1
|
Zhang Y, Yue W, Li J. The association of FKBP5 gene polymorphism with genetic susceptibility to depression and response to antidepressant treatment- a systematic review. BMC Psychiatry 2024; 24:274. [PMID: 38609904 PMCID: PMC11010372 DOI: 10.1186/s12888-024-05717-z] [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: 08/02/2023] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Given the inconsistencies in current studies regarding the impact of FKBP5 gene polymorphisms on depression, arising from variations in study methods, subjects, and treatment strategies, this paper provides a comprehensive review of the relationship between FKBP5 gene polymorphisms and genetic susceptibility to depression, as well as their influence on response to antidepressant treatment. METHODS Electronic databases were searched up to April 11, 2023, for all literature in English and Chinese on depression, FKBP5 gene polymorphisms, and antidepressant treatment. Data extraction and quality assessment were performed for key study characteristics. Qualitative methods were used to synthesize the study results. RESULTS A total of 21 studies were included, with the majority exhibiting average to moderate quality. Six SNPs (rs3800373, rs1360780, rs9470080, rs4713916, rs9296158, rs9394309) were broadly implicated in susceptibility to depression, while rs1360780 and rs3800373 were linked to antidepressant treatment sensitivity. Additionally, rs1360780 was associated with adverse reactions to antidepressant drug treatment. However, these associations were largely unconfirmed in replication studies. CONCLUSIONS Depression is recognized as a polygenic genetic disorder, with multiple genes contributing, each exerting relatively small effects. Future studies should explore not only multiple gene interactions but also epigenetic changes. Presently, research on FKBP5 in affective disorders remains notably limited, highlighting the necessity for further investigations in this domain.
Collapse
Affiliation(s)
- Ying Zhang
- Institute of Mental Health, Peking University Sixth Hospital, 100191, Beijing, China
- Tianjin Anding Hospital, Tianjin Municipal Mental Health Center, 300222, Tianjin, China
| | - Weihua Yue
- Institute of Mental Health, Peking University Sixth Hospital, 100191, Beijing, China.
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), 100191, Beijing, China.
- NHC Key Laboratory of Mental Health, Peking University, 100191, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- Chinese Institute for Brain Research, 102206, Beijing, China.
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 300222, Tianjin, China.
| |
Collapse
|
2
|
Malekpour M, Shekouh D, Safavinia ME, Shiralipour S, Jalouli M, Mortezanejad S, Azarpira N, Ebrahimi ND. Role of FKBP5 and its genetic mutations in stress-induced psychiatric disorders: an opportunity for drug discovery. Front Psychiatry 2023; 14:1182345. [PMID: 37398599 PMCID: PMC10313426 DOI: 10.3389/fpsyt.2023.1182345] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/24/2023] [Indexed: 07/04/2023] Open
Abstract
Stress-induced mental health disorders are affecting many people around the world. However, effective drug therapy for curing psychiatric diseases does not occur sufficiently. Many neurotransmitters, hormones, and mechanisms are essential in regulating the body's stress response. One of the most critical components of the stress response system is the hypothalamus-pituitary-adrenal (HPA) axis. The FKBP prolyl isomerase 51 (FKBP51) protein is one of the main negative regulators of the HPA axis. FKBP51 negatively regulates the cortisol effects (the end product of the HPA axis) by inhibiting the interaction between glucocorticoid receptors (GRs) and cortisol, causing reduced transcription of downstream cortisol molecules. By regulating cortisol effects, the FKBP51 protein can indirectly regulate the sensitivity of the HPA axis to stressors. Previous studies have indicated the influence of FKBP5 gene mutations and epigenetic changes in different psychiatric diseases and drug responses and recommended the FKBP51 protein as a drug target and a biomarker for psychological disorders. In this review, we attempted to discuss the effects of the FKBP5 gene, its mutations on different psychiatric diseases, and drugs affecting the FKBP5 gene.
Collapse
Affiliation(s)
- Mahdi Malekpour
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Dorsa Shekouh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Shadi Shiralipour
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Jalouli
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Mortezanejad
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
3
|
Yang Y, Babayan L, Mirzakhanian A, Sisliyan N, Zhang D, Hurtado C, Zahid A, Bergsneider M, Kim W, Wang MB, Heaney AP. Circulating FK506 binding protein 51 mRNA expression in patients with pituitary adenomas. Heliyon 2023; 9:e12678. [PMID: 36699264 PMCID: PMC9868367 DOI: 10.1016/j.heliyon.2022.e12678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/03/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Background FK506 binding protein 51 (FKBP5) is a co-chaperone regulator of the glucocorticoid receptor (GR). Recent studies have reported increased FKBP5 mRNA in the circulation from patients with Cushing disease (CD) which returned to comparable levels seen in healthy controls following successful trans-nasal trans-sphenoidal (TNTS) surgical corticotroph tumor removal. However, the expression of circulating FKBP5 mRNA levels in other pituitary tumor subtypes and its specificity to corticotroph tumors is unknown. Methods Pre-operative blood was collected from consecutive patients undergoing TNTS for pituitary tumors (n = 57) at our center between 2015 and 2019. Total RNA was isolated from whole blood using RiboPure blood RNA isolation kit and real-time qPCR was used to quantitate circulating FKBP5 mRNA expression. Results Consistent with the prior report, higher circulating FKBP5 mRNA levels were observed in 20 patients with CD prior to surgical tumor removal, compared to 21 healthy controls (p < 0.0005) and compared to 8 patients harboring gonadotroph pituitary tumors (p < 0.05) and 6 patients with silent corticotroph pituitary tumors (p < 0.05). However, circulating FKBP5 mRNA levels were higher in 10 patients with prolactin (PRL)-secreting pituitary tumors compared to healthy controls (p < 0.05), and did not differ between patients with CD and patients with growth hormone secreting tumors (GH-omas). Conclusions Although we confirm that circulating FKBP5 mRNA is higher in patients with corticotroph tumors compared to healthy subjects, measurement of circulating FKBP5 does not appear to be helpful to distinguish corticotroph tumors from other pituitary tumor sub-types.
