1
|
Ye J, Duan C, Han J, Chen J, Sun N, Li Y, Yuan T, Peng D. Peripheral mitochondrial DNA as a neuroinflammatory biomarker for major depressive disorder. Neural Regen Res 2025; 20:1541-1554. [PMID: 38934398 PMCID: PMC11688552 DOI: 10.4103/nrr.nrr-d-23-01878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/09/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
In the pathogenesis of major depressive disorder, chronic stress-related neuroinflammation hinders favorable prognosis and antidepressant response. Mitochondrial DNA may be an inflammatory trigger, after its release from stress-induced dysfunctional central nervous system mitochondria into peripheral circulation. This evidence supports the potential use of peripheral mitochondrial DNA as a neuroinflammatory biomarker for the diagnosis and treatment of major depressive disorder. Herein, we critically review the neuroinflammation theory in major depressive disorder, providing compelling evidence that mitochondrial DNA release acts as a critical biological substrate, and that it constitutes the neuroinflammatory disease pathway. After its release, mitochondrial DNA can be carried in the exosomes and transported to extracellular spaces in the central nervous system and peripheral circulation. Detectable exosomes render encaged mitochondrial DNA relatively stable. This mitochondrial DNA in peripheral circulation can thus be directly detected in clinical practice. These characteristics illustrate the potential for mitochondrial DNA to serve as an innovative clinical biomarker and molecular treatment target for major depressive disorder. This review also highlights the future potential value of clinical applications combining mitochondrial DNA with a panel of other biomarkers, to improve diagnostic precision in major depressive disorder.
Collapse
Affiliation(s)
- Jinmei Ye
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Duan
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxin Han
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jinrong Chen
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yuan Li
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Daihui Peng
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Angeloni E, Germelli L, Costa B, Martini C, Da Pozzo E. Neurosteroids and Translocator Protein (TSPO) in neuroinflammation. Neurochem Int 2025; 182:105916. [PMID: 39681140 DOI: 10.1016/j.neuint.2024.105916] [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: 10/03/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Neurosteroids have a crucial role in physiological intrinsic regulations of the Central Nervous System functions. They are derived from peripheral steroidogenic sources and from the de novo neurosteroidogenic capacity of brain cells. Significant alterations of neurosteroid levels have been frequently observed in neuroinflammation and neurodegenerative diseases. Such level fluctuations may be useful for both diagnosis and treatment of these pathological conditions. Beyond steroid administration, enhancing the endogenous production by Translocator Protein (TSPO) targeting has been proposed to restore these altered pathological levels. However, the neurosteroid quantification and the prediction of their final effects are often troublesome, sometimes controversial and context dependent, due to the complexity of neurosteroid biosynthetic pathway and to the low produced amounts. The aim of this review is to report recent advances, and technical limitations, in neurosteroid-related strategies against neuroinflammation.
Collapse
Affiliation(s)
- Elisa Angeloni
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Lorenzo Germelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Eleonora Da Pozzo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy.
| |
Collapse
|
3
|
Qiu Y, Lu G, Zhang S, Minping L, Xue X, Junyu W, Zheng Z, Qi W, Guo J, Zhou D, Huang H, Deng Z. Mitochondrial dysfunction of Astrocyte induces cell activation under high salt condition. Heliyon 2024; 10:e40621. [PMID: 39660210 PMCID: PMC11629238 DOI: 10.1016/j.heliyon.2024.e40621] [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: 11/03/2023] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
Excess dietary sodium can accumulate in brain and adversely affect human health. We have confirmed in previous studies that high salt can induce activation of astrocyte manifested by the secretion of various inflammatory factors. In order to further explore the effect of high salt on the internal cell metabolism of astrocytes, RNA sequencing was performed on astrocytes under high salt environment, which indicated the oxidative phosphorylation and glycolysis pathways of astrocytes were downregulated. Next, we found that high salt concentrations elicited astrocyte mitochondrial morphology change, as evidenced by swelling from a short rod to a round shape through a High Intelligent and Sensitive Structured Illumination Microscope (HIS-SIM). Furthermore, we found that high salt concentrations reduced astrocyte mitochondrial oxygen consumption and membrane potential. Treatment with 18-kDa translocator protein (TSPO) ligands FGIN-1-27 improved mitochondrial networks and reversed astrocyte activation under high-salt circumstances. Our study shows that high salt can directly disrupt astrocytic mitochondrial homeostasis and function. Targeting translocator protein signaling may have therapeutic potential against high-salt neurotoxicity.
Collapse
Affiliation(s)
- Yuemin Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
- Department of Neurology, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, 518100, China
| | - Gengxin Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Shifeng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Li Minping
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Xu Xue
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Wu Junyu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Zhihui Zheng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Weiwei Qi
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Junjie Guo
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Dongxiao Zhou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Haiwei Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Zhezhi Deng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| |
Collapse
|
4
|
Bader S, Jahner T, Dörfelt A, Melchner D, Cardon I, Siegmund HI, Brochhausen C, Rupprecht R, Milenkovic VM, Wetzel CH. A Comprehensive Functional Investigation of the Human Translocator Protein 18 kDa (TSPO) in a Novel Human Neuronal Cell Knockout Model. Int J Mol Sci 2024; 25:12882. [PMID: 39684592 DOI: 10.3390/ijms252312882] [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: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
The translocator protein 18 kDa (TSPO) is a multifunctional outer mitochondrial membrane protein associated with various aspects of mitochondrial physiology and multiple roles in health and disease. Here, we aimed to analyse the role of TSPO in the regulation of mitochondrial and cellular functions in a human neuronal cell model. We used the CRISPR/Cas9 technology and generated TSPO knockout (KO) and control (CTRL) variants of human-induced pluripotent stem cells (hiPSCs). In a multimodal phenotyping approach, we investigated cellular and mitochondrial functions in neural progenitor cells (NPCs), astrocytes, and neurons differentiated from hiPSC CTRL and TSPO KO cell lines. Our analysis revealed reduced mitochondrial respiration and glycolysis, altered Ca2+ levels in the cytosol and mitochondrial matrix, a depolarised MMP, and increased levels of reactive oxygen species, as well as a reduced cell size. Notably, TSPO deficiency was accompanied by reduced expression of the voltage-dependent anion channel (VDAC). We also observed a reduced TSPO and VDAC expression in cells derived from patients suffering from major depressive disorder (MDD). Considering the modulatory function of TSPO and the similar functional phenotype of cells derived from patients with depression, we discuss a role of TSPO in the etiology or pathology of MDD. In summary, our findings indicate a general impairment of mitochondrial function in TSPO knockout (KO) cells. This deepens our insight into the intricate role of TSPO in a range of physiological and pathological processes.
Collapse
Affiliation(s)
- Stefanie Bader
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Tatjana Jahner
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Anett Dörfelt
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Doris Melchner
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Iseline Cardon
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Heiko I Siegmund
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
| | | | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
5
|
Fairley LH, Lai KO, Grimm A, Eckert A, Barron AM. The mitochondrial translocator protein (TSPO) in Alzheimer's disease: Therapeutic and immunomodulatory functions. Biochimie 2024; 224:120-131. [PMID: 38971458 DOI: 10.1016/j.biochi.2024.07.003] [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: 01/12/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The translocator protein (TSPO) has been widely investigated as a PET-imaging biomarker of neuroinflammation and, more recently, as a therapeutic target for the treatment of neurodegenerative disease. TSPO ligands have been shown to exert neuroprotective effects in vivo and in vitro models of Alzheimer's disease (AD), by reducing toxic beta amyloid peptides, and attenuating brain atrophy. Recent transcriptomic and proteomic analyses, and the generation of TSPO-KO mice, have enabled new insights into the mechanistic function of TSPO in AD. Using a multi-omics approach in both TSPO-KO- and TSPO ligand-treated mice, we have demonstrated a key role for TSPO in microglial respiratory metabolism and phagocytosis in AD. In this review, we discuss emerging evidence for therapeutic and immunomodulatory functions of TSPO in AD, and new tools for studying TSPO in the brain.
