a useful in vivo tool to discriminate between Hsp90-dependent and independent functions of FKBP51.
Collapse
Affiliation(s)
- Lisha Wang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
| | - Jakub Wojcieszak
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
- Department of Pharmacodynamics, Medical University of Lodz, 90151, Lodz, Poland
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), 221005, Varanasi, India
| | - Zhe Zhao
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
- Department of Toxicology, School of Public Health, Peking University, 100191, Beijing, China
| | - Xuelian Sun
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
- National Clinical Research Center for Geriatrics and Department of Gerontology and Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Shaoxun Xie
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 14186, Huddinge, Sweden
| | - Pavel F Pavlov
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden.
| |
Collapse
|
10
|
Noddings CM, Johnson JL, Agard DA. Cryo-EM reveals how Hsp90 and FKBP immunophilins co-regulate the glucocorticoid receptor. Nat Struct Mol Biol 2023; 30:1867-1877. [PMID: 37945740 PMCID: PMC10716051 DOI: 10.1038/s41594-023-01128-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/18/2023] [Indexed: 11/12/2023]
Abstract
Hsp90 is an essential molecular chaperone responsible for the folding and activation of hundreds of 'client' proteins, including the glucocorticoid receptor (GR). Previously, we revealed that Hsp70 and Hsp90 remodel the conformation of GR to regulate ligand binding, aided by co-chaperones. In vivo, the co-chaperones FKBP51 and FKBP52 antagonistically regulate GR activity, but a molecular understanding is lacking. Here we present a 3.01 Å cryogenic electron microscopy structure of the human GR:Hsp90:FKBP52 complex, revealing how FKBP52 integrates into the GR chaperone cycle and directly binds to the active client, potentiating GR activity in vitro and in vivo. We also present a 3.23 Å cryogenic electron microscopy structure of the human GR:Hsp90:FKBP51 complex, revealing how FKBP51 competes with FKBP52 for GR:Hsp90 binding and demonstrating how FKBP51 can act as a potent antagonist to FKBP52. Altogether, we demonstrate how FKBP51 and FKBP52 integrate into the GR chaperone cycle to advance GR to the next stage of maturation.
Collapse
Affiliation(s)
- Chari M Noddings
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Jill L Johnson
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - David A Agard
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
11
|
Cruz B, Vozella V, Carper BA, Xu JC, Kirson D, Hirsch S, Nolen T, Bradley L, Fain K, Crawford M, Kosten TR, Zorrilla EP, Roberto M. FKBP5 inhibitors modulate alcohol drinking and trauma-related behaviors in a model of comorbid post-traumatic stress and alcohol use disorder. Neuropsychopharmacology 2023; 48:1144-1154. [PMID: 36396784 PMCID: PMC10267127 DOI: 10.1038/s41386-022-01497-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/19/2022]
Abstract
Post-traumatic stress disorder (PTSD) leads to enhanced alcohol drinking and development of alcohol use disorder (AUD). Identifying shared neural mechanisms might help discover new therapies for PTSD/AUD. Here, we employed a rat model of comorbid PTSD/AUD to evaluate compounds that inhibit FK506-binding protein 51 (FKBP5), a co-chaperone modulator of glucocorticoid receptors implicated in stress-related disorders. Male and female rats received a familiar avoidance-based shock stress followed by voluntary alcohol drinking. We then assessed trauma-related behaviors through sleep bout cycles, hyperarousal, fear overgeneralization, and irritability. To evaluate the role of stress and alcohol history on the sensitivity to FKBP5 inhibitors, in two separate studies, we administered two FKBP5 inhibitors, benztropine (Study 1) or SAFit2 (Study 2). FKBP5 inhibitors were administered on the last alcohol drinking session and prior to each trauma-related behavioral assessment. We also measured plasma corticosterone to assess the actions of FKBP5 inhibitors after familiar shock stress and alcohol drinking. Benztropine reduced alcohol preference in stressed males and females, while aggressive bouts were reduced in benztropine-treated stressed females. During hyperarousal, benztropine reduced several startle response outcomes across stressed males and females. Corticosterone was reduced in benztropine-treated stressed males. The selective FKBP5 inhibitor, SAFit2, reduced alcohol drinking in stressed males but not females, with no differences in irritability. Importantly, SAFit2 decreased fear overgeneralization in stressed males and females. SAFit2 also reduced corticosterone across stressed males and females. Neither FKBP5 inhibitor changed sleep bout structure. These findings indicate that FKBP5 inhibitors modulate stress-related alcohol drinking and partially modulate trauma-related behaviors. This work supports the hypothesis that targeting FKBP5 may alleviate PTSD/AUD comorbidity.
Collapse
Affiliation(s)
- Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92073, USA
| | - Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92073, USA
| | - Benjamin A Carper
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, 27709, USA
| | - Joy C Xu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92073, USA
| | - Dean Kirson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92073, USA
- Department of Pharmacology, Addiction Science, and Toxicology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Shawn Hirsch
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, 27709, USA
| | - Tracy Nolen
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, 27709, USA
| | - Lauren Bradley
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, 27709, USA
| | - Katie Fain
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, 27709, USA
| | - Meg Crawford
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, 27709, USA
| | - Thomas R Kosten
- Division of Alcohol and Addiction Psychiatry, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Eric P Zorrilla
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92073, USA.
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92073, USA.