Collapse
Affiliation(s)
- Yingying Yang
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lilit Babayan
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Argishty Mirzakhanian
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Nvard Sisliyan
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Dongyun Zhang
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Carolina Hurtado
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Abdul Zahid
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Marvin Bergsneider
- Departments of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Won Kim
- Departments of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Marilene B. Wang
- Departments of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Anthony P. Heaney
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA,Departments of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA,Corresponding author. Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Zheng Y, Zhang L, He S, Xie Z, Zhang J, Ge C, Sun G, Huang J, Li H. Integrated Module of Multidimensional Omics for Peripheral Biomarkers (iMORE) in patients with major depressive disorder: rationale and design of a prospective multicentre cohort study. BMJ Open 2022; 12:e067447. [PMID: 36418119 PMCID: PMC9685190 DOI: 10.1136/bmjopen-2022-067447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Major depressive disorder (MDD) represents a worldwide burden on healthcare and the response to antidepressants remains limited. Systems biology approaches have been used to explore the precision therapy. However, no reliable biomarker clinically exists for prognostic prediction at present. The objectives of the Integrated Module of Multidimensional Omics for Peripheral Biomarkers (iMORE) study are to predict the efficacy of antidepressants by integrating multidimensional omics and performing validation in a real-world setting. As secondary aims, a series of potential biomarkers are explored for biological subtypes. METHODS AND ANALYSIS iMore is an observational cohort study in patients with MDD with a multistage design in China. The study is performed by three mental health centres comprising an observation phase and a validation phase. A total of 200 patients with MDD and 100 healthy controls were enrolled. The protocol-specified antidepressants are selective serotonin reuptake inhibitors and serotonin-norepinephrine reuptake inhibitors. Clinical visits (baseline, 4 and 8 weeks) include psychiatric rating scales for symptom assessment and biospecimen collection for multiomics analysis. Participants are divided into responders and non-responders based on treatment response (>50% reduction in Montgomery-Asberg Depression Rating Scale). Antidepressants' responses are predicted and biomarkers are explored using supervised learning approach by integration of metabolites, cytokines, gut microbiomes and immunophenotypic cells. The accuracy of the prediction models constructed is verified in an independent validation phase. ETHICS AND DISSEMINATION The study was approved by the ethics committee of Shanghai Mental Health Center (approval number 2020-87). All participants need to sign a written consent for the study entry. Study findings will be published in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT04518592.
Collapse
Affiliation(s)
- Yuzhen Zheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linna Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shen He
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zuoquan Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jing Zhang
- Shanghai Green Valley Pharmaceutical Co Ltd, Shanghai, China
| | - Changrong Ge
- Shanghai Green Valley Pharmaceutical Co Ltd, Shanghai, China
| | - Guangqiang Sun
- Shanghai Green Valley Pharmaceutical Co Ltd, Shanghai, China
| | - Jingjing Huang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Research Center for Mental Health, Shanghai Mental Health Center, Shanghai, China
| | - Huafang Li
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Research Center for Mental Health, Shanghai Mental Health Center, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Galkin S, Ivanova S, Bokhan N. Current methods for predicting therapeutic response in patients with depressive disorders. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:15-21. [DOI: 10.17116/jnevro202212202115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
6
|
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
|
7
|
Kraus C, Kadriu B, Lanzenberger R, Zarate CA, Kasper S. Prognosis and Improved Outcomes in Major Depression: A Review. FOCUS: JOURNAL OF LIFE LONG LEARNING IN PSYCHIATRY 2020; 18:220-235. [PMID: 33343240 DOI: 10.1176/appi.focus.18205] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
(Reprinted from Transl Psychiatry. 2019 Apr 3; 9(1):127. Open access; is licensed under a Creative Commons Attribution 4.0 International License).
Collapse
|
8
|
Palumbo MC, Dominguez S, Dong H. Sex differences in hypothalamic-pituitary-adrenal axis regulation after chronic unpredictable stress. Brain Behav 2020; 10:e01586. [PMID: 32154650 PMCID: PMC7177572 DOI: 10.1002/brb3.1586] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/06/2020] [Accepted: 02/10/2020] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION Exposure to stress, mediated through the hypothalamic-pituitary-adrenal (HPA) axis, elicits sex differences in endocrine, neurological, and behavioral responses. However, the sex-specific factors that confer resilience or vulnerability to stress and stress-associated psychiatric disorders remain largely unknown. The evident sex differences in stress-related disease prevalence suggest the underlying differences in the neurobiological underpinnings of HPA axis regulation. METHOD Here, we used a chronic unpredictable stress (CUS) model to investigate the behavioral and biochemical responses of the HPA axis in C57BL/6 mice. Animals were tested in the open field and forced swim test to examine anxiety-like and depressive-like behaviors. Plasma corticosterone levels were measured after behavior and CUS, and glucocorticoid receptor (GR) expression and cytosolic and nuclear fractions of binding protein FKBP51 expression were taken to measure function and regulation of the stress response. RESULTS Our results indicate increased depressive-like behavior in males and females which correlated with increased corticosterone levels following CUS. However, females displayed more anxiety-like behaviors with and without CUS. Interestingly, we found trends toward dysregulation of GR protein expression in CUS females, and an increase in the GR inhibitory protein, FKBP51, in the cytosol of CUS males but not females. CONCLUSION These results suggest biochemical alterations to the HPA axis regulation which may elicit a glucocorticoid resistance in females after chronic stress and may contribute to the sex-biased vulnerability to stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michelle C Palumbo
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sky Dominguez
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
9
|
Calabrò M, Porcelli S, Crisafulli C, Albani D, Kasper S, Zohar J, Souery D, Montgomery S, Mantovani V, Mendlewicz J, Bonassi S, Vieta E, Frustaci A, Ducci G, Landi S, Boccia S, Bellomo A, Di Nicola M, Janiri L, Colombo R, Benedetti F, Mandelli L, Fabbri C, Serretti A. Genetic variants associated with psychotic symptoms across psychiatric disorders. Neurosci Lett 2020; 720:134754. [PMID: 31945448 DOI: 10.1016/j.neulet.2020.134754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/06/2019] [Accepted: 01/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent evidence suggests that psychiatric symptoms share a common genetic liability across diagnostic categories. The present study investigated the effects of variants within previously identified relevant genes on specific symptom clusters, independently from the diagnosis. METHODS 1550 subjects affected by Schizophrenia (SCZ), Major Depressive Disorder or Bipolar Disorder were included. Symptoms were assessed using the Positive and Negative Syndrome Scale (PANSS) and the Hamilton Depression Rating Scale (HDRS). Principal component analysis and a further clinical refinement were used to define symptom clusters. Clusters scores were tested for association with 46 genetic variants within nine genes previously linked to one or more major psychiatric disorders by large genome wide association studies (ANK3, CACNA1C, CACNB2, FKBP5, FZD3, GRM7, ITIH3, SYNE1, TCF4). Exploratory analyses were performed in each disorder separately to further elucidate the SNPs effects. RESULTS five PANSS clusters (Negative; Impulsiveness; Cognitive; Psychotic; Depressive) and four HDRS clusters (Core Depressive; Somatic; Psychotic-like; Insomnia) were identified. CACNA1C rs11615998 was associated with HDRS Psychotic cluster in the whole sample. In the SCZ sample, CACNA1C rs11062296 was associated with PANSS Impulsiveness cluster and CACNA1C rs2238062 was associated with PANSS negative cluster. DISCUSSION CACNA1C rs11615998 was associated with psychotic symptoms (C-allele carriers have decreased psychotic-risk) independently from the diagnosis, in line with the evidence of a cross disorder effect of many risk variants. This gene was previously associated with SCZ and cross-disorder liability to psychiatric disorders. Our findings confirmed that deep phenotyping is pivotal to clarify the role of genetic variants on symptoms patterns.