Collapse
Affiliation(s)
- Lauren H Fairley
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Kei Onn Lai
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Amandine Grimm
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anne Eckert
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore.
| |
Collapse
|
6
|
Jolly AA, Brown RB, Tozer DJ, Hong YT, Fryer TD, Aigbirhio FI, O’Brien JT, Markus HS. Are central and systemic inflammation associated with fatigue in cerebral small vessel disease? Int J Stroke 2024; 19:705-713. [PMID: 38533609 PMCID: PMC11292988 DOI: 10.1177/17474930241245613] [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: 01/10/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Fatigue is a common symptom in cerebral small vessel disease (SVD), but its pathogenesis is poorly understood. It has been suggested that inflammation may play a role. We determined whether central (neuro) inflammation and peripheral inflammation were associated with fatigue in SVD. METHODS Notably, 36 patients with moderate-to-severe SVD underwent neuropsychometric testing, combined positron emission tomography and magnetic resonance imaging (PET-MRI) scan, and blood draw for the analysis of inflammatory blood biomarkers. Microglial signal was taken as a proxy for neuroinflammation, assessed with radioligand 11C-PK11195. Of these, 30 subjects had full PET datasets for analysis. We assessed global 11C-PK11195 binding and hotspots of 11C-PK11195 binding in the normal-appearing white matter, lesioned tissue, and combined total white matter. Peripheral inflammation was assessed with serum C-reactive protein (CRP) and using the Olink cardiovascular III proteomic panel comprising 92 biomarkers of cardiovascular inflammation and endothelial activation. Fatigue was assessed using the fatigue severity scale (FSS), the visual analog fatigue scale, and a subscale of the Geriatric Depression Scale. RESULTS Mean (SD) age was 68.7 (11.2) years, and 63.9% were male. Of these, 55.6% showed fatigue on the FSS. Fatigued participants had higher disability scores (p = 0.02), higher total GDS scores (p = 0.02), and more commonly reported a history of depression (p = 0.04). 11C-PK11195 ligand binding in the white matter was not associated with any measure of fatigue. Serum CRP was significantly associated with average fatigue score on FSS (ρ = 0.48, p = 0.004); this association persisted when controlling for age, sex, disability score, and depression (β = 0.49, 95% CI (0.17, 2.26), p = 0.03). Blood biomarkers from the Olink panel showed no association with fatigue. CONCLUSION In symptomatic SVD patients, neuroinflammation, assessed with microglial marker 11C-PK11195, was not associated with fatigue. We found some evidence for a role of systematic inflammation, evidenced by an association between fatigue severity and raised CRP, but further studies are required to understand this relationship and inform whether it could be therapeutically modified to reduce fatigue severity. DATA ACCESS STATEMENT Data for this study are available from the corresponding author upon reasonable request.
Collapse
Affiliation(s)
- Amy A Jolly
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Robin B Brown
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel J Tozer
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Firth W, Robb JL, Stewart D, Pye KR, Bamford R, Oguro-Ando A, Beall C, Ellacott KLJ. Regulation of astrocyte metabolism by mitochondrial translocator protein 18 kDa. J Neurochem 2024; 168:1374-1401. [PMID: 38482552 DOI: 10.1111/jnc.16089] [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: 10/09/2023] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 03/26/2024]
Abstract
The mitochondrial translocator protein 18 kDa (TSPO) has been linked to functions from steroidogenesis to regulation of cellular metabolism and is an attractive therapeutic target for chronic CNS inflammation. Studies in Leydig cells and microglia indicate that TSPO function may vary between cells depending on their specialized roles. Astrocytes are critical for providing trophic and metabolic support in the brain. Recent work has highlighted that TSPO expression increases in astrocytes under inflamed conditions and may drive astrocyte reactivity. Relatively little is known about the role TSPO plays in regulating astrocyte metabolism and whether this protein is involved in immunometabolic processes in these cells. Using TSPO-deficient (TSPO-/-) mouse primary astrocytes in vitro (MPAs) and a human astrocytoma cell line (U373 cells), we performed extracellular metabolic flux analyses. We found that TSPO deficiency reduced basal cellular respiration and attenuated the bioenergetic response to glucopenia. Fatty acid oxidation was increased, and lactate production was reduced in TSPO-/- MPAs and U373 cells. Co-immunoprecipitation studies revealed that TSPO forms a complex with carnitine palmitoyltransferase 1a in U373 and MPAs, presenting a mechanism wherein TSPO may regulate FAO in these cells. Compared to TSPO+/+ cells, in TSPO-/- MPAs we observed attenuated tumor necrosis factor release following 3 h lipopolysaccharide (LPS) stimulation, which was enhanced at 24 h post-LPS stimulation. Together these data suggest that while TSPO acts as a regulator of metabolic flexibility, TSPO deficiency does not appear to modulate the metabolic response of MPAs to inflammation, at least in response to the model used in this study.
Collapse
Affiliation(s)
- Wyn Firth
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Josephine L Robb
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Daisy Stewart
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Katherine R Pye
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Rosemary Bamford
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Asami Oguro-Ando
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Craig Beall
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kate L J Ellacott
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
8
|
Triebelhorn J, Cardon I, Kuffner K, Bader S, Jahner T, Meindl K, Rothhammer-Hampl T, Riemenschneider MJ, Drexler K, Berneburg M, Nothdurfter C, Manook A, Brochhausen C, Baghai TC, Hilbert S, Rupprecht R, Milenkovic VM, Wetzel CH. Induced neural progenitor cells and iPS-neurons from major depressive disorder patients show altered bioenergetics and electrophysiological properties. Mol Psychiatry 2024; 29:1217-1227. [PMID: 35732695 PMCID: PMC11189806 DOI: 10.1038/s41380-022-01660-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022]
Abstract
The molecular pathomechanisms of major depressive disorder (MDD) are still not completely understood. Here, we follow the hypothesis, that mitochondria dysfunction which is inevitably associated with bioenergetic disbalance is a risk factor that contributes to the susceptibility of an individual to develop MDD. Thus, we investigated molecular mechanisms related to mitochondrial function in induced neuronal progenitor cells (NPCs) which were reprogrammed from fibroblasts of eight MDD patients and eight non-depressed controls. We found significantly lower maximal respiration rates, altered cytosolic basal calcium levels, and smaller soma size in NPCs derived from MDD patients. These findings are partially consistent with our earlier observations in MDD patient-derived fibroblasts. Furthermore, we differentiated MDD and control NPCs into iPS-neurons and analyzed their passive biophysical and active electrophysiological properties to investigate whether neuronal function can be related to altered mitochondrial activity and bioenergetics. Interestingly, MDD patient-derived iPS-neurons showed significantly lower membrane capacitance, a less hyperpolarized membrane potential, increased Na+ current density and increased spontaneous electrical activity. Our findings indicate that functional differences evident in fibroblasts derived from MDD patients are partially present after reprogramming to induced-NPCs, could relate to altered function of iPS-neurons and thus might be associated with the aetiology of major depressive disorder.
Collapse
Affiliation(s)
- Julian Triebelhorn
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Iseline Cardon
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Kerstin Kuffner
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Stefanie Bader
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Tatjana Jahner
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Katrin Meindl
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Tanja Rothhammer-Hampl
- Department of Neuropathology, Regensburg University Hospital, 93053, Regensburg, Germany
| | | | - Konstantin Drexler
- Department of Dermatology, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Caroline Nothdurfter
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - André Manook
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
- Central Biobank of the University of Regensburg and the Regensburg University Hospital, 93053, Regensburg, Germany
| | - Thomas C Baghai
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Sven Hilbert
- Institute of Educational Research, Faculty of Human Sciences, University of Regensburg, 93053, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
9
|
Hector M, Langmann T, Wolf A. Translocator protein (18 kDa) (Tspo) in the retina and implications for ocular diseases. Prog Retin Eye Res 2024; 100:101249. [PMID: 38430990 DOI: 10.1016/j.preteyeres.2024.101249] [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: 12/05/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Translocator protein (18 kDa) (Tspo), formerly known as peripheral benzodiazepine receptor is a highly conserved transmembrane protein primarily located in the outer mitochondrial membrane. In the central nervous system (CNS), especially in glia cells, Tspo is upregulated upon inflammation. Consequently, Tspo was used as a tool for diagnostic in vivo imaging of neuroinflammation in the brain and as a potential therapeutic target. Several synthetic Tspo ligands have been explored as immunomodulatory and neuroprotective therapy approaches. Although the function of Tspo and how its ligands exert these beneficial effects is not fully clear, it became a research topic of interest, especially in ocular diseases in the past few years. This review summarizes state-of-the-art knowledge of Tspo expression and its proposed functions in different cells of the retina including microglia, retinal pigment epithelium and Müller cells. Tspo is involved in cytokine signaling, oxidative stress and reactive oxygen species production, calcium signaling, neurosteroid synthesis, energy metabolism, and cholesterol efflux. We also highlight recent developments in preclinical models targeting Tspo and summarize the relevance of Tspo biology for ocular and retinal diseases. We conclude that glial upregulation of Tspo in different ocular pathologies and the use of Tspo ligands as promising therapeutic approaches in preclinical studies underline the importance of Tspo as a potential disease-modifying protein.