| |
Collapse
|
12
|
Wang X, Song J, Yuan Y, Li L, Abu-Taha I, Heijman J, Sun L, Dobrev S, Kamler M, Xie L, Wehrens XH, Horrigan FT, Dobrev D, Li N. Downregulation of FKBP5 Promotes Atrial Arrhythmogenesis. Circ Res 2023; 133:e1-e16. [PMID: 37154033 PMCID: PMC10330339 DOI: 10.1161/circresaha.122.322213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 04/21/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Atrial fibrillation (AF), the most common arrhythmia, is associated with the downregulation of FKBP5 (encoding FKBP5 [FK506 binding protein 5]). However, the function of FKBP5 in the heart remains unknown. Here, we elucidate the consequences of cardiomyocyte-restricted loss of FKBP5 on cardiac function and AF development and study the underlying mechanisms. METHODS Right atrial samples from patients with AF were used to assess the protein levels of FKBP5. A cardiomyocyte-specific FKBP5 knockdown mouse model was established by crossbreeding Fkbp5flox/flox mice with Myh6MerCreMer/+ mice. Cardiac function and AF inducibility were assessed by echocardiography and programmed intracardiac stimulation. Histology, optical mapping, cellular electrophysiology, and biochemistry were employed to elucidate the proarrhythmic mechanisms due to loss of cardiomyocyte FKBP5. RESULTS FKBP5 protein levels were lower in the atrial lysates of patients with paroxysmal AF or long-lasting persistent (chronic) AF. Cardiomyocyte-specific knockdown mice exhibited increased AF inducibility and duration compared with control mice. Enhanced AF susceptibility in cardiomyocyte-specific knockdown mice was associated with the development of action potential alternans and spontaneous Ca2+ waves, and increased protein levels and activity of the NCX1 (Na+/Ca2+-exchanger 1), mimicking the cellular phenotype of chronic AF patients. FKBP5-deficiency enhanced transcription of Slc8a1 (encoding NCX1) via transcription factor hypoxia-inducible factor 1α. In vitro studies revealed that FKBP5 negatively modulated the protein levels of hypoxia-inducible factor 1α by competitively interacting with heat-shock protein 90. Injections of the heat-shock protein 90 inhibitor 17-AAG normalized protein levels of hypoxia-inducible factor 1α and NCX1 and reduced AF susceptibility in cardiomyocyte-specific knockdown mice. Furthermore, the atrial cardiomyocyte-selective knockdown of FKBP5 was sufficient to enhance AF arrhythmogenesis. CONCLUSIONS This is the first study to demonstrate a role for the FKBP5-deficiency in atrial arrhythmogenesis and to establish FKBP5 as a negative regulator of hypoxia-inducible factor 1α in cardiomyocytes. Our results identify a potential molecular mechanism for the proarrhythmic NCX1 upregulation in chronic AF patients.
Collapse
Affiliation(s)
- Xiaolei Wang
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Jia Song
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Yue Yuan
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Luge Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Issam Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Jordi Heijman
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Liang Sun
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Shokoufeh Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Liang Xie
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Xander H.T. Wehrens
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Frank T. Horrigan
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
13
|
Bassil K, Krontira AC, Leroy T, Escoto AIH, Snijders C, Pernia CD, Pasterkamp RJ, de Nijs L, van den Hove D, Kenis G, Boks MP, Vadodaria K, Daskalakis NP, Binder EB, Rutten BPF. In vitro modeling of the neurobiological effects of glucocorticoids: A review. Neurobiol Stress 2023; 23:100530. [PMID: 36891528 PMCID: PMC9986648 DOI: 10.1016/j.ynstr.2023.100530] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Hypothalamic-pituitary adrenal (HPA)axis dysregulation has long been implicated in stress-related disorders such as major depression and post-traumatic stress disorder. Glucocorticoids (GCs) are released from the adrenal glands as a result of HPA-axis activation. The release of GCs is implicated with several neurobiological changes that are associated with negative consequences of chronic stress and the onset and course of psychiatric disorders. Investigating the underlying neurobiological effects of GCs may help to better understand the pathophysiology of stress-related psychiatric disorders. GCs impact a plethora of neuronal processes at the genetic, epigenetic, cellular, and molecular levels. Given the scarcity and difficulty in accessing human brain samples, 2D and 3D in vitro neuronal cultures are becoming increasingly useful in studying GC effects. In this review, we provide an overview of in vitro studies investigating the effects of GCs on key neuronal processes such as proliferation and survival of progenitor cells, neurogenesis, synaptic plasticity, neuronal activity, inflammation, genetic vulnerability, and epigenetic alterations. Finally, we discuss the challenges in the field and offer suggestions for improving the use of in vitro models to investigate GC effects.
Collapse
Affiliation(s)
- Katherine Bassil
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Anthi C Krontira
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Thomas Leroy
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Alana I H Escoto
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Clara Snijders
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Cameron D Pernia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center (UMC) Utrecht Brain Center, Utrecht University, Utrecht, 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, Maastricht, the Netherlands
| | - 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, Maastricht, the Netherlands
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Marco P Boks
- Psychiatry, UMC Utrecht Brain Center, Utrecht, the Netherlands
| | - Krishna Vadodaria
- Salk Institute for Biological Studies, La Jolla, San Diego, United States
| | | | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - 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, Maastricht, the Netherlands
| |
Collapse
|
14
|
Noddings CM, Johnson JL, Agard DA. Cryo-EM reveals how Hsp90 and FKBP immunophilins co-regulate the Glucocorticoid Receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523504. [PMID: 36711821 PMCID: PMC9882067 DOI: 10.1101/2023.01.10.523504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hsp90 is an essential molecular chaperone responsible for the folding and activation of hundreds of 'client' proteins, including the glucocorticoid receptor (GR)1-3. Previously, we revealed that GR ligand binding activity is inhibited by Hsp70 and restored by Hsp90, aided by co-chaperones4. We then presented cryo-EM structures mechanistically detailing how Hsp70 and Hsp90 remodel the conformation of GR to regulate ligand binding5,6. In vivo, GR-chaperone complexes are found associated with numerous Hsp90 co-chaperones, but the most enigmatic have been the immunophilins FKBP51 and FKBP52, which further regulate the activity of GR and other steroid receptors7-9. A molecular understanding of how FKBP51 and FKBP52 integrate with the GR chaperone cycle to differentially regulate GR activation in vivo is lacking due to difficulties reconstituting these interactions. Here, we present a 3.01 Å cryo-EM structure of the GR:Hsp90:FKBP52 complex, revealing , for the first time, that FKBP52 directly binds to the folded, ligand-bound GR using three novel interfaces, each of which we demonstrate are critical for FKBP52-dependent potentiation of GR activity in vivo. In addition, we present a 3.23 Å cryo-EM structure of the GR:Hsp90:FKBP51 complex, which, surprisingly, largely mimics the GR:Hsp90:FKBP52 structure. In both structures, FKBP51 and FKBP52 directly engage the folded GR and unexpectedly facilitate release of p23 through an allosteric mechanism. We also reveal that FKBP52, but not FKBP51, potentiates GR ligand binding in vitro, in a manner dependent on FKBP52-specific interactions. Altogether, we reveal how FKBP51 and FKBP52 integrate into the GR chaperone cycle to advance GR to the next stage of maturation and how FKBP51 and FKBP52 compete for GR:Hsp90 binding, leading to functional antagonism.