Collapse
Affiliation(s)
- Marco Calabrò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| | - Stefano Porcelli
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy
| | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| | - Diego Albani
- Laboratory of Biology of Neurodegenerative Disorders, Neuroscience Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Austria
| | - Joseph Zohar
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, and Sackler School of Medicine, Tel Aviv University, Israel
| | - Daniel Souery
- Laboratoire de Psychologie Medicale, Universitè Libre de Bruxelles and Psy Pluriel, Centre Européen de Psychologie Medicale, Brussels, Belgium
| | | | - Vilma Mantovani
- Center for Applied Biomedical Research (CRBA), St. Orsola University Hospital, Bologna, Italy
| | | | - Stefano Bonassi
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Pisana, Rome, Italy; Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | - Eduard Vieta
- Bipolar Disorders Unit, Institute of Neuroscience, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Alessandra Frustaci
- Barnet, Enfield and Haringey Mental Health NHS Trust, St.Ann's Hospital, St.Ann's Road, N15 3 TH, London, UK
| | | | - Stefano Landi
- Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Stefania Boccia
- Sezione di Igiene, Istituto di Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italy; Department of Woman and Child Health and Public Health - Public Health Area, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Antonello Bellomo
- Dipartimento di Medicina Clinica e Sperimentale, Foggia University, Italy
| | - Marco Di Nicola
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigi Janiri
- Faculty of Medicine "Agostino Gemelli", Catholic University of the Sacred Heart, Rome, Italy
| | - Roberto Colombo
- Division of Neuroscience, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Francesco Benedetti
- Faculty of Medicine "Agostino Gemelli", Catholic University of the Sacred Heart, Rome, Italy
| | - Laura Mandelli
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy
| | - Chiara Fabbri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy
| | - Alessandro Serretti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy.
| |
Collapse
|
10
|
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: 201] [Impact Index Per Article: 33.5] [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
|
11
|
Corponi F, Fabbri C, Serretti A. Pharmacogenetics and Depression: A Critical Perspective. Psychiatry Investig 2019; 16:645-653. [PMID: 31455064 PMCID: PMC6761796 DOI: 10.30773/pi.2019.06.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2019] [Accepted: 06/16/2019] [Indexed: 12/17/2022] Open
Abstract
Depression leads the higher personal and socio-economical burden within psychiatric disorders. Despite the fact that over 40 antidepressants (ADs) are available, suboptimal response still poses a major challenge and is thought to be partially a result of genetic variation. Pharmacogenetics studies the effects of genetic variants on treatment outcomes with the aim of providing tailored treatments, thereby maximizing efficacy and tolerability. After two decades of pharmacogenetic research, variants in genes coding for the cytochromes involved in ADs metabolism (CYP2D6 and CYP2C19) are now considered biomarkers with sufficient scientific support for clinical application, despite the lack of conclusive cost/effectiveness evidence. The effect of variants in genes modulating ADs mechanisms of action (pharmacodynamics) is still controversial, because of the much higher complexity of ADs pharmacodynamics compared to ADs metabolism. Considerable progress has been made since the era of candidate gene studies: the genomic revolution has made possible to assess genetic variance on an unprecedented scale, throughout the whole genome, and to analyze the cumulative effect of different variants. The results have revealed key information on the biological mechanisms mediating ADs effect and identified hypothetical new pharmacological targets. They also paved the way for future availability of polygenic pharmacogenetic panels to predict treatment outcome, which are expected to explain much higher variance in ADs response compared to CYP2D6 and CYP2C19 only. As the demand and availability of AD pharmacogenetic testing is projected to increase, it is important for clinicians to keep abreast of this evolving area to facilitate informed discussions with their patients.
Collapse
Affiliation(s)
- Filippo Corponi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Fabbri
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, United Kingdom
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Abstract
The FKBP5 protein is of importance for the function of the glucocorticoid receptor. The purpose of the present study was to examine the possible association between the different genotypes of rs1360780 in the FKBP5 gene, and clinical symptoms in patients with unipolar depression. Seven hundred eighteen patients and 673 controls from the Danish Psychiatric Biobank were participated. No association was found between any genotype and diagnosis of unipolar depression. It was found that the group of depressed patients with the CC genotype showed significantly earlier start of treatment with medicine, had a significantly greater tendency to be treated with electroconvulsive therapy and showed a significantly higher frequency of family history of depression compared with the combined group of patients with the CT and TT genotypes. The only informations about controls were their sex and that they were recruited from the blood bank. The clinical data were not complete for all patients.