Collapse
Affiliation(s)
- Mandy Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
10
|
Liu Y, Zhang Z, He Y, Li R, Zhang Y, Liu H, Wang Y, Ma W. Mitochondria protective and anti-apoptotic effects of peripheral benzodiazepine receptor and its ligands on the treatment of asthma in vitro and vivo. J Inflamm (Lond) 2024; 21:11. [PMID: 38641850 PMCID: PMC11031857 DOI: 10.1186/s12950-024-00383-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 04/03/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Asthma is a prevalent respiratory inflammatory disease. Abnormal apoptosis of bronchial epithelial cells is one of the major factors in the progression of asthma. Peripheral benzodiazepine receptors are highly expressed in bronchial epithelial cells, which act as a component of the mitochondrial permeability transition pore to regulate its opening and closing and apoptosis of bronchial epithelial cells. We aimed to investigate the mechanisms by which peripheral benzodiazepine receptor and its ligands, agonist 4'-Chlorodiazepam (Ro5-4864) and antagonist 1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide (PK 11,195), modulate the mitochondrial function and cell apoptosis in the treatment of asthma. METHODS In vitro study, Ro5-4864 and PK 11,195 were utilized to pretreat cells prior to the inflammatory injury induced by Lipopolysaccharide. The reactive oxygen species, the apoptosis of cell, the mitochondrial membrane potentials, the ultrastructures of the mitochondria and the expression levels of peripheral benzodiazepine receptors and apoptosis-related proteins and genes were detected. In vivo study, mice were administrated intraperitoneally with Ro5-4864 and PK 11,195 before sensitized and challenged by ovalbumin. Serum IgE and bronchoalveolar lavage fluid cytokines were detected, and lung tissues were underwent the histopathological examination. RESULTS The ligands of peripheral benzodiazepine receptor counteracted the effects of the increase of reactive oxygen species, the elevated extent of apoptosis, the decrease of mitochondrial membrane potentials and the disruption of mitochondrial ultrastructures induced by Lipopolysaccharide. The ligands also promoted the expression of anti-apoptosis-related proteins and genes and inhibited the expression of pro-apoptosis-related proteins and genes. Besides, the ligands reduced the levels of serum IgE and bronchoalveolar lavage fluid cytokines in asthmatic mice and attenuated the histopathological damage of lungs. CONCLUSION Peripheral benzodiazepine receptor serves as a potential therapeutic target for the treatment of asthma, with its ligands exerting mitochondrial protective and anti-apoptotic effects on bronchial epithelial cells.
Collapse
Affiliation(s)
- Yurui Liu
- Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China
| | - Zhengze Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China
| | - Yuewen He
- Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China
| | - Ruogen Li
- Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China
| | - Yuhao Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China
| | - Hao Liu
- Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China
| | - Yong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China
| | - Wuhua Ma
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, 510405, Guangzhou, P.R. China.
| |
Collapse
|
11
|
Wheeler S, Bhardwaj M, Kenyon V, Ferraz MJ, Aerts JMFG, Sillence DJ. Mitochondrial dysfunction in NPC1-deficiency is not rescued by drugs targeting the glucosylceramidase GBA2 and the cholesterol-binding proteins TSPO and StARD1. FEBS Lett 2024; 598:477-484. [PMID: 38302739 DOI: 10.1002/1873-3468.14802] [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: 10/03/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 02/03/2024]
Abstract
Niemann-Pick type C disease (NPCD) is a rare neurodegenerative disorder most commonly caused by mutations in the lysosomal protein Niemann-Pick C1 (NPC1), which is implicated in cholesterol export. Mitochondrial insufficiency forms a significant feature of the pathology of this disease, yet studies attempting to address this are rare. The working hypothesis is that mitochondria become overloaded with cholesterol which renders them dysfunctional. We examined two potential protein targets-translocator protein (TSPO) and steroidogenic acute regulatory protein D1 (StARD1)-which are implicated in cholesterol transport to mitochondria, in addition to glucocerbrosidase 2 (GBA2), the target of miglustat, which is currently the only approved treatment for NPCD. However, inhibiting these proteins did not correct the mitochondrial defect in NPC1-deficient cells.
Collapse
Affiliation(s)
- Simon Wheeler
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | | | | | - Maria J Ferraz
- Leiden Institute of Chemistry, Leiden University, The Netherlands
| | | | - Dan J Sillence
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| |
Collapse
|
12
|
Zhang S, Deng Z, Qiu Y, Lu G, Wu J, Huang H. FGIN-1-27 Mitigates Radiation-induced Mitochondrial Hyperfunction and Cellular Hyperactivation in Cultured Astrocytes. Neuroscience 2023; 535:23-35. [PMID: 37913861 DOI: 10.1016/j.neuroscience.2023.10.017] [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: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
Radiation-induced brain injury (RBI) poses a significant challenge in the context of radiotherapy for intracranial tumors, necessitating a comprehensive understanding of the cellular and molecular mechanisms involved. While prior investigations have underscored the role of astrocyte activation and excessive vascular endothelial growth factor production in microvascular damage associated with RBI, there remains a scarcity of studies examining the impact of radiation on astrocytes, particularly regarding organelles such as mitochondria. Thus, our study aimed to elucidate alterations in astrocyte and mitochondrial functionality following radiation exposure, with a specific focus on evaluating the potential ameliorative effects of translocator protein 18 kDa(TSPO) ligands. In this study, cultured astrocytes were subjected to X-ray irradiation, and their cellular states and mitochondrial functions were examined and compared to control cells. Our findings revealed that radiation-induced astrocytic hyperactivation, transforming them into the neurotoxic A1-type, concomitant with reduced cell proliferation. Additionally, radiation triggered mitochondrial hyperfunction, heightened the mitochondrial membrane potential, and increased oxidative metabolite production. However, following treatment with FGIN-1-27, a TSPO ligand, we observed a restoration of mitochondrial function and a reduction in oxidative metabolite production. Moreover, this intervention mitigated astrocyte hyperactivity, decreased the number of A1-type astrocytes, and restored cell proliferative capacity. In conclusion, our study has unveiled additional manifestations of radiation-induced astrocyte dysfunction and validated that TSPO ligands may serve as a promising therapeutic strategy to mitigate this dysfunction. It has potential clinical implications for the treatment of RBI.
Collapse
Affiliation(s)
- Shifeng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Zhezhi Deng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yuemin Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Gengxin Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Junyu Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Haiwei Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China.
| |
Collapse
|
13
|
Chen H, Guo Z, Sun Y, Dai X. The immunometabolic reprogramming of microglia in Alzheimer's disease. Neurochem Int 2023; 171:105614. [PMID: 37748710 DOI: 10.1016/j.neuint.2023.105614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder (NDD). In the central nervous system (CNS), immune cells like microglia could reprogram intracellular metabolism to alter or exert cellular immune functions in response to environmental stimuli. In AD, microglia could be activated and differentiated into pro-inflammatory or anti-inflammatory phenotypes, and these differences in cellular phenotypes resulted in variance in cellular energy metabolism. Considering the enormous energy requirement of microglia for immune functions, the changes in mitochondria-centered energy metabolism and substrates of microglia are crucial for the cellular regulation of immune responses. Here we reviewed the mechanisms of microglial metabolic reprogramming by analyzing their flexible metabolic patterns and changes that occurred in their metabolism during the development of AD. Further, we summarized the role of drugs in modulating immunometabolic reprogramming to prevent neuroinflammation, which may shed light on a new research direction for AD treatment.
Collapse
Affiliation(s)
- Hongli Chen
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Zichen Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| |
Collapse
|
14
|
Firth W, Robb JL, Stewart D, Pye KR, Bamford R, Oguro-Ando A, Beall C, Ellacott KLJ. Regulation of astrocyte metabolism by mitochondrial translocator protein 18kDa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560159. [PMID: 37873215 PMCID: PMC10592862 DOI: 10.1101/2023.09.29.560159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The mitochondrial translocator protein 18kDa (TSPO) has been linked to a variety of functions from steroidogenesis to regulation of cellular metabolism and is an attractive therapeutic target for chronic CNS inflammation. Studies in the periphery using Leydig cells and hepatocytes, as well as work in microglia, indicate that the function of TSPO may vary between cells depending on their specialised roles. Astrocytes are critical for providing trophic and metabolic support in the brain as part of their role in maintaining brain homeostasis. Recent work has highlighted that TSPO expression increases in astrocytes under inflamed conditions and may drive astrocyte reactivity. However, relatively little is known about the role TSPO plays in regulating astrocyte metabolism and whether this protein is involved in immunometabolic processes in these cells. Using TSPO-deficient (TSPO-/-) mouse primary astrocytes in vitro (MPAs) and a human astrocytoma cell line (U373 cells), we performed metabolic flux analyses. We found that loss of TSPO reduced basal astrocyte respiration and increased the bioenergetic response to glucose reintroduction following glucopenia, while increasing fatty acid oxidation (FAO). Lactate production was significantly reduced in TSPO-/- astrocytes. Co-immunoprecipitation studies in U373 cells revealed that TSPO forms a complex with carnitine palmitoyltransferase 1a, which presents a mechanism wherein TSPO may regulate FAO in astrocytes. Compared to TSPO+/+ cells, inflammation induced by 3h lipopolysaccharide (LPS) stimulation of TSPO-/- MPAs revealed attenuated tumour necrosis factor release, which was enhanced in TSPO-/- MPAs at 24h LPS stimulation. Together these data suggest that while TSPO acts as a regulator of metabolic flexibility in astrocytes, loss of TSPO does not appear to modulate the metabolic response of astrocytes to inflammation, at least in response to the stimulus/time course used in this study.