Collapse
Affiliation(s)
- Chari M. Noddings
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jill L. Johnson
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| | - David A. Agard
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
15
|
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
|
16
|
Bassil K, De Nijs L, Rutten BPF, Van Den Hove DLA, Kenis G. In vitro modeling of glucocorticoid mechanisms in stress-related mental disorders: Current challenges and future perspectives. Front Cell Dev Biol 2022; 10:1046357. [DOI: 10.3389/fcell.2022.1046357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
In the last decade, in vitro models has been attracting a great deal of attention for the investigation of a number of mechanisms underlying neurological and mental disorders, including stress-related disorders, for which human brain material has rarely been available. Neuronal cultures have been extensively used to investigate the neurobiological effects of stress hormones, in particular glucocorticoids. Despite great advancements in this area, several challenges and limitations of studies attempting to model and investigate stress-related mechanisms in vitro exist. Such experiments often come along with non-standardized definitions stress paradigms in vitro, variations in cell models and cell types investigated, protocols with differing glucocorticoid concentrations and exposure times, and variability in the assessment of glucocorticoid-induced phenotypes, among others. Hence, drawing consensus conclusions from in-vitro stress studies is challenging. Addressing these limitations and aligning methodological aspects will be the first step towards an improved and standardized way of conducting in vitro studies into stress-related disorders, and is indispensable to reach the full potential of in vitro neuronal models. Here, we consider the most important challenges that need to be overcome and provide initial guidelines to achieve improved use of in vitro neuronal models for investigating mechanisms underlying the development of stress-related mental disorders.
Collapse
|
17
|
Rotoli D, Díaz-Flores L, Gutiérrez R, Morales M, Ávila J, Martín-Vasallo P. AmotL2, IQGAP1, and FKBP51 Scaffold Proteins in Glioblastoma Stem Cell Niches. J Histochem Cytochem 2022; 70:9-16. [PMID: 34165350 PMCID: PMC8721575 DOI: 10.1369/00221554211025480] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Glioma stem cells (GSCs) live in a continuous process of stemness reprogramming to achieve specific cell commitment within the so-called GSC niches, specifically located in periarteriolar regions. In this review, we analyze the expression levels, cellular and subcellular location, and role of three scaffold proteins (IQGAP1, FKBP51, and AmotL2) in GSC niches. Scaffold proteins contribute to cell differentiation, migration, and angiogenesis in glioblastoma. It could be of diagnostic interest for establishing stages, for therapeutic targets, and for improving glioblastoma prognosis, which is still at the experimental level.
Collapse
Affiliation(s)
- Deborah Rotoli
- Department of Bioquímica, Microbiología, Biología Celular y Genética, Universidad de La Laguna, San Cristóbal de La Laguna, Spain,Instituto de Tecnología Biomédicas de Canarias, Universidad de La Laguna, San Cristóbal de La Laguna, Spain,Istituto per l’Endocrinologia e l’Oncologia Gaetano Salvatore, Naples, Italy
| | - Lucio Díaz-Flores
- Department of Basic Medical Sciences and Department of Anatomy, Pathology, Histology and Radiology, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Ricardo Gutiérrez
- Department of Basic Medical Sciences and Department of Anatomy, Pathology, Histology and Radiology, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Manuel Morales
- Oncología Médica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Julio Ávila
- Department of Bioquímica, Microbiología, Biología Celular y Genética, Universidad de La Laguna, San Cristóbal de La Laguna, Spain,Instituto de Tecnología Biomédicas de Canarias, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Pablo Martín-Vasallo
- Pablo Martín-Vasallo, UD Bioquímica y Biología Molecular, Universidad de La Laguna, Av/Astrofísico Sánchez s/n, 38206 San Cristóbal de La Laguna, Tenerife, Spain. E-mail:
| |
Collapse
|
18
|
FKBP5 and early life stress affect the hippocampus by an age-dependent mechanism. Brain Behav Immun Health 2021; 9:100143. [PMID: 34589890 PMCID: PMC8474669 DOI: 10.1016/j.bbih.2020.100143] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 01/30/2023] Open
Abstract
Early life stress (ELS) adversely affects the brain and is commonly associated with the etiology of mental health disorders, like depression. In addition to the mood-related symptoms, patients with depression show dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, increased peripheral inflammation, and structural brain alterations. Although the underlying causes are unknown, polymorphisms in the FK506-binding protein 5 (FKBP5) gene, a regulator of glucocorticoid receptor (GR) activity, interact with childhood adversities to increase vulnerability to depressive disorders. We hypothesized that high FKBP5 protein levels combined with early life stress (ELS) would alter the HPA axis and brain, promoting depressive-like behaviors. To test this, we exposed males and females of a mouse model overexpressing FKBP5 in the brain (rTgFKBP5 mice), or littermate controls, to maternal separation for 14 days after birth. Then, we evaluated neuroendocrine, behavioral, and brain changes in young adult and aged mice. We observed lower basal corticosterone (CORT) levels in rTgFKBP5 mice, which was exacerbated in females. Aged, but not young, rTgFKBP5 mice showed increased depressive-like behaviors. Moreover, FKBP5 overexpression reduced hippocampal neuron density in aged mice, while promoting markers of microglia expression, but these effects were reversed by ELS. Together, these results demonstrate that high FKBP5 affects basal CORT levels, depressive-like symptoms, and numbers of neurons and microglia in the hippocampus in an age-dependent manner. High FKBP5 reduces basal corticosterone levels in mice, especially in females. ELS prevents FKBP5-induced susceptibility to depressive-like behavior in aged mice. FKBP5 overexpression reduces hippocampal neuron density in aged mice, while increasing microglial markers.
Collapse
|
19
|
Poletti F, González-Fernández R, García MDP, Rotoli D, Ávila J, Mobasheri A, Martín-Vasallo P. Molecular-Morphological Relationships of the Scaffold Protein FKBP51 and Inflammatory Processes in Knee Osteoarthritis. Cells 2021; 10:2196. [PMID: 34571845 PMCID: PMC8468871 DOI: 10.3390/cells10092196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/03/2021] [Accepted: 08/22/2021] [Indexed: 12/25/2022] Open
Abstract
Knee osteoarthritis (OA) is one of the most prevalent chronic conditions affecting the adult population. OA is no longer thought to come from a purely biomechanical origin but rather one that has been increasingly recognized to include a persistent low-grade inflammatory component. Intra-articular corticosteroid injections (IACSI) have become a widely used method for treating pain in patients with OA as an effective symptomatic treatment. However, as the disease progresses, IACSI become ineffective. FKBP51 is a regulatory protein of the glucocorticoid receptor function and have been shown to be dysregulated in several pathological scenario's including chronic inflammation. Despite of these facts, to our knowledge, there are no previous studies of the expression and possible role of FKBP51 in OA. We investigated by double and triple immunofluorescence confocal microscopy the cellular and subcellular expression of FKBP51 and its relations with inflammation factors in osteoarthritic knee joint tissues: specifically, in the tibial plateau knee cartilage, Hoffa's fat pad and suprapatellar synovial tissue of the knee. Our results show co-expression of FKBP51 with TNF-α, IL-6, CD31 and CD34 in OA chondrocytes, synovial membrane cells and adipocytes in Hoffa's fat pad. FKBP51 is also abundant in nerve fibers within the fat pad. Co-expression of FKBP51 protein with these markers may be indicative of its contribution to inflammatory processes and associated chronic pain in OA.