Collapse
|
13
|
Müller DJ, Brandl EJ, Degenhardt F, Domschke K, Grabe H, Gruber O, Hebebrand J, Maier W, Menke A, Riemenschneider M, Rietschel M, Rujescu D, Schulze TG, Tebartz van Elst L, Tüscher O, Deckert J. [Pharmacogenetics in psychiatry: state of the art]. DER NERVENARZT 2019; 89:290-299. [PMID: 29383410 DOI: 10.1007/s00115-017-0479-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this article, the current literature on pharmacogenetics of antidepressants, antipsychotics and lithium are summarized by the section of Neurobiology and Genetics of the German Society of Psychiatry, Psychotherapy and Neurology (DGPPN). The publications of international expert groups and regulatory authorities are reviewed and discussed. In Germany, a statement on pharmacogenetics was also made by the gene diagnostics committee of the Ministry of Health. The DGPPN supports two recommendations: 1) to perform CYP2D6 genetic testing prior to prescription of tricyclic antidepressants and 2) to determine the HLA-B*1502 genotype in patients of Asian origin before using carbamazepine. The main obstacle for a broad application of pharmacogenetic tests in psychiatry remains the lack of large prospective studies, for both single gene-drug pair and cobinatorial pharmacogenetic tests, to evaluate the benefits of genetic testing. Psychiatrists, geneticists and funding agencies are encouraged to increase their efforts for the future benefit of psychiatric patients.
Collapse
Affiliation(s)
- D J Müller
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., R132, Toronto, ON, M5T 1R8, Kanada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Kanada.
| | - E J Brandl
- Klinik für Psychiatrie und Psychotherapie, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Deutschland.,Berlin Institute of Health, Berlin, Deutschland
| | - F Degenhardt
- Institut für Humangenetik, Universitätsklinikum Bonn, Bonn, Deutschland
| | - K Domschke
- Klinik für Psychiatrie und Psychotherapie, Universität Freiburg, Freiburg, Deutschland
| | - H Grabe
- Klinik und Poliklinik für Psychiatrie und Psychotherapie an der Universitätsmedizin Greifswald, Universität Greifswald, Greifswald, Deutschland
| | - O Gruber
- Klinik für Allgemeine Psychiatrie, Zentrum für Psychosoziale Medizin, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - J Hebebrand
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie des Kindes- und Jugendalters, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Deutschland
| | - W Maier
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Bonn, Bonn, Deutschland
| | - A Menke
- Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie, Zentrum für Psychische Gesundheit, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - M Riemenschneider
- Klinik für Psychiatrie, Universitätsklinikum des Saarlandes, Homburg/Saar, Deutschland
| | - M Rietschel
- Zentralinstitut für Seelische Gesundheit, Mannheim, Deutschland
| | - D Rujescu
- Klinik und Poliklinik für Psychiatrie, Psychotherapie und Psychosomatik, Martin-Luther-Universität Halle-Wittenberg, Halle, Deutschland
| | - T G Schulze
- Institut für Psychiatrische Phänomik und Genomik (IPPG), Klinikum der Universität München, LMU München, München, Deutschland
| | - L Tebartz van Elst
- Klinik für Psychiatrie und Psychotherapie, Universität Freiburg, Freiburg, Deutschland
| | - O Tüscher
- Klinik für Psychiatrie und Psychotherapie, Universitätsmedizin der Johannes-Gutenberg Universität, Mainz, Deutschland
| | - J Deckert
- Klinik und Poliklinik für Psychiatrie, Psychosomatik und Psychotherapie, Zentrum für Psychische Gesundheit, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | | |
Collapse
|
14
|
Kraus C, Kadriu B, Lanzenberger R, Zarate CA, Kasper S. Prognosis and improved outcomes in major depression: a review. Transl Psychiatry 2019; 9:127. [PMID: 30944309 PMCID: PMC6447556 DOI: 10.1038/s41398-019-0460-3] [Citation(s) in RCA: 257] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/10/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023] Open
Abstract
Treatment outcomes for major depressive disorder (MDD) need to be improved. Presently, no clinically relevant tools have been established for stratifying subgroups or predicting outcomes. This literature review sought to investigate factors closely linked to outcome and summarize existing and novel strategies for improvement. The results show that early recognition and treatment are crucial, as duration of untreated depression correlates with worse outcomes. Early improvement is associated with response and remission, while comorbidities prolong course of illness. Potential biomarkers have been explored, including hippocampal volumes, neuronal activity of the anterior cingulate cortex, and levels of brain-derived neurotrophic factor (BDNF) and central and peripheral inflammatory markers (e.g., translocator protein (TSPO), interleukin-6 (IL-6), C-reactive protein (CRP), tumor necrosis factor alpha (TNFα)). However, their integration into routine clinical care has not yet been fully elucidated, and more research is needed in this regard. Genetic findings suggest that testing for CYP450 isoenzyme activity may improve treatment outcomes. Strategies such as managing risk factors, improving clinical trial methodology, and designing structured step-by-step treatments are also beneficial. Finally, drawing on existing guidelines, we outline a sequential treatment optimization paradigm for selecting first-, second-, and third-line treatments for acute and chronically ill patients. Well-established treatments such as electroconvulsive therapy (ECT) are clinically relevant for treatment-resistant populations, and novel transcranial stimulation methods such as theta-burst stimulation (TBS) and magnetic seizure therapy (MST) have shown promising results. Novel rapid-acting antidepressants, such as ketamine, may also constitute a paradigm shift in treatment optimization for MDD.
Collapse
Affiliation(s)
- Christoph Kraus
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Carlos A Zarate
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
15
|
Fabbri C, Zohar J, Serretti A. Pharmacogenetic tests to guide drug treatment in depression: Comparison of the available testing kits and clinical trials. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:36-44. [PMID: 29777729 DOI: 10.1016/j.pnpbp.2018.05.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 12/26/2022]
Abstract
The empirical approach to drug choice and dosing in depression often results into inadequate response and side effects. Pharmacogenetic (PGx) testing appears a promising way to implement personalized treatments. A systematic review was performed to identify available PGx tests, compare the genes they include with clinical guidelines and drug labels, and assess the quality of published clinical studies. ~40 commercial PGx tests are available and potential benefits were estimated for nine of them by clinical studies. The most part of studies are observational (9/21) or non-randomized case-control trials that compared standard care with PGx-guided treatment (6/21), six randomized controlled trials (RCTs) are available. The only genes included in all the available PGx tests and with recommendations in current clinical guidelines and drug labels are CYP2D6 and CYP2C19. There is heterogeneity among outcome measures across studies (response, remission, improvement, health care utilization, medication tolerability), as well as in trial design. Relatively weak clinical benefits were reported by RCTs and higher clinical benefits by non-RCTs, but the last group showed greater risk of bias. Lack of patient and rater's blindness, retrospective design and possible confounders (concomitant medications and medical diseases, lack of wash out prior to inclusion, no assessment of compliance etc.) were the main issues. Estimations of cost savings provided heterogeneous findings. Variants in CYP2D6 and CYP2C19 have already adequate support for clinical application. The development of future PGx tests should include best practices for clinical evidence development and for health economic assessment.