Collapse
Affiliation(s)
- Wyn Firth
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Josephine L Robb
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Daisy Stewart
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Katherine R Pye
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Rosemary Bamford
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Asami Oguro-Ando
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Craig Beall
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kate LJ Ellacott
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
15
|
Tournier B, Bouteldja F, Amossé Q, Nicolaides A, Duarte Azevedo M, Tenenbaum L, Garibotto V, Ceyzériat K, Millet P. 18 kDa Translocator Protein TSPO Is a Mediator of Astrocyte Reactivity. ACS OMEGA 2023; 8:31225-31236. [PMID: 37663488 PMCID: PMC10468775 DOI: 10.1021/acsomega.3c03368] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/01/2023] [Indexed: 09/05/2023]
Abstract
An increase in astrocyte reactivity has been described in Alzheimer's disease and seems to be related to the presence of a pro-inflammatory environment. Reactive astrocytes show an increase in the density of the 18 kDa translocator protein (TSPO), but TSPO involvement in astrocyte functions remains poorly understood. The goal of this study was to better characterize the mechanisms leading to the increase in TSPO under inflammatory conditions and the associated consequences. For this purpose, the C6 astrocytic cell line was used in the presence of lipopolysaccharide (LPS) or TSPO overexpression mediated by the transfection of a plasmid encoding TSPO. The results show that nonlethal doses of LPS induced TSPO expression at mRNA and protein levels through a STAT3-dependent mechanism and increased the number of mitochondria per cell. LPS stimulated reactive oxygen species (ROS) production and decreased glucose consumption (quantified by the [18F]FDG uptake), and these effects were diminished by FEPPA, a TSPO antagonist. The transfection-mediated overexpression of TSPO induced ROS production, and this effect was blocked by FEPPA. In addition, a synergistic effect of overexpression of TSPO and LPS on ROS production was observed. These data show that the increase of TSPO in astrocytic cells is involved in the regulation of glucose metabolism and in the pro-inflammatory response. These data suggest that the overexpression of TSPO by astrocytes in Alzheimer's disease would have rather deleterious effects by promoting the pro-inflammatory response.
Collapse
Affiliation(s)
- Benjamin
B. Tournier
- Department
of Psychiatry, University Hospitals of Geneva, Geneva 1206, Switzerland
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Farha Bouteldja
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Quentin Amossé
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Alekos Nicolaides
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Marcelo Duarte Azevedo
- Laboratory
of Cellular and Molecular Neurotherapies, Center for Neuroscience
Research, Clinical Neuroscience Department, Lausanne University Hospital, Lausanne 1011, Switzerland
| | - Liliane Tenenbaum
- Laboratory
of Cellular and Molecular Neurotherapies, Center for Neuroscience
Research, Clinical Neuroscience Department, Lausanne University Hospital, Lausanne 1011, Switzerland
| | - Valentina Garibotto
- Division
of Nuclear Medicine, Diagnostic Department, University Hospitals of Geneva, Geneva 1206, Switzerland
- CIBM
Center for BioMedical Imaging; NIMTLab, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Kelly Ceyzériat
- Department
of Psychiatry, University Hospitals of Geneva, Geneva 1206, Switzerland
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
- Division
of Nuclear Medicine, Diagnostic Department, University Hospitals of Geneva, Geneva 1206, Switzerland
- CIBM
Center for BioMedical Imaging; NIMTLab, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Philippe Millet
- Department
of Psychiatry, University Hospitals of Geneva, Geneva 1206, Switzerland
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
16
|
Cheung G, Lin YC, Papadopoulos V. Translocator protein in the rise and fall of central nervous system neurons. Front Cell Neurosci 2023; 17:1210205. [PMID: 37416505 PMCID: PMC10322222 DOI: 10.3389/fncel.2023.1210205] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
Translocator protein (TSPO), a 18 kDa protein found in the outer mitochondrial membrane, has historically been associated with the transport of cholesterol in highly steroidogenic tissues though it is found in all cells throughout the mammalian body. TSPO has also been associated with molecular transport, oxidative stress, apoptosis, and energy metabolism. TSPO levels are typically low in the central nervous system (CNS), but a significant upregulation is observed in activated microglia during neuroinflammation. However, there are also a few specific regions that have been reported to have higher TSPO levels than the rest of the brain under normal conditions. These include the dentate gyrus of the hippocampus, the olfactory bulb, the subventricular zone, the choroid plexus, and the cerebellum. These areas are also all associated with adult neurogenesis, yet there is no explanation of TSPO's function in these cells. Current studies have investigated the role of TSPO in microglia during neuron degeneration, but TSPO's role in the rest of the neuron lifecycle remains to be elucidated. This review aims to discuss the known functions of TSPO and its potential role in the lifecycle of neurons within the CNS.
Collapse
|
17
|
Bader S, Würfel T, Jahner T, Nothdurfter C, Rupprecht R, Milenkovic VM, Wetzel CH. Impact of Translocator Protein 18 kDa (TSPO) Deficiency on Mitochondrial Function and the Inflammatory State of Human C20 Microglia Cells. Cells 2023; 12:cells12060954. [PMID: 36980295 PMCID: PMC10046935 DOI: 10.3390/cells12060954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/01/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Microglia are the resident immune cells of the central nervous system. Upon stimulus presentation, microglia polarize from a resting to an activated state. Microglial translocator protein 18 kDa (TSPO) is considered a marker of inflammation. Here, we characterized the role of TSPO by investigating the impact of TSPO deficiency on human microglia. We used TSPO knockout (TSPO-/-) variants of the human C20 microglia cell line. We found a significant reduction in the TSPO-associated protein VDAC1 in TSPO-/- cells compared to control cells. Moreover, we assessed the impact of TSPO deficiency on calcium levels and the mitochondrial membrane potential. Cytosolic and mitochondrial calcium concentrations were increased in TSPO-/- cell lines, whereas the mitochondrial membrane potential tended to be lower. Assessment of the mitochondrial DNA copy number via RT-PCR revealed a decreased amount of mtDNA in the TSPO-/- cells when compared to controls. Moreover, the metabolic profiles of C20 cells were strongly dependent on the glycolytic pathway. However, TSPO depletion did not affect the cellular metabolic profile. Measurement of the mRNA expression levels of the pro-inflammatory mediators revealed an attenuated response to pro-inflammatory stimuli in TSPO-depleted cells, implying a role for the TSPO protein in the process of microglial polarization.
Collapse
Affiliation(s)
- Stefanie Bader
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Thea Würfel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Tatjana Jahner
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Caroline Nothdurfter
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
18
|
Rupprecht R, Wetzel CH, Dorostkar M, Herms J, Albert NL, Schwarzbach J, Schumacher M, Neumann ID. Translocator protein (18kDa) TSPO: a new diagnostic or therapeutic target for stress-related disorders? Mol Psychiatry 2022; 27:2918-2926. [PMID: 35444254 DOI: 10.1038/s41380-022-01561-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022]
Abstract
Efficient treatment of stress-related disorders, such as depression, is still a major challenge. The onset of antidepressant drug action is generally quite slow, while the anxiolytic action of benzodiazepines is considerably faster. However, their long-term use is impaired by tolerance development, abuse liability and cognitive impairment. Benzodiazepines act as positive allosteric modulators of ɣ-aminobutyric acid type A (GABAA) receptors. 3α-reduced neurosteroids such as allopregnanolone also are positive allosteric GABAA receptor modulators, however, through a site different from that targeted by benzodiazepines. Recently, the administration of neurosteroids such as brexanolone or zuranolone has been shown to rapidly ameliorate symptoms in post-partum depression or major depressive disorder. An attractive alternative to the administration of exogenous neurosteroids is promoting endogenous neurosteroidogenesis via the translocator protein 18k Da (TSPO). TSPO is a transmembrane protein located primarily in mitochondria, which mediates numerous biological functions, e.g., steroidogenesis and mitochondrial bioenergetics. TSPO ligands have been used in positron emission tomography (PET) studies as putative markers of microglia activation and neuroinflammation in stress-related disorders. Moreover, TSPO ligands have been shown to modulate neuroplasticity and to elicit antidepressant and anxiolytic therapeutic effects in animals and humans. As such, TSPO may open new avenues for understanding the pathophysiology of stress-related disorders and for the development of novel treatment options.