Collapse
Affiliation(s)
- Fabián Poletti
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, La Laguna, Av. Astrofísico Sánchez s/n, 38206 La Laguna Tenerife, Spain; (F.P.); (R.G.-F.); (D.R.); (J.Á.)
- Orthopaedic Surgery and Trauma Unit, Royal Berkshire Hospital NHS Foundation Trust, Reading RG1 5AN, UK
- Unidad de Cirugía Ortopédica y Traumatología, Hospital San Juan de Dios-Tenerife, Ctra. Santa Cruz Laguna 53, 38009 Santa Cruz de Tenerife, Spain
| | - Rebeca González-Fernández
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, La Laguna, Av. Astrofísico Sánchez s/n, 38206 La Laguna Tenerife, Spain; (F.P.); (R.G.-F.); (D.R.); (J.Á.)
| | - María-del-Pino García
- Department of Pathology, Eurofins® Megalab-Hospiten Hospitals, 38001 Santa Cruz de Tenerife, Spain;
| | - Deborah Rotoli
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, La Laguna, Av. Astrofísico Sánchez s/n, 38206 La Laguna Tenerife, Spain; (F.P.); (R.G.-F.); (D.R.); (J.Á.)
- Institute of Endocrinology and Experimental Oncology (IEOS), CNR-National Research Council, 80131 Naples, Italy
| | - Julio Ávila
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, La Laguna, Av. Astrofísico Sánchez s/n, 38206 La Laguna Tenerife, Spain; (F.P.); (R.G.-F.); (D.R.); (J.Á.)
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, 90570 Oulu, Finland;
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000 Liège, Belgium
| | - Pablo Martín-Vasallo
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, La Laguna, Av. Astrofísico Sánchez s/n, 38206 La Laguna Tenerife, Spain; (F.P.); (R.G.-F.); (D.R.); (J.Á.)
| |
Collapse
|
20
|
Stassen HH, Bachmann S, Bridler R, Cattapan K, Herzig D, Schneeberger A, Seifritz E. Inflammatory processes linked to major depression and schizophrenic disorders and the effects of polypharmacy in psychiatry: evidence from a longitudinal study of 279 patients under therapy. Eur Arch Psychiatry Clin Neurosci 2021; 271:507-520. [PMID: 32696276 PMCID: PMC7981316 DOI: 10.1007/s00406-020-01169-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/13/2020] [Indexed: 12/27/2022]
Abstract
Over the past 2 decades, polypharmacy has become the de-facto standard of acute treatment in psychiatry where patients with psychiatric disorders receive a multiple medication regimen. There is growing evidence for a potential link between major psychiatric disorders and inflammatory processes. Combining these two aspects aims at avoiding polypharmacy attempts among patients with inflammatory activation through alternative treatment strategies. In this study, we addressed the following questions: (1) to what extent can polypharmacy be explained through the factors "diagnosis", "previous history", "severity at baseline", "age", "gender", and "psychiatrist in charge"; (2) what are the differences between polypharmacy and monotherapy regarding efficacy and side effect profiles; and (3) what amount of between-patient variance is explainable by the natural antibody immunoglobulin M (IgM) within each diagnostic group. This naturalistic longitudinal study was comprised of 279 patients under therapy with a clinical diagnosis of depressive (ICD-10: "F3x.x"; n = 195) or schizophrenic disorders (ICD-10: "F2x.x"; n = 84). The study protocol included (1) assessment of previous history by the SADS Syndrome Check List SSCL-16 (lifetime version); (2) repeated measurements over 5 weeks assessing the time course of improvement by the Hamilton Depression Scale HAM-D and the Positive and Negative Syndrome Scale PANSS, along with medication and unwanted side effects through the Medication and Side Effects Inventory MEDIS; and (3) the collection of blood samples from which DNA and serum were extracted. The association between inflammatory response system and psychiatric disorders was detailed by fitting multi-layer Neural Net (NN) models to the observed data ("supervised learning"). The same approach was used to set up prediction models of side effects. Our data showed that polypharmacy was omnipresent. Yet the various polypharmacy regimens had no advantage over monotherapy: we even found slightly larger baseline score reductions under monotherapy, independent of primary diagnoses and for comparable baseline severities. Most patients experienced unwanted side effects. The close link between side effects and treatment regimen was revealed by a linear model in which the mere number of drugs explained a significant (p < 0.001) proportion of the observed variance. As to the inflammatory response system: For the F2 patients, our NN model identified a 22.5% subgroup exhibiting a significant correlation of r = 0.746 (p = 0.0004) between global schizophrenia scores and IgM levels, along with a correct prediction of response of 94.4%, thus explaining 55.7% of the observed between-patient variance. For the F3 patients, our NN model identified a 19.6% subgroup exhibiting a significant correlation of r = 0.644 (p = 0.00003) between global depression scores and IgM levels, along a correct prediction of response of 89.6%, thus explaining 41.4% of the observed between-patient variance. Polypharmacy is omnipresent in today's acute treatment of psychiatric disorders. Given the large proportion of patients with unwanted side effects and the strong correlation between side effects and the number of drugs, polypharmacy approaches are not equally suited for every patient. In terms of efficacy, there are no advantages of polypharmacy over monotherapy. Most notably, our study appears to have cleared the way for the reliable identification of a subgroup of patients for whom the inflammatory response system is a promising target of therapeutic intervention.