Collapse
Affiliation(s)
- Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Joseph Zohar
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, and Sackler School of Medicine, Tel Aviv University, Israel
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
16
|
Criado-Marrero M, Rein T, Binder EB, Porter JT, Koren J, Blair LJ. Hsp90 and FKBP51: complex regulators of psychiatric diseases. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0532. [PMID: 29203717 DOI: 10.1098/rstb.2016.0532] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 09/11/2017] [Indexed: 01/30/2023] Open
Abstract
Mood disorders affect nearly a quarter of the world's population. Therefore, understanding the molecular mechanisms underlying these conditions is of great importance. FK-506 binding protein 5 (FKBP5) encodes the FKBP51 protein, a heat shock protein 90 kDa (Hsp90) co-chaperone, and is a risk factor for several affective disorders. FKBP51, in coordination with Hsp90, regulates glucocorticoid receptor (GR) activity via a short negative feedback loop. This signalling pathway rapidly restores homeostasis in the hypothalamic-pituitary-adrenal (HPA) axis following stress. Expression of FKBP5 increases with age through reduced DNA methylation. High levels of FKBP51 are linked to GR resistance and reduced stress coping behaviour. Moreover, common allelic variants in the FKBP5 gene are associated with increased risk of developing affective disorders like anxiety, depression and post-traumatic stress disorder (PTSD). This review highlights the current understanding of the Hsp90 co-chaperone, FKBP5, in disease from both human and animal studies. In addition, FKBP5 genetic implications in the clinic involving life stress exposure, gender differences and treatment outcomes are discussed.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Marangelie Criado-Marrero
- Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Theo Rein
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - James T Porter
- Department of Basic Sciences, Ponce Health Sciences University-School of Medicine/Ponce Research Institute, Ponce, Puerto Rico 00732, USA
| | - John Koren
- Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Laura J Blair
- Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| |
Collapse
|
17
|
Abstract
Mental illness represents a major health issue both at the individual and at the socioeconomical level. This is partly due to the current suboptimal treatment options: existing psychotropic medications, including antidepressants, antipsychotics, and mood stabilizers, are effective only in a subset of patients or produce partial response and they are often associated with debilitating side effects that discourage adherence. Pharmacogenetics is the study of how genetic information impacts on drug response/side effects with the goal to provide tailored treatments, thereby maximizing efficacy and tolerability. The first pharmacogenetic studies focused on candidate genes, previously known to be relevant to the pharmacokinetics and pharmacodynamics of psychotropic drugs. Results were mainly inconclusive, but some replicated candidates were identified and included as pharmacogenetic biomarkers in drug labeling and in some commercial kits. With the advent of the genomic revolution, it became possible to study the genetic variation on an unprecedented scale, throughout the whole genome with no need of a priori hypothesis. This may lead to the personalized prescription of existing medications and potentially to the development of innovative ones, thanks to new insights into the genetics of mental illness. Promising findings were obtained, but methods for the generation and analysis of genome-wide and sequencing data are still in evolution. Future pharmacogenetic tests may consist of hundreds/thousands of polymorphisms throughout the genome or selected pathways in order to take into account the complex interactions across variants in a number of genes.
Collapse
Affiliation(s)
- Filippo Corponi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
18
|
DNA methylation and clinical response to antidepressant medication in major depressive disorder: A review and recommendations. Neurosci Lett 2018; 669:14-23. [DOI: 10.1016/j.neulet.2016.12.071] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 12/28/2022]
|
19
|
Fabbri C, Corponi F, Albani D, Raimondi I, Forloni G, Schruers K, Kasper S, Kautzky A, Zohar J, Souery D, Montgomery S, Cristalli CP, Mantovani V, Mendlewicz J, Serretti A. Pleiotropic genes in psychiatry: Calcium channels and the stress-related FKBP5 gene in antidepressant resistance. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:203-210. [PMID: 28989100 DOI: 10.1016/j.pnpbp.2017.10.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 11/27/2022]
Abstract
A candidate gene and a genome-wide approach were combined to study the pharmacogenetics of antidepressant response and resistance. Investigated genes were selected on the basis of pleiotropic effect across psychiatric phenotypes in previous genome-wide association studies and involvement in antidepressant response. Three samples with major depressive disorder (total=671) were genotyped for 44 SNPs in 8 candidate genes (CACNA1C, CACNB2, ANK3, GRM7, TCF4, ITIH3, SYNE1, FKBP5). Phenotypes were response/remission after 4weeks of treatment and treatment-resistant depression (TRD). Genome-wide data from STAR*D were used to replicate findings for response/remission (n=1409) and TRD (n=620). Pathways including the most promising candidate genes were investigated in STAR*D for involvement in TRD. FKBP5 polymorphisms showed replicated but nominal associations with response, remission or TRD. CACNA1C rs1006737 and rs10848635 were the only polymorphisms that survived multiple-testing correction. In STAR*D the best pathway associated with TRD included CACNA1C (GO:0006942, permutated p=0.15). Machine learning models showed that independent SNPs in this pathway predicted TRD with a mean sensitivity of 0.83 and specificity of 0.56 after 10-fold cross validation repeated 100 times. FKBP5 polymorphisms appear good candidates for inclusion in antidepressant pharmacogenetic tests. Pathways including the CACNA1C gene may be involved in TRD and they may provide the base for developing multi-marker predictors of TRD.