Collapse
Affiliation(s)
- Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Mario Dorostkar
- Center for Neuropathology and Prion Research, Ludwig-Maximilian-University Munich, 81377, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilian-University Munich, 81377, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilian-University Munich, 81377, Munich, Germany
| | - Jens Schwarzbach
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Michael Schumacher
- Research Unit 1195, INSERM and University Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
| | - Inga D Neumann
- Department of Neurobiology and Animal Physiology, University Regensburg, 93040, Regensburg, Germany
| |
Collapse
|
19
|
Lin YC, Cheung G, Porter E, Papadopoulos V. The neurosteroid pregnenolone is synthesized by a mitochondrial P450 enzyme other than CYP11A1 in human glial cells. J Biol Chem 2022; 298:102110. [PMID: 35688208 PMCID: PMC9278081 DOI: 10.1016/j.jbc.2022.102110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 11/03/2022] Open
Abstract
Neurosteroids, modulators of neuronal and glial cell functions, are synthesized in the nervous system from cholesterol. In peripheral steroidogenic tissues, cholesterol is converted to the major steroid precursor pregnenolone by the CYP11A1 enzyme. Although pregnenolone is one of the most abundant neurosteroids in the brain, expression of CYP11A1 is difficult to detect. We found that human glial cells produced pregnenolone, detectable by mass spectrometry and ELISA, despite the absence of observable immunoreactive CYP11A1 protein. Unlike testicular and adrenal cortical cells, pregnenolone production in glial cells was not inhibited by CYP11A1 inhibitors DL-aminoglutethimide and ketoconazole. Furthermore, addition of hydroxycholesterols increased pregnenolone synthesis, suggesting desmolase activity that was not blocked by DL-aminoglutethimide or ketoconazole. We explored three different possibilities for an alternative pathway for glial cell pregnenolone synthesis: (1) regulation by reactive oxygen species, (2) metabolism via a different CYP11A1 isoform, and (3) metabolism via another CYP450 enzyme. First, we found oxidants and antioxidants had no significant effects on pregnenolone synthesis, suggesting it is not regulated by reactive oxygen species. Second, overexpression of CYP11A1 isoform b did not alter synthesis, indicating use of another CYP11A1 isoform is unlikely. Finally, we show nitric oxide and iron chelators deferoxamine and deferiprone significantly inhibited pregnenolone production, indicating involvement of another CYP450 enzyme. Ultimately, knockdown of endoplasmic reticulum cofactor NADPH-cytochrome P450 reductase had no effect, while knockdown of mitochondrial CYP450 cofactor ferredoxin reductase inhibited pregnenolone production. These data suggest that pregnenolone is synthesized by a mitochondrial cytochrome P450 enzyme other than CYP11A1 in human glial cells.
Collapse
|
20
|
Brown RB, Tozer DJ, Loubière L, Hong YT, Fryer TD, Williams GB, Graves MJ, Aigbirhio FI, O’Brien JT, Markus HS. MINocyclinE to Reduce inflammation and blood brain barrier leakage in small Vessel diseAse (MINERVA) trial study protocol. Eur Stroke J 2022; 7:323-330. [PMID: 36082255 PMCID: PMC9445404 DOI: 10.1177/23969873221100338] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Cerebral small vessel disease (SVD) is a common cause of stroke and cognitive impairment. Recent data has implicated neuroinflammation and increased blood-brain barrier (BBB) permeability in its pathogenesis, but whether such processes are causal and can be therapeutically modified is uncertain. In a rodent model of SVD, minocycline was associated with reduced white matter lesions, inflammation and BBB permeability. Aims: To determine whether blood-brain barrier permeability (measured using dynamic contrast-enhanced MRI) and microglial activation (measured by positron emission tomography using the radioligand 11C-PK11195) can be modified in SVD. Design: Phase II randomised double blind, placebo-controlled trial of minocycline 100 mg twice daily for 3 months in 44 participants with moderate to severe SVD defined as a clinical lacunar stroke and confluent white matter hyperintensities. Outcomes: Primary outcome measures are volume and intensity of focal increases of blood-brain barrier permeability and microglial activation determined using PET-MRI imaging. Secondary outcome measures include inflammatory biomarkers in serum, and change in conventional MRI markers and cognitive performance over 1 year follow up. Discussion: The MINERVA trial aims to test whether minocycline can influence novel pathological processes thought to be involved in SVD progression, and will provide insights into whether central nervous system inflammation in SVD can be therapeutically modulated.
Collapse
Affiliation(s)
- Robin B Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel J Tozer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Laurence Loubière
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Guy B Williams
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Singh P, Adhikari A, Singh D, Gond C, Tiwari AK. The 18-kDa Translocator Protein PET Tracers as a Diagnostic Marker for Neuroinflammation: Development and Current Standing. ACS OMEGA 2022; 7:14412-14429. [PMID: 35557664 PMCID: PMC9089361 DOI: 10.1021/acsomega.2c00588] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/05/2022] [Indexed: 05/13/2023]
Abstract
Translocator protein (TSPO, 18 kDa) is an evolutionary, well-preserved, and tryptophan-rich 169-amino-acid protein which localizes on the contact sites between the outer and inner mitochondrial membranes of steroid-synthesizing cells. This mitochondrial protein is implicated in an extensive range of cellular activities, including steroid synthesis, cholesterol transport, apoptosis, mitochondrial respiration, and cell proliferation. The upregulation of TSPO is well documented in diverse disease conditions including neuroinflammation, cancer, brain injury, and inflammation in peripheral organs. On the basis of these outcomes, TSPO has been assumed to be a fascinating subcellular target for early stage imaging of the diseased state and for therapeutic purposes. The main outline of this Review is to give an update on dealing with the advances made in TSPO PET tracers for neuroinflammation, synchronously emphasizing the approaches applied for the design and advancement of new tracers with reference to their structure-activity relationship (SAR).
Collapse
Affiliation(s)
- Priya Singh
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Anupriya Adhikari
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Deepika Singh
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Chandraprakash Gond
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Anjani Kumar Tiwari
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
- Address:
Department of Chemistry,
Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh. Tel.: +91-7503381343. Fax: +91-522-2440821. E-mail:
| |
Collapse
|
22
|
Nicotinic Acetylcholine Receptors and Microglia as Therapeutic and Imaging Targets in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092780. [PMID: 35566132 PMCID: PMC9102429 DOI: 10.3390/molecules27092780] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022]
Abstract
Amyloid-β (Aβ) accumulation and tauopathy are considered the pathological hallmarks of Alzheimer’s disease (AD), but attenuation in choline signaling, including decreased nicotinic acetylcholine receptors (nAChRs), is evident in the early phase of AD. Currently, there are no drugs that can suppress the progression of AD due to a limited understanding of AD pathophysiology. For this, diagnostic methods that can assess disease progression non-invasively before the onset of AD symptoms are essential, and it would be valuable to incorporate the concept of neurotheranostics, which simultaneously enables diagnosis and treatment. The neuroprotective pathways activated by nAChRs are attractive targets as these receptors may regulate microglial-mediated neuroinflammation. Microglia exhibit both pro- and anti-inflammatory functions that could be modulated to mitigate AD pathogenesis. Currently, single-cell analysis is identifying microglial subpopulations that may have specific functions in different stages of AD pathologies. Thus, the ability to image nAChRs and microglia in AD according to the stage of the disease in the living brain may lead to the development of new diagnostic and therapeutic methods. In this review, we summarize and discuss the recent findings on the nAChRs and microglia, as well as their methods for live imaging in the context of diagnosis, prophylaxis, and therapy for AD.