Collapse
Affiliation(s)
- H. H. Stassen
- grid.412004.30000 0004 0478 9977Institute for Response-Genetics, Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Psychiatric Hospital, 8032 Zurich, Switzerland
| | - S. Bachmann
- grid.9018.00000 0001 0679 2801Department of Psychiatry, Psychotherapy, and Psychosomatics, University of Halle, 06112 Halle, Germany ,Clienia AG, Psychiatric Hospital, 9573 Littenheid, Switzerland
| | - R. Bridler
- grid.492890.e0000 0004 0627 5312Sanatorium Kilchberg, 8802 Kilchberg, Switzerland
| | - K. Cattapan
- grid.492890.e0000 0004 0627 5312Sanatorium Kilchberg, 8802 Kilchberg, Switzerland
| | - D. Herzig
- grid.412004.30000 0004 0478 9977Institute for Response-Genetics, Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Psychiatric Hospital, 8032 Zurich, Switzerland ,grid.9018.00000 0001 0679 2801Department of Psychiatry, Psychotherapy, and Psychosomatics, University of Halle, 06112 Halle, Germany ,Clienia AG, Psychiatric Hospital, 9573 Littenheid, Switzerland
| | - A. Schneeberger
- grid.412004.30000 0004 0478 9977Institute for Response-Genetics, Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Psychiatric Hospital, 8032 Zurich, Switzerland
| | - E. Seifritz
- grid.412004.30000 0004 0478 9977Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Psychiatric Hospital, 8032 Zurich, Switzerland
| |
Collapse
|
21
|
Paladino A, Woodford MR, Backe SJ, Sager RA, Kancherla P, Daneshvar MA, Chen VZ, Bourboulia D, Ahanin EF, Prodromou C, Bergamaschi G, Strada A, Cretich M, Gori A, Veronesi M, Bandiera T, Vanna R, Bratslavsky G, Serapian SA, Mollapour M, Colombo G. Chemical Perturbation of Oncogenic Protein Folding: from the Prediction of Locally Unstable Structures to the Design of Disruptors of Hsp90-Client Interactions. Chemistry 2020; 26:9459-9465. [PMID: 32167602 PMCID: PMC7415569 DOI: 10.1002/chem.202000615] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Indexed: 12/20/2022]
Abstract
Protein folding quality control in cells requires the activity of a class of proteins known as molecular chaperones. Heat shock protein-90 (Hsp90), a multidomain ATP driven molecular machine, is a prime representative of this family of proteins. Interactions between Hsp90, its co-chaperones, and client proteins have been shown to be important in facilitating the correct folding and activation of clients. Hsp90 levels and functions are elevated in tumor cells. Here, we computationally predict the regions on the native structures of clients c-Abl, c-Src, Cdk4, B-Raf and Glucocorticoid Receptor, that have the highest probability of undergoing local unfolding, despite being ordered in their native structures. Such regions represent potential ideal interaction points with the Hsp90-system. We synthesize mimics spanning these regions and confirm their interaction with partners of the Hsp90 complex (Hsp90, Cdc37 and Aha1) by Nuclear Magnetic Resonance (NMR). Designed mimics selectively disrupt the association of their respective clients with the Hsp90 machinery, leaving unrelated clients unperturbed and causing apoptosis in cancer cells. Overall, selective targeting of Hsp90 protein-protein interactions is achieved without causing indiscriminate degradation of all clients, setting the stage for the development of therapeutics based on specific chaperone:client perturbation.
Collapse
Affiliation(s)
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Priyanka Kancherla
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Michael A Daneshvar
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Victor Z Chen
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Elham F Ahanin
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | | | | | | | | | | | - Marina Veronesi
- D3-PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Tiziano Bandiera
- D3-PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Renzo Vanna
- Institute for Photonics and Nanotechnologies, IFN-CNR, c/o Dept. of Physics, Politecnico di Milano, Piazza L. Da Vinci 32, 20133, Milano, Italy
| | - Gennady Bratslavsky
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Stefano A Serapian
- University of Pavia, Department of Chemistry, Viale Taramelli 10, 27100, Pavia, Italy
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Giorgio Colombo
- SCITEC-CNR, via Mario Bianco 9, 20131, Milano, Italy
- University of Pavia, Department of Chemistry, Viale Taramelli 10, 27100, Pavia, Italy
| |
Collapse
|
22
|
Yuan Y, Zhen L, Li Z, Xu W, Leng H, Xu W, Zheng V, Luria V, Pan J, Tao Y, Zhang H, Cao S, Xu Y. trans-Resveratrol ameliorates anxiety-like behaviors and neuropathic pain in mouse model of post-traumatic stress disorder. J Psychopharmacol 2020; 34:726-736. [PMID: 32308103 DOI: 10.1177/0269881120914221] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND trans-Resveratrol has been extensively investigated for its anti-inflammatory, antioxidant, and anti-psychiatric properties. However, whether it could rescue posttraumatic stress disorder-like stress-induced pain abnormality is unknown. AIM The present study examined the effects of trans-resveratrol on anxiety-like behavior and neuropathic pain induced by single-prolonged stress, which is a classical animal model for mimicking posttraumatic stress disorder. METHODS The single-prolonged stress-induced anxiety-like behavior and pain response were detected by the novelty suppressed feeding, marble burying, locomotor activity, von Frey, and acetone-induced cold allodynia tests in mice. The serum corticosterone levels and glucocorticoid receptor, protein kinase A, phosphorylated cAMP response element binding protein, and brain-derived neurotrophic factor expression were detected by enzyme-linked immunosorbent assay and immunoblot analyses. RESULTS trans-Resveratrol reversed single-prolonged stress-induced increased latency to feed and the number of marbles buried in the novelty suppressed feeding and marble burying tests, but did not significantly influence locomotion distance in the locomotor activity test. trans-Resveratrol also reversed single-prolonged stress-induced cold and mechanical allodynia. Moreover, single-prolonged stress induced abnormality in the limbic hypothalamus-pituitary-adrenal axis was reversed by trans-resveratrol, as evidenced by the fact that trans-resveratrol reversed the differential expression of glucocorticoid receptor in the anxiety- and pain-related regions. In addition, trans-resveratrol increased protein kinase A, phosphorylated cAMP response element binding protein, and brain-derived neurotrophic factor levels, which were decreased in mice subjected to single-prolonged stress. CONCLUSIONS These results provide compelling evidence that trans-resveratrol protects neurons against posttraumatic stress disorder-like stress insults through regulation of limbic hypothalamus-pituitary-adrenal axis function and activation of downstream neuroprotective molecules such as protein kinase A, phosphorylated cAMP response element binding protein, and brain-derived neurotrophic factor expression.