Collapse
Affiliation(s)
- Chiara Fabbri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy
| | - Filippo Corponi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy
| | - Diego Albani
- Laboratory of Biology of Neurodegenerative Disorders, Neuroscience Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Ilaria Raimondi
- Laboratory of Biology of Neurodegenerative Disorders, Neuroscience Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Gianluigi Forloni
- Laboratory of Biology of Neurodegenerative Disorders, Neuroscience Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Koen Schruers
- School of Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Austria
| | - Alexander Kautzky
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Austria
| | - Joseph Zohar
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Israel
| | - Daniel Souery
- Laboratoire de Psychologie Medicale, Universitè Libre de Bruxelles and Psy Pluriel, Centre Européen de Psychologie Medicale, Brussels, Belgium
| | | | - Carlotta Pia Cristalli
- Center for Applied Biomedical Research (CRBA), St. Orsola University Hospital, Bologna, Italy
| | - Vilma Mantovani
- Center for Applied Biomedical Research (CRBA), St. Orsola University Hospital, Bologna, Italy
| | - Julien Mendlewicz
- Universite´ Libre de Bruxelles, Avenue Franklin Roosevelt 50, Bruxelles, Belgium
| | - Alessandro Serretti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy.
| |
Collapse
|
20
|
Fries GR, Gassen NC, Rein T. The FKBP51 Glucocorticoid Receptor Co-Chaperone: Regulation, Function, and Implications in Health and Disease. Int J Mol Sci 2017; 18:ijms18122614. [PMID: 29206196 PMCID: PMC5751217 DOI: 10.3390/ijms18122614] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/27/2022] Open
Abstract
Among the chaperones and co-chaperones regulating the glucocorticoid receptor (GR), FK506 binding protein (FKBP) 51 is the most intensely investigated across different disciplines. This review provides an update on the role of the different co-chaperones of Hsp70 and Hsp90 in the regulation of GR function. The development leading to the focus on FKBP51 is outlined. Further, a survey of the vast literature on the mechanism and function of FKBP51 is provided. This includes its structure and biochemical function, its regulation on different levels—transcription, post-transcription, and post-translation—and its function in signaling pathways. The evidence portraying FKBP51 as a scaffolding protein organizing protein complexes rather than a chaperone contributing to the folding of individual proteins is collated. Finally, FKBP51’s involvement in physiology and disease is outlined, and the promising efforts in developing drugs targeting FKBP51 are discussed.
Collapse
Affiliation(s)
- Gabriel R Fries
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.
| | - Nils C Gassen
- Department of Translational Science in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | - Theo Rein
- Department of Translational Science in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
21
|
Abstract
Pharmacogenomic testing in psychiatry is becoming an established clinical procedure. Several vendors provide clinical interpretation of combinatorial pharmacogenomic testing of gene variants that have documented predictive implications regarding either pharmacologic response or adverse effects in depression and other psychiatric conditions. Such gene profiles have demonstrated improvements in outcome in depression, and reduction of cost of care of patients with inadequate clinical response. Additionally, several new gene variants are being studied to predict specific response in individuals. Many of these genes have demonstrated a role in the pathophysiology of depression or specific depressive symptoms. This article reviews the current state-of-the-art application of psychiatric pharmacogenomics.
Collapse
|
22
|
Merkulov VM, Merkulova TI, Bondar NP. Mechanisms of Brain Glucocorticoid Resistance in Stress-Induced Psychopathologies. BIOCHEMISTRY (MOSCOW) 2017; 82:351-365. [PMID: 28320277 DOI: 10.1134/s0006297917030142] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Exposure to stress activates the hypothalamic-pituitary-adrenal axis and leads to increased levels of glucocorticoid (GC) hormones. Prolonged elevation of GC levels causes neuronal dysfunction, decreases the density of synapses, and impairs neuronal plasticity. Decreased sensitivity to glucocorticoids (glucocorticoid resistance) that develops as a result of chronic stress is one of the characteristic features of stress-induced psychopathologies. In this article, we reviewed the published data on proposed molecular mechanisms that contribute to the development of glucocorticoid resistance in brain, including changes in the expression of the glucocorticoid receptor (GR) gene, biosynthesis of GR isoforms, and GR posttranslational modifications. We also present data on alterations in the expression of the FKBP5 gene encoding the main component of cell ultra-short negative feedback loop of GC signaling regulation. Recent discoveries on stress- and GR-induced changes in epigenetic modification patterns as well as normalizing action of antidepressants are discussed. GR and FKBP5 gene polymorphisms associated with stress-induced psychopathologies are described, and their role in glucocorticoid resistance is discussed.
Collapse
Affiliation(s)
- V M Merkulov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | | | | |
Collapse
|
23
|
Fabbri C, Hosak L, Mössner R, Giegling I, Mandelli L, Bellivier F, Claes S, Collier DA, Corrales A, Delisi LE, Gallo C, Gill M, Kennedy JL, Leboyer M, Lisoway A, Maier W, Marquez M, Massat I, Mors O, Muglia P, Nöthen MM, O'Donovan MC, Ospina-Duque J, Propping P, Shi Y, St Clair D, Thibaut F, Cichon S, Mendlewicz J, Rujescu D, Serretti A. Consensus paper of the WFSBP Task Force on Genetics: Genetics, epigenetics and gene expression markers of major depressive disorder and antidepressant response. World J Biol Psychiatry 2017; 18:5-28. [PMID: 27603714 DOI: 10.1080/15622975.2016.1208843] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 06/29/2016] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a heritable disease with a heavy personal and socio-economic burden. Antidepressants of different classes are prescribed to treat MDD, but reliable and reproducible markers of efficacy are not available for clinical use. Further complicating treatment, the diagnosis of MDD is not guided by objective criteria, resulting in the risk of under- or overtreatment. A number of markers of MDD and antidepressant response have been investigated at the genetic, epigenetic, gene expression and protein levels. Polymorphisms in genes involved in antidepressant metabolism (cytochrome P450 isoenzymes), antidepressant transport (ABCB1), glucocorticoid signalling (FKBP5) and serotonin neurotransmission (SLC6A4 and HTR2A) were among those included in the first pharmacogenetic assays that have been tested for clinical applicability. The results of these investigations were encouraging when examining patient-outcome improvement. Furthermore, a nine-serum biomarker panel (including BDNF, cortisol and soluble TNF-α receptor type II) showed good sensitivity and specificity in differentiating between MDD and healthy controls. These first diagnostic and response-predictive tests for MDD provided a source of optimism for future clinical applications. However, such findings should be considered very carefully because their benefit/cost ratio and clinical indications were not clearly demonstrated. Future tests may include combinations of different types of biomarkers and be specific for MDD subtypes or pathological dimensions.