Collapse
|
23
|
Bloms-Funke P, Schumacher M, Liu S, Su D, Li J, Liere P, Rupprecht R, Nothdurfter C, Bahrenberg G, Christoph T, Habermann C, Kneip C, Schröder W, Tzschentke TM, Saunders D. A novel dual mode-of-action anti-hyperalgesic compound in rats which is neuroprotective and promotes neuroregeneration. Eur J Pharmacol 2022; 923:174935. [PMID: 35378102 DOI: 10.1016/j.ejphar.2022.174935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 12/25/2022]
Abstract
Chronic neuropathic pain (CNP) can result from surgery or traumatic injury, but also from peripheral neuropathies caused by diseases, viral infections, or toxic treatments. Opioids, although very effective for acute pain, do not prevent the development of CNP, and are considered as insufficient treatment. Therefore, there is high need for effective and safe non-opioid options to treat, prevent and eventually reverse CNP. A more effective approach to alleviating CNP would constitute a treatment that acts concurrently on various mechanisms involved in relieving pain symptoms and preventing or reversing chronification by enhancing both neuroprotection and neuroregeneration. We have identified and characterized GRT-X (N-[(3-fluorophenyl)-methyl]-1-(2-methoxyethyl)-4-methyl-2-oxo-(7-trifluoromethyl)-1H-quinoline-3-caboxylic acid amide), a novel drug which is able to activate both voltage-gated potassium channels of the Kv7 family and the mitochondrial translocator protein 18 kDa (TSPO). The dual mode-of-action (MoA) of GRT-X was indicated in in vitro studies and in vivo in a rat model of diabetic neuropathy. In this model, mechanical hyperalgesia was dose-dependently inhibited. After severe crush lesion of cervical spinal nerves in rats, GRT-X promoted survival, speeded up regrowth of sensory and motor neurons, and accelerated recovery of behavioral and neuronal responses to heat, cold, mechanical and electrical stimuli. These properties may reduce the likelihood of chronification of acute pain, and even potentially relieve established CNP. The absence of a conditioned place preference in rats suggests lack of abuse potential. In conclusion, GRT-X offers a promising preclinical profile with a novel dual MoA.
Collapse
Affiliation(s)
- Petra Bloms-Funke
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany.
| | - Michael Schumacher
- U1195 Inserm and University Paris-Saclay, 80, rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Song Liu
- U1195 Inserm and University Paris-Saclay, 80, rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Diya Su
- U1195 Inserm and University Paris-Saclay, 80, rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Jing Li
- U1195 Inserm and University Paris-Saclay, 80, rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Philippe Liere
- U1195 Inserm and University Paris-Saclay, 80, rue du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstraße 84, D-93053, Regensburg, Germany
| | - Caroline Nothdurfter
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstraße 84, D-93053, Regensburg, Germany
| | - Gregor Bahrenberg
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany
| | - Thomas Christoph
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany
| | - Christopher Habermann
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany
| | - Christa Kneip
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany
| | - Wolfgang Schröder
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany
| | - Thomas M Tzschentke
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany
| | - Derek Saunders
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078, Aachen, Germany
| |
Collapse
|
24
|
Nutma E, Ceyzériat K, Amor S, Tsartsalis S, Millet P, Owen DR, Papadopoulos V, Tournier BB. Cellular sources of TSPO expression in healthy and diseased brain. Eur J Nucl Med Mol Imaging 2021; 49:146-163. [PMID: 33433698 PMCID: PMC8712293 DOI: 10.1007/s00259-020-05166-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022]
Abstract
The 18 kDa translocator protein (TSPO) is a highly conserved protein located in the outer mitochondrial membrane. TSPO binding, as measured with positron emission tomography (PET), is considered an in vivo marker of neuroinflammation. Indeed, TSPO expression is altered in neurodegenerative, neuroinflammatory, and neuropsychiatric diseases. In PET studies, the TSPO signal is often viewed as a marker of microglial cell activity. However, there is little evidence in support of a microglia-specific TSPO expression. This review describes the cellular sources and functions of TSPO in animal models of disease and human studies, in health, and in central nervous system diseases. A discussion of methods of analysis and of quantification of TSPO is also presented. Overall, it appears that the alterations of TSPO binding, their cellular underpinnings, and the functional significance of such alterations depend on many factors, notably the pathology or the animal model under study, the disease stage, and the involved brain regions. Thus, further studies are needed to fully determine how changes in TSPO binding occur at the cellular level with the ultimate goal of revealing potential therapeutic pathways.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Division of Nuclear medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
- Division of Radiation Oncology, Department of Oncology, University Hospitals of Geneva, Geneva, Switzerland
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - David R Owen
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
25
|
Benz R. Historical Perspective of Pore-Forming Activity Studies of Voltage-Dependent Anion Channel (Eukaryotic or Mitochondrial Porin) Since Its Discovery in the 70th of the Last Century. Front Physiol 2021; 12:734226. [PMID: 35547863 PMCID: PMC9083909 DOI: 10.3389/fphys.2021.734226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/24/2021] [Indexed: 01/07/2023] Open
Abstract
Eukaryotic porin, also known as Voltage-Dependent Anion Channel (VDAC), is the most frequent protein in the outer membrane of mitochondria that are responsible for cellular respiration. Mitochondria are most likely descendants of strictly aerobic Gram-negative bacteria from the α-proteobacterial lineage. In accordance with the presumed ancestor, mitochondria are surrounded by two membranes. The mitochondrial outer membrane contains besides the eukaryotic porins responsible for its major permeability properties a variety of other not fully identified channels. It encloses also the TOM apparatus together with the sorting mechanism SAM, responsible for the uptake and assembly of many mitochondrial proteins that are encoded in the nucleus and synthesized in the cytoplasm at free ribosomes. The recognition and the study of electrophysiological properties of eukaryotic porin or VDAC started in the late seventies of the last century by a study of Schein et al., who reconstituted the pore from crude extracts of Paramecium mitochondria into planar lipid bilayer membranes. Whereas the literature about structure and function of eukaryotic porins was comparatively rare during the first 10years after the first study, the number of publications started to explode with the first sequencing of human Porin 31HL and the recognition of the important function of eukaryotic porins in mitochondrial metabolism. Many genomes contain more than one gene coding for homologs of eukaryotic porins. More than 100 sequences of eukaryotic porins are known to date. Although the sequence identity between them is relatively low, the polypeptide length and in particular, the electrophysiological characteristics are highly preserved. This means that all eukaryotic porins studied to date are anion selective in the open state. They are voltage-dependent and switch into cation-selective substates at voltages in the physiological relevant range. A major breakthrough was also the elucidation of the 3D structure of the eukaryotic pore, which is formed by 19 β-strands similar to those of bacterial porin channels. The function of the presumed gate an α-helical stretch of 20 amino acids allowed further studies with respect to voltage dependence and function, but its exact role in channel gating is still not fully understood.
Collapse
Affiliation(s)
- Roland Benz
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| |
Collapse
|
26
|
Troike KM, Acanda de la Rocha AM, Alban TJ, Grabowski MM, Otvos B, Cioffi G, Waite KA, Barnholtz Sloan JS, Lathia JD, Guilarte TR, Azzam DJ. The Translocator Protein ( TSPO) Genetic Polymorphism A147T Is Associated with Worse Survival in Male Glioblastoma Patients. Cancers (Basel) 2021; 13:cancers13184525. [PMID: 34572751 PMCID: PMC8471762 DOI: 10.3390/cancers13184525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary The translocator protein 18 kDa (TSPO) gene is highly expressed in glioblastoma (GBM), the most common primary malignant brain tumor, which remains one of the most difficult tumors to treat. TSPO is located in the outer mitochondrial membrane and binds cholesterol through its C-terminal domain. One frequent single-nucleotide polymorphism (SNP) rs6971, which changes the alanine 147 into threonine (Ala147Thr), has been found in the C-terminal domain of the TSPO region and dramatically alters the affinity with which TSPO binds drug ligands. However, the potential association between the TSPO genetic variants and GBM clinical outcomes is not known. Here, we evaluated the effects of the Ala147Thr SNP localized in this TSPO region on biological, sex-specific, overall, and progression-free GBM survival. Our findings suggest an association between the TSPO rs6971 variant and adverse outcomes in male GBM patients but not in females. These findings also suggest that the TSPO rs6971 SNP could be used as a prognostic marker of survival in GBM patients. Abstract Glioblastoma (GBM) is the most common primary brain tumor in adults, with few available therapies and a five-year survival rate of 7.2%. Hence, strategies for improving GBM prognosis are urgently needed. The translocator protein 18kDa (TSPO) plays crucial roles in essential mitochondria-based physiological processes and is a validated biomarker of neuroinflammation, which is implicated in GBM progression. The TSPO gene has a germline single nucleotide polymorphism, rs6971, which is the most common SNP in the Caucasian population. High TSPO gene expression is associated with reduced survival in GBM patients; however, the relation between the most frequent TSPO genetic variant and GBM pathogenesis is not known. The present study retrospectively analyzed the correlation of the TSPO polymorphic variant rs6971 with overall and progression-free survival in GBM patients using three independent cohorts. TSPO rs6971 polymorphism was significantly associated with shorter overall survival and progression-free survival in male GBM patients but not in females in one large cohort of 441 patients. We observed similar trends in two other independent cohorts. These observations suggest that the TSPO rs6971 polymorphism could be a significant predictor of poor prognosis in GBM, with a potential for use as a prognosis biomarker in GBM patients. These results reveal for the first time a biological sex-specific relation between rs6971 TSPO polymorphism and GBM.
Collapse
Affiliation(s)
- Katie M. Troike
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (K.M.T.); (T.J.A.); (M.M.G.); (B.O.); (J.D.L.)