Collapse
Affiliation(s)
- Yirong Yuan
- Department of Neurosurgery, The People's Hospital of Yichun City, Yichun, China
| | - Linlin Zhen
- Department of Breast and Thyroid Surgery, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Zhi Li
- Department of Breast and Thyroid Surgery, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, China.,Department of Pharmaceutical Sciences, The State University of New York, Buffalo, USA
| | - Wenhua Xu
- Department of Orthopedics, The People's Hospital of Yichun City, Yichun, China
| | - Huilin Leng
- Department of Neurology, The People's Hospital of Yichun City, Yichun, China
| | - Wen Xu
- Brain Institute, Wenzhou Medical University, Wenzhou, China
| | - Victor Zheng
- Department of Pharmaceutical Sciences, The State University of New York, Buffalo, USA
| | - Victor Luria
- Department of System Biology, Harvard University Medical School, Boston, USA
| | - Jianchun Pan
- Brain Institute, Wenzhou Medical University, Wenzhou, China
| | - Yuanxiang Tao
- Department of Anesthesiology, The State University of New Jersey, Newark, USA
| | - Hanting Zhang
- Department of Behavioral Medicine and Psychiatry and Physiology, Pharmacology and Neuroscience, West Virginia University Health Sciences Center, Morgantown, USA
| | - Shengsheng Cao
- Department of Orthopedics, The People's Hospital of Yichun City, Yichun, China.,These authors jointly directed this work
| | - Ying Xu
- Department of Pharmaceutical Sciences, The State University of New York, Buffalo, USA.,These authors jointly directed this work
| |
Collapse
|
23
|
Hippocampal Neurogenesis Is Enhanced in Adult Tau Deficient Mice. Cells 2020; 9:cells9010210. [PMID: 31947657 PMCID: PMC7016791 DOI: 10.3390/cells9010210] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 12/22/2022] Open
Abstract
Tau dysfunction is common in several neurodegenerative diseases including Alzheimer’s disease (AD) and frontotemporal dementia (FTD). Affective symptoms have often been associated with aberrant tau pathology and are commonly comorbid in patients with tauopathies, indicating a connection between tau functioning and mechanisms of depression. The current study investigated depression-like behavior in Mapt−/− mice, which contain a targeted deletion of the gene coding for tau. We show that 6-month Mapt−/− mice are resistant to depressive behaviors, as evidenced by decreased immobility time in the forced swim and tail suspension tests, as well as increased escape behavior in a learned helplessness task. Since depression has also been linked to deficient adult neurogenesis, we measured neurogenesis in the hippocampal dentate gyrus and subventricular zone using 5-bromo-2-deoxyuridine (BrdU) labeling. We found that neurogenesis is increased in the dentate gyrus of 14-month-old Mapt−/− brains compared to wild type, providing a potential mechanism for their behavioral phenotypes. In addition to the hippocampus, an upregulation of proteins involved in neurogenesis was observed in the frontal cortex and amygdala of the Mapt−/− mice using proteomic mass spectrometry. All together, these findings suggest that tau may have a role in the depressive symptoms observed in many neurodegenerative diseases and identify tau as a potential molecular target for treating depression.
Collapse
|
24
|
Liberman AC, Budziñski ML, Sokn C, Gobbini RP, Ugo MB, Arzt E. SUMO conjugation as regulator of the glucocorticoid receptor-FKBP51 cellular response to stress. Steroids 2020; 153:108520. [PMID: 31604074 DOI: 10.1016/j.steroids.2019.108520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/20/2019] [Accepted: 10/01/2019] [Indexed: 01/19/2023]
Abstract
In order to adequately respond to stressful stimuli, glucocorticoids (GCs) target almost every tissue of the body. By exerting a negative feedback loop in the hypothalamic-pituitary-adrenal (HPA) axis GCs inhibit their own synthesis and restore homeostasis. GCs actions are mostly mediated by the GC receptor (GR), a member of the nuclear receptor superfamily. Alterations of the GR activity have been associatedto different diseases including mood disorders and can lead to severe complication. Therefore, understanding the molecular complexity of GR modulation is mandatory for the development of new and effective drugs for treating GR-associated disorders. FKBP51 is a GR chaperone that has gained much attention because it is a strong inhibitor of GR activity and has a crucial role in psychiatric diseases. Both GR and FKBP51 activity are regulated by SUMOylation, a posttranslational (PTM). In this review, we focus on the impact of SUMO-conjugation as a regulator of this pathway.
Collapse
Affiliation(s)
- Ana C Liberman
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina.
| | - Maia L Budziñski
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Clara Sokn
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Romina P Gobbini
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Maria B Ugo
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina; Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina.
| |
Collapse
|
25
|
Maguire DG, Ruddock MW, Milanak ME, Moore T, Cobice D, Armour C. Sleep, a Governor of Morbidity in PTSD: A Systematic Review of Biological Markers in PTSD-Related Sleep Disturbances. Nat Sci Sleep 2020; 12:545-562. [PMID: 32801980 PMCID: PMC7402856 DOI: 10.2147/nss.s260734] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Sleep disturbances (SD) are the most impactful and commonly reported symptoms in post-traumatic stress disorder (PTSD). Yet, they are often resistant to primary PTSD therapies. Research has identified two distinct SDs highly prevalent in PTSD; insomnia and nightmares. Those who report SDs prior to a traumatic event are at greater risk for developing PTSD; highlighting that sleep potentially plays a role in PTSD's pathology. To further understand the pathobiological mechanisms that lead to the development of PTSD, it is first imperative to understand the interplay which exists between sleep and PTSD on a biological level. The aim of this systematic review is to determine if biological or physiological markers are related to SD in PTSD. METHODS A systematic literature search was conducted on the electronic databases; Medline, Embase, AMED and PsycINFO, using Medical Subject Headings and associated keywords. RESULTS Sixteen studies were included in the final analyses. Physiological makers of autonomic function, and biochemical markers of HPA-axis activity; inflammatory processes; and trophic factor regulation were related to the severity of SDs in PTSD. CONCLUSION These findings add to the growing literature base supporting a central focus on sleep in research aiming to define the pathophysiological processes which result in PTSD, as well as emphasising the importance of specifically targeting sleep as part of a successful PTSD intervention strategy. Resolving SDs will not only reduce PTSD symptom severity and improve quality of life but will also reduce all-cause mortality, hospital admissions and lifetime healthcare costs for those with PTSD. Limitations of the current literature are discussed, and key recommendations future research must adhere to are made within.