Collapse
Affiliation(s)
- Chiara Fabbri
- a Department of Biomedical and Neuromotor Sciences , University of Bologna , Bologna , Italy
| | - Ladislav Hosak
- b Department of Psychiatrics , Charles University, Faculty of Medicine and University Hospital, Hradec Králové , Czech Republic
| | - Rainald Mössner
- c Department of Psychiatry and Psychotherapy , University of Tübingen , Tübingen , Germany
| | - Ina Giegling
- d Department of Psychiatry, Psychotherapy and Psychosomatics , Martin Luther University of Halle-Wittenberg , Halle , Germany
| | - Laura Mandelli
- a Department of Biomedical and Neuromotor Sciences , University of Bologna , Bologna , Italy
| | - Frank Bellivier
- e Fondation Fondamental, Créteil, France AP-HP , GH Saint-Louis-Lariboisière-Fernand-Widal, Pôle Neurosciences , Paris , France
| | - Stephan Claes
- f GRASP-Research Group, Department of Neuroscience , University of Leuven , Leuven , Belgium
| | - David A Collier
- g Social, Genetic and Developmental Psychiatry Centre , Institute of Psychiatry, King's College London , London , UK
| | - Alejo Corrales
- h National University (UNT) Argentina, Argentinean Association of Biological Psychiatry , Buenos Aires , Argentina
| | - Lynn E Delisi
- i VA Boston Health Care System , Brockton , MA , USA
| | - Carla Gallo
- j Departamento de Ciencias Celulares y Moleculares, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía , Universidad Peruana Cayetano Heredia , Lima , Peru
| | - Michael Gill
- k Neuropsychiatric Genetics Research Group, Department of Psychiatry , Trinity College Dublin , Dublin , Ireland
| | - James L Kennedy
- l Neurogenetics Section, Centre for Addiction and Mental Health , Toronto , Ontario , Canada
| | - Marion Leboyer
- m Faculté de Médecine , Université Paris-Est Créteil, Inserm U955, Equipe Psychiatrie Translationnelle , Créteil , France
| | - Amanda Lisoway
- l Neurogenetics Section, Centre for Addiction and Mental Health , Toronto , Ontario , Canada
| | - Wolfgang Maier
- n Department of Psychiatry , University of Bonn , Bonn , Germany
| | - Miguel Marquez
- o Director of ADINEU (Asistencia, Docencia e Investigación en Neurociencia) , Buenos Aires , Argentina
| | - Isabelle Massat
- p UNI - ULB Neurosciences Institute, ULB , Bruxelles , Belgium
| | - Ole Mors
- q Department P , Aarhus University Hospital , Risskov , Denmark
| | | | - Markus M Nöthen
- s Institute of Human Genetics , University of Bonn , Bonn , Germany
| | - Michael C O'Donovan
- t MRC Centre for Neuropsychiatric Genetics and Genomics , Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University , Cardiff , UK
| | - Jorge Ospina-Duque
- u Grupo de Investigación en Psiquiatría, Departamento de Psiquiatría, Facultad de Medicina , Universidad de Antioquia , Medellín , Colombia
| | | | - Yongyong Shi
- w Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education , Shanghai Jiao Tong University , Shanghai , China
| | - David St Clair
- x University of Aberdeen, Institute of Medical Sciences , Aberdeen , UK
| | - Florence Thibaut
- y University Hospital Cochin (Site Tarnier), University Sorbonne Paris Cité (Faculty of Medicine Paris Descartes), INSERM U 894 Centre Psychiatry and Neurosciences , Paris , France
| | - Sven Cichon
- z Division of Medical Genetics, Department of Biomedicine , University of Basel , Basel , Switzerland
| | - Julien Mendlewicz
- aa Laboratoire de Psychologie Medicale, Centre Européen de Psychologie Medicale , Université Libre de Bruxelles and Psy Pluriel , Brussels , Belgium
| | - Dan Rujescu
- d Department of Psychiatry, Psychotherapy and Psychosomatics , Martin Luther University of Halle-Wittenberg , Halle , Germany
| | - Alessandro Serretti
- a Department of Biomedical and Neuromotor Sciences , University of Bologna , Bologna , Italy
| |
Collapse
|
24
|
Ramos M, Berrogain C, Concha J, Lomba L, García CB, Ribate MP. Pharmacogenetic studies: a tool to improve antidepressant therapy. Drug Metab Pers Ther 2016; 31:197-204. [PMID: 27889704 DOI: 10.1515/dmpt-2016-0019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/20/2016] [Indexed: 06/06/2023]
Abstract
The World Health Organization (WHO) predicts that major depressive disorder (MDD) will be the second leading cause of death and disability by 2020. Nowadays, approximately 60-70% of patients with this disorder have shown the lack of effectiveness and tolerability of the therapy with antidepressants. The US Food and Drug Administration (FDA) and the European Medicine Agency (EMA) are including pharmacogenetic information in the labeling of several antidepressants. The presence of this information represents the relevance of genetic polymorphisms in drug response. These pharmacogenetic studies have been based on the knowledge of genes involved in pharmacokinetic (CYP2D6, CYP2C19 and ABCB1) and pharmacodynamic (SLC6A4, HTR2A, BDNF, GNB3 and FKBP5) processes of antidepressant medications. The knowledge of the genotype of patients with MDD is an important tool for personalized therapy that can improve their clinical response to treatment. In this review, we highlight the most relevant genes involved in the metabolism of antidepressants (ADs) or the genes related to the presence of adverse reactions.