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Arlet M. Acanda de la Rocha
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA;
| | - Tyler J. Alban
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (K.M.T.); (T.J.A.); (M.M.G.); (B.O.); (J.D.L.)
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Matthew M. Grabowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (K.M.T.); (T.J.A.); (M.M.G.); (B.O.); (J.D.L.)
- Department of Neurosurgery, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Balint Otvos
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (K.M.T.); (T.J.A.); (M.M.G.); (B.O.); (J.D.L.)
- Department of Neurosurgery, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gino Cioffi
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Trans-Divisional Research Program, Bethesda, MD 20892, USA; (G.C.); (K.A.W.); (J.S.B.S.)
| | - Kristin A. Waite
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Trans-Divisional Research Program, Bethesda, MD 20892, USA; (G.C.); (K.A.W.); (J.S.B.S.)
| | - Jill S. Barnholtz Sloan
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Trans-Divisional Research Program, Bethesda, MD 20892, USA; (G.C.); (K.A.W.); (J.S.B.S.)
- National Cancer Institute, Center for Biomedical Informatics and Information Technology, Bethesda, MD 20892, USA
| | - Justin D. Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (K.M.T.); (T.J.A.); (M.M.G.); (B.O.); (J.D.L.)
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tomás R. Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA;
- Brain, Behavior & the Environment Program, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA
- Correspondence: (T.R.G.); (D.J.A.)
| | - Diana J. Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA;
- Correspondence: (T.R.G.); (D.J.A.)
| |
Collapse
|
27
|
Wang J, Beecher K. TSPO: an emerging role in appetite for a therapeutically promising biomarker. Open Biol 2021; 11:210173. [PMID: 34343461 PMCID: PMC8331234 DOI: 10.1098/rsob.210173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
There is accumulating evidence that an obesogenic Western diet causes neuroinflammatory damage to the brain, which then promotes further appetitive behaviour. Neuroinflammation has been extensively studied by analysing the translocator protein of 18 kDa (TSPO), a protein that is upregulated in the inflamed brain following a damaging stimulus. As a result, there is a rich supply of TSPO-specific agonists, antagonists and positron emission tomography ligands. One TSPO ligand, etifoxine, is also currently used clinically for the treatment of anxiety with a minimal side-effect profile. Despite the neuroinflammatory pathogenesis of diet-induced obesity, and the translational potential of targeting TSPO, there is sparse literature characterizing the effect of TSPO on appetite. Therefore, in this review, the influence of TSPO on appetite is discussed. Three putative mechanisms for TSPO's appetite-modulatory effect are then characterized: the TSPO–allopregnanolone–GABAAR signalling axis, glucosensing in tanycytes and association with the synaptic protein RIM-BP1. We highlight that, in addition to its plethora of functions, TSPO is a regulator of appetite. This review ultimately suggests that the appetite-modulating function of TSPO should be further explored due to its potential therapeutic promise.
Collapse
Affiliation(s)
- Joshua Wang
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kate Beecher
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
28
|
Lin YC, Papadopoulos V. Neurosteroidogenic enzymes: CYP11A1 in the central nervous system. Front Neuroendocrinol 2021; 62:100925. [PMID: 34015388 DOI: 10.1016/j.yfrne.2021.100925] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/07/2021] [Accepted: 05/14/2021] [Indexed: 01/08/2023]
Abstract
Neurosteroids, steroid hormones synthesized locally in the nervous system, have important neuromodulatory and neuroprotective effects in the central nervous system. Progress in neurosteroid research has led to the successful translation of allopregnanolone into an approved therapy for postpartum depression. However, there is insufficient evidence to support the assumption that steroidogenesis is exactly the same between the nervous system and the periphery. This review focuses on CYP11A1, the only enzyme currently known to catalyze the first reaction in steroidogenesis to produce pregnenolone, the precursor to all other steroids. Although CYP11A1 mRNA has been found in brain of many mammals, the presence of CYP11A1 protein has been difficult to detect, particularly in humans. Here, we highlight the discrepancies in the current evidence for CYP11A1 in the central nervous system and propose new directions for understanding neurosteroidogenesis, which will be crucial for developing neurosteroid-based therapies for the future.
Collapse
Affiliation(s)
- Yiqi Christina Lin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
29
|
Rivière G, Jaipuria G, Andreas LB, Leonov A, Giller K, Becker S, Zweckstetter M. Membrane-embedded TSPO: an NMR view. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2021; 50:173-180. [PMID: 33354729 PMCID: PMC8071791 DOI: 10.1007/s00249-020-01487-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/19/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022]
Abstract
Translocator Protein (18 kDa) (TSPO) is a mitochondrial transmembrane protein commonly used as a biomarker for neuroinflammation and is also a potential therapeutic target in neurodegenerative diseases. Despite intensive research efforts, the function of TSPO is still largely enigmatic. Deciphering TSPO structure in the native lipid environment is essential to gain insight into its cellular activities and to design improved diagnostic and therapeutic ligands. Here, we discuss the influence of lipid composition on the structure of mammalian TSPO embedded into lipid bilayers on the basis of solid-state NMR experiments. We further highlight that cholesterol can influence both the tertiary and quaternary TSPO structure and also influence TSPO localization in mitochondria-associated endoplasmic reticulum membranes.
Collapse
Affiliation(s)
- Gwladys Rivière
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Garima Jaipuria
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Loren B Andreas
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Andrei Leonov
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Karin Giller
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Stefan Becker
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Markus Zweckstetter
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany.
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
30
|
Inflammation-Related Changes in Mood Disorders and the Immunomodulatory Role of Lithium. Int J Mol Sci 2021; 22:ijms22041532. [PMID: 33546417 PMCID: PMC7913492 DOI: 10.3390/ijms22041532] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Mood disorders are chronic, recurrent diseases characterized by changes in mood and emotions. The most common are major depressive disorder (MDD) and bipolar disorder (BD). Molecular biology studies have indicated an involvement of the immune system in the pathogenesis of mood disorders, and showed their correlation with altered levels of inflammatory markers and energy metabolism. Previous reports, including meta-analyses, also suggested the role of microglia activation in the M1 polarized macrophages, reflecting the pro-inflammatory phenotype. Lithium is an effective mood stabilizer used to treat both manic and depressive episodes in bipolar disorder, and as an augmentation of the antidepressant treatment of depression with a multidimensional mode of action. This review aims to summarize the molecular studies regarding inflammation, microglia activation and energy metabolism changes in mood disorders. We also aimed to outline the impact of lithium on these changes and discuss its immunomodulatory effect in mood disorders.
Collapse
|
31
|
Feng H, Liu Y, Zhang R, Liang Y, Sun L, Lan N, Ma B. TSPO Ligands PK11195 and Midazolam Reduce NLRP3 Inflammasome Activation and Proinflammatory Cytokine Release in BV-2 Cells. Front Cell Neurosci 2020; 14:544431. [PMID: 33362467 PMCID: PMC7759202 DOI: 10.3389/fncel.2020.544431] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation related to microglial activation plays an important role in neurodegenerative diseases. Translocator protein 18 kDa (TSPO), a biomarker of reactive gliosis, its ligands can reduce neuroinflammation and can be used to treat neurodegenerative diseases. Therefore, we explored whether TSPO ligands exert an anti-inflammatory effect by affecting the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome, thereby inhibiting the release of inflammatory cytokines in microglial cells. In the present study, BV-2 cells were exposed to lipopolysaccharide (LPS) for 6 h to induce an inflammatory response. We found that the levels of reactive oxygen species (ROS), NLRP3 inflammasome, interleukin-1β (IL-1β), and interleukin-18 (IL-18) were significantly increased. However, pretreatment with TSPO ligands inhibited BV-2 microglial and NLRP3 inflammasome activation and significantly reduced the levels of ROS, IL-1β, and IL-18. Furthermore, a combination of LPS and ATP was used to activate the NLRP3 inflammasome. Both pretreatment and post-treatment with TSPO ligand can downregulate the activation of NLRP3 inflammasome and IL-1β expression. Finally, we found that TSPO was involved in the regulation of NLRP3 inflammasome with TSPO ligands treatment in TSPO knockdown BV2 cells. Collectively, these results indicate that TSPO ligands are promising targets to control microglial reactivity and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Hao Feng
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Yongxin Liu
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Rui Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Yingxia Liang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Lina Sun
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Nannan Lan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Baoyu Ma
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| |
Collapse
|
32
|
Tuominen S, Keller T, Petruk N, López-Picón F, Eichin D, Löyttyniemi E, Verhassel A, Rajander J, Sandholm J, Tuomela J, Grönroos TJ. Evaluation of [ 18F]F-DPA as a target for TSPO in head and neck cancer under normal conditions and after radiotherapy. Eur J Nucl Med Mol Imaging 2020; 48:1312-1326. [PMID: 33340054 PMCID: PMC8113193 DOI: 10.1007/s00259-020-05115-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/09/2020] [Indexed: 11/05/2022]
Abstract
Background Many malignant tumours have increased TSPO expression, which has been related to a poor prognosis. TSPO-PET tracers have not comprehensively been evaluated in peripherally located tumours. This study aimed to evaluate whether N,N-diethyl-2-(2-(4-([18F]fluoro)phenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide ([18F]F-DPA) can reflect radiotherapy (RT)-induced changes in TSPO activity in head and neck squamous cell carcinoma (HNSCC). Methods RT was used to induce inflammatory responses in HNSCC xenografts and cells. [18F]F-DPA uptake was measured in vivo in non-irradiated and irradiated tumours, followed by ex vivo biodistribution, autoradiography, and radiometabolite analysis. In vitro studies were performed in parental and TSPO-silenced (TSPO siRNA) cells. TSPO protein and mRNA expression, as well as tumour-associated macrophages (TAMs), were also assessed. Results In vivo imaging and ex vivo measurement revealed significantly higher [18F]F-DPA uptake in irradiated, compared to non-irradiated tumours. In vitro labelling studies with cells confirmed this finding, whereas no effect of RT on [18F]F-DPA uptake was detected in TSPO siRNA cells. Radiometabolite analysis showed that the amount of unchanged [18F]F-DPA in tumours was 95%, also after irradiation. PK11195 pre-treatment reduced the tumour-to-blood ratio of [18F]F-DPA by 73% in xenografts and by 88% in cells. TSPO protein and mRNA levels increased after RT, but were highly variable. The proportion of M1/M2 TAMs decreased after RT, whereas the proportion of monocytes and migratory monocytes/macrophages increased. Conclusions [18F]F-DPA can detect changes in TSPO expression levels after RT in HNSCC, which does not seem to reflect inflammation. Further studies are however needed to clarify the physiological mechanisms regulated by TSPO after RT. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-020-05115-z.