Collapse
Affiliation(s)
- Daniel G Maguire
- Biomedical Sciences Research Institute, Ulster University, Coleraine BT52 1SA, Northern Ireland
| | - Mark W Ruddock
- Randox Laboratories Ltd, Clinical Studies, Crumlin, County Antrim BT29 4QY, Northern Ireland
| | - Melissa E Milanak
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tara Moore
- Biomedical Sciences Research Institute, Ulster University, Coleraine BT52 1SA, Northern Ireland
| | - Diego Cobice
- Biomedical Sciences Research Institute, Ulster University, Coleraine BT52 1SA, Northern Ireland
| | - Cherie Armour
- School of Psychology, David Keir Building, Queen's University Belfast, Belfast BT9 5BN, Northern Ireland
| |
Collapse
|
26
|
Biebl MM, Buchner J. Structure, Function, and Regulation of the Hsp90 Machinery. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a034017. [PMID: 30745292 DOI: 10.1101/cshperspect.a034017] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone involved in the maturation of a plethora of substrates ("clients"), including protein kinases, transcription factors, and E3 ubiquitin ligases, positioning Hsp90 as a central regulator of cellular proteostasis. Hsp90 undergoes large conformational changes during its ATPase cycle. The processing of clients by cytosolic Hsp90 is assisted by a cohort of cochaperones that affect client recruitment, Hsp90 ATPase function or conformational rearrangements in Hsp90. Because of the importance of Hsp90 in regulating central cellular pathways, strategies for the pharmacological inhibition of the Hsp90 machinery in diseases such as cancer and neurodegeneration are being developed. In this review, we summarize recent structural and mechanistic progress in defining the function of organelle-specific and cytosolic Hsp90, including the impact of individual cochaperones on the maturation of specific clients and complexes with clients as well as ways of exploiting Hsp90 as a drug target.
Collapse
Affiliation(s)
- Maximilian M Biebl
- Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, D-85748 Garching, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, D-85748 Garching, Germany
| |
Collapse
|
27
|
Locci A, Pinna G. Social isolation as a promising animal model of PTSD comorbid suicide: neurosteroids and cannabinoids as possible treatment options. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:243-259. [PMID: 30586627 DOI: 10.1016/j.pnpbp.2018.12.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric condition characterized by drastic alterations in mood, emotions, social abilities and cognition. Notably, one aspect of PTSD, particularly in veterans, is its comorbidity with suicide. Elevated aggressiveness predicts high-risk to suicide in humans and despite the difficulty in reproducing a complex human suicidal behavior in rodents, aggressive behavior is a well reproducible behavioral trait of suicide. PTSD animal models are based on a peculiar phenotype, including exaggerated fear memory and impaired fear extinction associated with neurochemical dysregulations in the brain circuitry regulating emotion. The endocannabinoid and the neurosteroid systems regulate emotions and stress responses, and recent evidence shows these two systems are interrelated and critically compromised in neuropsychiatric disorders. For instance, levels of the neurosteroid, allopregnanolone, as well as those of the endocannabinoids, anandamide and its congener, palmitoylethanolamide are decreased in PTSD. Similarly, the endocannabinoid system and neurosteroid biosynthesis are altered in suicidal individuals. Selective serotonin reuptake inhibitors (SSRIs), the only FDA-approved treatments for PTSD, fail to help half of the treatment-seeking patients. This highlights the need for developing biomarker-based efficient therapies. One promising alternative to SSRIs points to stimulation of allopregnanolone biosynthesis as a treatment and a valid end-point to predict treatment response in PTSD patients. This review highlights running findings on the role of the endocannabinoid and neurosteroid systems in PTSD and suicidal behavior both in a preclinical and clinical perspective. A specific focus is given to predictive PTSD/suicide animal models. Ultimately, we discuss the idea that disruption of neurosteroid and endocannabinoid biosynthesis may offer a novel promising biomarker axis to develop new treatments for PTSD and, perhaps, suicidal behavior.
Collapse
Affiliation(s)
- Andrea Locci
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, 1601 W. Taylor St., Chicago, IL 60612, USA
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, 1601 W. Taylor St., Chicago, IL 60612, USA.
| |
Collapse
|
28
|
Zgajnar NR, De Leo SA, Lotufo CM, Erlejman AG, Piwien-Pilipuk G, Galigniana MD. Biological Actions of the Hsp90-binding Immunophilins FKBP51 and FKBP52. Biomolecules 2019; 9:biom9020052. [PMID: 30717249 PMCID: PMC6406450 DOI: 10.3390/biom9020052] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
Immunophilins are a family of proteins whose signature domain is the peptidylprolyl-isomerase domain. High molecular weight immunophilins are characterized by the additional presence of tetratricopeptide-repeats (TPR) through which they bind to the 90-kDa heat-shock protein (Hsp90), and via this chaperone, immunophilins contribute to the regulation of the biological functions of several client-proteins. Among these Hsp90-binding immunophilins, there are two highly homologous members named FKBP51 and FKBP52 (FK506-binding protein of 51-kDa and 52-kDa, respectively) that were first characterized as components of the Hsp90-based heterocomplex associated to steroid receptors. Afterwards, they emerged as likely contributors to a variety of other hormone-dependent diseases, stress-related pathologies, psychiatric disorders, cancer, and other syndromes characterized by misfolded proteins. The differential biological actions of these immunophilins have been assigned to the structurally similar, but functionally divergent enzymatic domain. Nonetheless, they also require the complementary input of the TPR domain, most likely due to their dependence with the association to Hsp90 as a functional unit. FKBP51 and FKBP52 regulate a variety of biological processes such as steroid receptor action, transcriptional activity, protein conformation, protein trafficking, cell differentiation, apoptosis, cancer progression, telomerase activity, cytoskeleton architecture, etc. In this article we discuss the biology of these events and some mechanistic aspects.
Collapse
Affiliation(s)
- Nadia R Zgajnar
- Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires 1428, Argentina.
| | - Sonia A De Leo
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Buenos Aires 1428, Argentina.
| | - Cecilia M Lotufo
- Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires 1428, Argentina.
| | - Alejandra G Erlejman
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Buenos Aires 1428, Argentina.
| | | | - Mario D Galigniana
- Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires 1428, Argentina.