Collapse
|
25
|
Pharmacogenetics and Imaging-Pharmacogenetics of Antidepressant Response: Towards Translational Strategies. CNS Drugs 2016; 30:1169-1189. [PMID: 27752945 DOI: 10.1007/s40263-016-0385-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Genetic variation underlies both the response to antidepressant treatment and the occurrence of side effects. Over the past two decades, a number of pharmacogenetic variants, among these the SCL6A4, BDNF, FKBP5, GNB3, GRIK4, and ABCB1 genes, have come to the forefront in this regard. However, small effects sizes, mixed results in independent samples, and conflicting meta-analyses results led to inherent difficulties in the field of pharmacogenetics translating these findings into clinical practice. Nearly all antidepressant pharmacogenetic variants have potentially pleiotropic effects in which they are associated with major depressive disorder, intermediate phenotypes involved in emotional processes, and brain areas affected by antidepressant treatment. The purpose of this article is to provide a comprehensive review of the advances made in the field of pharmacogenetics of antidepressant efficacy and side effects, imaging findings of antidepressant response, and the latest results in the expanding field of imaging-pharmacogenetics studies. We suggest there is mounting evidence that genetic factors exert their impact on treatment response by influencing brain structural and functional changes during antidepressant treatment, and combining neuroimaging and genetic methods may be a more powerful way to detect biological mechanisms of response than either method alone. The most promising imaging-pharmacogenetics findings exist for the SCL6A4 gene, with converging associations with antidepressant response, frontolimbic predictors of affective symptoms, and normalization of frontolimbic activity following antidepressant treatment. More research is required before imaging-pharmacogenetics informed personalized medicine can be applied to antidepressant treatment; nevertheless, inroads have been made towards assessing genetic and neuroanatomical liability and potential clinical application.
Collapse
|
26
|
Western High-Fat Diet Consumption during Adolescence Increases Susceptibility to Traumatic Stress while Selectively Disrupting Hippocampal and Ventricular Volumes. eNeuro 2016; 3:eN-NWR-0125-16. [PMID: 27844058 PMCID: PMC5099604 DOI: 10.1523/eneuro.0125-16.2016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 09/23/2016] [Accepted: 10/26/2016] [Indexed: 11/29/2022] Open
Abstract
Psychological trauma and obesity co-occur frequently and have been identified as major risk factors for psychiatric disorders. Surprisingly, preclinical studies examining how obesity disrupts the ability of the brain to cope with psychological trauma are lacking. The objective of this study was to determine whether an obesogenic Western-like high-fat diet (WD) predisposes rats to post-traumatic stress responsivity. Adolescent Lewis rats (postnatal day 28) were fed ad libitum for 8 weeks with either the experimental WD diet (41.4% kcal from fat) or the control diet (16.5% kcal from fat). We modeled psychological trauma by exposing young adult rats to a cat odor threat. The elevated plus maze and the open field test revealed increased psychological trauma-induced anxiety-like behaviors in the rats that consumed the WD when compared with control animals 1 week after undergoing traumatic stress (p < 0.05). Magnetic resonance imaging showed significant hippocampal atrophy (20% reduction) and lateral ventricular enlargement (50% increase) in the animals fed the WD when compared with controls. These volumetric abnormalities were associated with behavioral indices of anxiety, increased leptin and FK506-binding protein 51 (FKBP51) levels, and reduced hippocampal blood vessel density. We found asymmetric structural vulnerabilities to the WD, particularly the ventral and left hippocampus and lateral ventricle. This study highlights how WD consumption during adolescence impacts key substrates implicated in post-traumatic stress disorder. Understanding how consumption of a WD affects the developmental trajectories of the stress neurocircuitry is critical, as stress susceptibility imposes a marked vulnerability to neuropsychiatric disorders.
Collapse
|
27
|
De la Cruz-Cano E. Association between FKBP5 and CRHR1 genes with suicidal behavior: A systematic review. Behav Brain Res 2016; 317:46-61. [PMID: 27638035 DOI: 10.1016/j.bbr.2016.09.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 12/26/2022]
Abstract
Suicide is one of the leading causes of death around the world with approximately one million suicides per year. An increasing number of neurobiological studies implicate HPA system dysfunction in suicide behavior, stimulating genetic research to focus on genes related to this system. This systematic review was focused on searching a correlation between FKBP5 and CRHR1 genes with suicidal behavior. Therefore, an electronic search strategy, using PubMed, EBSCO and Cochrane Library databases, was conducted from the inception of the studies into the databases to July 2016. The inclusion criteria were: use of at least one analysis investigating the relation between either the genetic variants in FKBP5 and/or CRHR1 genes with suicidal behavior. 2) use of a case-control design; 3) investigation about suicidal behavior in the form of suicide completion or history of at least one suicide attempt, as defined by each individual study; 4) inclusion of samples comprising control subjects; and 6) inclusion of reports written only in English language. The PRISMA guidelines were followed and the search strategy ensured that all possible studies were identified to compile the review. Using the keyword combinations, the search strategy provided 3334 articles, of which only 15 case-control studies were included in this systematic review. The included studies comprised 2526 subjects with suicidal behavior. A quantitative synthesis of results from the included studies was not undertaken due to marked methodological heterogeneity. This review showed a significant genetic association in most studies in FKBP5 and CRHR1 genes with a high rate of attempted suicide, pointing out that the expression of these genes and its polymorphisms could be a key predictor of suicide risk. In conclusion, this systematic review supports an association between suicidal behavior and genetic variants in FKBP5 and CRHR1 genes.
Collapse
Affiliation(s)
- Eduardo De la Cruz-Cano
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, C.P. 86100, Mexico; Secretaría de Salud, Hospital General de Comalcalco, Departamento de Laboratorio de Análisis Clínicos, Comalcalco, Tabasco C.P. 86300, Mexico.
| |
Collapse
|
28
|
Fabbri C, Crisafulli C, Calabrò M, Spina E, Serretti A. Progress and prospects in pharmacogenetics of antidepressant drugs. Expert Opin Drug Metab Toxicol 2016; 12:1157-68. [DOI: 10.1080/17425255.2016.1202237] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Chiara Fabbri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Concetta Crisafulli
- Department of Biomedical Science, Odontoiatric and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Marco Calabrò
- Department of Biomedical Science, Odontoiatric and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Edoardo Spina
- Department of Biomedical Science, Odontoiatric and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Alessandro Serretti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|