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Buenos Aires 1428, Argentina.
| |
Collapse
|
29
|
Ising M, Maccarrone G, Brückl T, Scheuer S, Hennings J, Holsboer F, Turck CW, Uhr M, Lucae S. FKBP5 Gene Expression Predicts Antidepressant Treatment Outcome in Depression. Int J Mol Sci 2019; 20:ijms20030485. [PMID: 30678080 PMCID: PMC6387218 DOI: 10.3390/ijms20030485] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/13/2019] [Accepted: 01/20/2019] [Indexed: 12/12/2022] Open
Abstract
Adverse experiences and chronic stress are well-known risk factors for the development of major depression, and an impaired stress response regulation is frequently observed in acute depression. Impaired glucocorticoid receptor (GR) signalling plays an important role in these alterations, and a restoration of GR signalling appears to be a prerequisite of successful antidepressant treatment. Variants in genes of the stress response regulation contribute to the vulnerability to depression in traumatized subjects. Consistent findings point to an important role of FKBP5, the gene expressing FK506-binding protein 51 (FKBP51), which is a strong inhibitor of the GR, and thus, an important regulator of the stress response. We investigated the role of FKBP5 and FKB51 expression with respect to stress response regulation and antidepressant treatment outcome in depressed patients. This study included 297 inpatients, who participated in the Munich Antidepressant Response Signature (MARS) project and were treated for acute depression. In this open-label study, patients received antidepressant treatment according to the attending doctor’s choice. In addition to the FKBP5 genotype, changes in blood FKBP51 expression during antidepressant treatment were analyzed using RT-PCR and ZeptoMARKTM reverse phase protein microarray (RPPM). Stress response regulation was evaluated in a subgroup of patients using the combined dexamethasone (dex)/corticotropin releasing hormone (CRH) test. As expected, increased FKBP51 expression was associated with an impaired stress response regulation at baseline and after six weeks was accompanied by an elevated cortisol response to the combined dex/CRH test. Further, we demonstrated an active involvement of FKBP51 in antidepressant treatment outcome. While patients responding to antidepressant treatment had a pronounced reduction of FKBP5 gene and FKBP51 protein expression, increasing expression levels were observed in nonresponders. This effect was moderated by the genotype of the FKBP5 single nucleotide polymorphism (SNP) rs1360780, with carriers of the minor allele showing the most pronounced association. Our findings demonstrate that FKBP5 and, specifically, its expression product FKBP51 are important modulators of antidepressant treatment outcome, pointing to a new, promising target for future antidepressant drug development.
Collapse
Affiliation(s)
- Marcus Ising
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | | | - Tanja Brückl
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | - Sandra Scheuer
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | | | - Florian Holsboer
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
- HMNC Brain Health GmbH, 80807 Munich, Germany.
| | | | - Manfred Uhr
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | - Susanne Lucae
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
30
|
Abstract
The FK506-binding protein 51 (FKBP51) has emerged as a key regulator of endocrine stress responses in mammals and as a potential therapeutic target for stress-related disorders (depression, post-traumatic stress disorder), metabolic disorders (obesity and diabetes) and chronic pain. Recently, FKBP51 has been implicated in several cellular pathways and numerous interacting protein partners have been reported. However, no consensus on the underlying molecular mechanisms has yet emerged. Here, we review the protein interaction partners reported for FKBP51, the proposed pathways involved, their relevance to FKBP51’s physiological function(s), the interplay with other FKBPs, and implications for the development of FKBP51-directed drugs.
Collapse
|
31
|
Thomas N, Gurvich C, Kulkarni J. Borderline personality disorder, trauma, and the hypothalamus-pituitary-adrenal axis. Neuropsychiatr Dis Treat 2019; 15:2601-2612. [PMID: 31564884 PMCID: PMC6743631 DOI: 10.2147/ndt.s198804] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
Borderline personality disorder (BPD) is a complex psychiatric illness for which treatment poses a significant challenge due to limited effective pharmacologic treatments, and under-resourced psychological interventions. BPD is one of the most stigmatized conditions in psychiatry today, but can be understood as a modifiable, neurodevelopmental disorder that arises from maladaptive responses to trauma and stress. Stress susceptibility and reactivity in BPD is thought to mediate both the development and maintenance of BPD symptomatology, with trauma exposure considered an early life risk factor of development, and acute stress moderating symptom trajectory. An altered stress response has been characterized in BPD at the structural, neural, and neurobiological level, and is believed to underlie the maladaptive behavioral and cognitive symptomatology presented in BPD. The endocrine hypothalamus-pituitary-adrenal (HPA) axis represents a key stress response system, and growing evidence suggests it is dysfunctional in the BPD patient population. This theoretical review examines BPD in the context of a neurodevelopmental stress-related disorder, providing an overview of measurements of stress with a focus on HPA-axis measurement. Potential confounding factors associated with measurement of the HPA system are discussed, including sex and sex hormones, genetic factors, and the influence of sample collection methods. HPA-axis dysfunction in BPD largely mirrors findings demonstrated in post-traumatic stress disorder and may represent a valuable neuroendocrine target for diagnostic or treatment response biomarkers, or for which novel treatments can be investigated.
Collapse
Affiliation(s)
- Natalie Thomas
- Central Clinical School , Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Victoria, Australia
| | - Caroline Gurvich
- Central Clinical School , Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Victoria, Australia
| | - Jayashri Kulkarni
- Central Clinical School , Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Baker JD, Ozsan I, Rodriguez Ospina S, Gulick D, Blair LJ. Hsp90 Heterocomplexes Regulate Steroid Hormone Receptors: From Stress Response to Psychiatric Disease. Int J Mol Sci 2018; 20:ijms20010079. [PMID: 30585227 PMCID: PMC6337637 DOI: 10.3390/ijms20010079] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 01/30/2023] Open
Abstract
The hypothalamus-pituitary-adrenal (HPA) axis directly controls the stress response. Dysregulation of this neuroendocrine system is a common feature among psychiatric disorders. Steroid hormone receptors, like glucocorticoid receptor (GR), function as transcription factors of a diverse set of genes upon activation. This activity is regulated by molecular chaperone heterocomplexes. Much is known about the structure and function of these GR/heterocomplexes. There is strong evidence suggesting altered regulation of steroid receptor hormones by chaperones, particularly the 51 kDa FK506-binding protein (FKBP51), may work with environmental factors to increase susceptibility to various psychiatric illnesses including post-traumatic stress disorder (PTSD), major depressive disorder (MDD), and anxiety. This review highlights the regulation of steroid receptor dynamics by the 90kDa heat shock protein (Hsp90)/cochaperone heterocomplexes with an in depth look at how the structural regulation and imbalances in cochaperones can cause functional effects on GR activity. Links between the stress response and circadian systems and the development of novel chaperone-targeting therapeutics are also discussed.
Collapse
Affiliation(s)
- Jeremy D Baker
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Ilayda Ozsan
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Santiago Rodriguez Ospina
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Danielle Gulick
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Laura J Blair
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| |
Collapse
|
33
|
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: 105] [Impact Index Per Article: 15.0] [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
|