Collapse
Affiliation(s)
- Sanni Tuominen
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland.,Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Thomas Keller
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Nataliia Petruk
- Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Francisco López-Picón
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Dominik Eichin
- MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Alejandra Verhassel
- Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Johanna Tuomela
- Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Tove J Grönroos
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland. .,MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland. .,Department of Oncology and Radiotherapy, Turku University Hospital, Hämeenkatu 11, FI-20520, Turku, Finland.
| |
Collapse
|
33
|
Lejri I, Grimm A, Hallé F, Abarghaz M, Klein C, Maitre M, Schmitt M, Bourguignon JJ, Mensah-Nyagan AG, Bihel F, Eckert A. TSPO Ligands Boost Mitochondrial Function and Pregnenolone Synthesis. J Alzheimers Dis 2020; 72:1045-1058. [PMID: 31256132 PMCID: PMC6971832 DOI: 10.3233/jad-190127] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Translocator protein 18 kDa (TSPO) is located in the mitochondrial outer membrane and plays an important role in steroidogenesis and cell survival. In the central nervous system (CNS), its expression is upregulated in neuropathologies such as Alzheimer's disease (AD). Previously, we demonstrated that two new TSPO ligands based on an imidazoquinazolinone termed 2a and 2b, stimulated pregnenolone synthesis and ATP production in vitro. In the present study, we compared their effects to those of TSPO ligands described in the literature (XBD173, SSR-180,575, and Ro5-4864) by profiling the mitochondrial bioenergetic phenotype before and after treatment and investigating the protective effects of these ligands after oxidative injury in a cellular model of AD overexpressing amyloid-β (Aβ). Of note, ATP levels increased with rising pregnenolone levels suggesting that the energetic performance of mitochondria is linked to an increased production of this neurosteroid via TSPO modulation. Our results further demonstrate that the TSPO ligands 2a and 2b exerted neuroprotective effects by improving mitochondrial respiration, reducing reactive oxygen species and thereby decreasing oxidative stress-induced cell death as well as lowering Aβ levels. The compounds 2a and 2b show similar or even better functional effects than those obtained with the reference TSPO ligands XBD173 and SSR-180.575. These findings indicate that the new TSPO ligands modulate mitochondrial bioenergetic phenotype and protect against oxidative injury probably through the de novo synthesis of neurosteroids, suggesting that these compounds could be potential new therapeutic tools for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Imane Lejri
- University of Basel, Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Basel, Switzerland.,Psychiatric University Clinics, Basel, Switzerland
| | - Amandine Grimm
- University of Basel, Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Basel, Switzerland.,Psychiatric University Clinics, Basel, Switzerland
| | - François Hallé
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS, Université de Strasbourg, Faculté de pharmacie, Illkirch, France
| | - Mustapha Abarghaz
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS, Université de Strasbourg, Faculté de pharmacie, Illkirch, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Michel Maitre
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Martine Schmitt
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS, Université de Strasbourg, Faculté de pharmacie, Illkirch, France
| | - Jean-Jacques Bourguignon
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS, Université de Strasbourg, Faculté de pharmacie, Illkirch, France
| | - Ayikoe Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Frederic Bihel
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS, Université de Strasbourg, Faculté de pharmacie, Illkirch, France
| | - Anne Eckert
- University of Basel, Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Basel, Switzerland.,Psychiatric University Clinics, Basel, Switzerland
| |
Collapse
|
34
|
Ammer LM, Vollmann-Zwerenz A, Ruf V, Wetzel CH, Riemenschneider MJ, Albert NL, Beckhove P, Hau P. The Role of Translocator Protein TSPO in Hallmarks of Glioblastoma. Cancers (Basel) 2020; 12:cancers12102973. [PMID: 33066460 PMCID: PMC7602186 DOI: 10.3390/cancers12102973] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The translocator protein (TSPO) has been under extensive investigation as a specific marker in positron emission tomography (PET) to visualize brain lesions following injury or disease. In recent years, TSPO is increasingly appreciated as a potential novel therapeutic target in cancer. In Glioblastoma (GBM), the most malignant primary brain tumor, TSPO expression levels are strongly elevated and scientific evidence accumulates, hinting at a pivotal role of TSPO in tumorigenesis and glioma progression. The aim of this review is to summarize the current literature on TSPO with respect to its role both in diagnostics and especially with regard to the critical hallmarks of cancer postulated by Hanahan and Weinberg. Overall, our review contributes to a better understanding of the functional significance of TSPO in Glioblastoma and draws attention to TSPO as a potential modulator of treatment response and thus an important factor that may influence the clinical outcome of GBM. Abstract Glioblastoma (GBM) is the most fatal primary brain cancer in adults. Despite extensive treatment, tumors inevitably recur, leading to an average survival time shorter than 1.5 years. The 18 kDa translocator protein (TSPO) is abundantly expressed throughout the body including the central nervous system. The expression of TSPO increases in states of inflammation and brain injury due to microglia activation. Not least due to its location in the outer mitochondrial membrane, TSPO has been implicated with a broad spectrum of functions. These include the regulation of proliferation, apoptosis, migration, as well as mitochondrial functions such as mitochondrial respiration and oxidative stress regulation. TSPO is frequently overexpressed in GBM. Its expression level has been positively correlated to WHO grade, glioma cell proliferation, and poor prognosis of patients. Several lines of evidence indicate that TSPO plays a functional part in glioma hallmark features such as resistance to apoptosis, invasiveness, and proliferation. This review provides a critical overview of how TSPO could regulate several aspects of tumorigenesis in GBM, particularly in the context of the hallmarks of cancer proposed by Hanahan and Weinberg in 2011.
Collapse
Affiliation(s)
- Laura-Marie Ammer
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
| | - Arabel Vollmann-Zwerenz
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
| | - Viktoria Ruf
- Center for Neuropathology and Prion Research, Ludwig Maximilians University of Munich, 81377 Munich, Germany;
| | - Christian H. Wetzel
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany;
| | | | - Nathalie L. Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, 81377 Munich, Germany;
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI) and Department Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Peter Hau
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
- Correspondence:
| |
Collapse
|
35
|
Sambri I, Massa F, Gullo F, Meneghini S, Cassina L, Carraro M, Dina G, Quattrini A, Patanella L, Carissimo A, Iuliano A, Santorelli F, Codazzi F, Grohovaz F, Bernardi P, Becchetti A, Casari G. Impaired flickering of the permeability transition pore causes SPG7 spastic paraplegia. EBioMedicine 2020; 61:103050. [PMID: 33045469 PMCID: PMC7553352 DOI: 10.1016/j.ebiom.2020.103050] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Background Mutations of the mitochondrial protein paraplegin cause hereditary spastic paraplegia type 7 (SPG7), a so-far untreatable degenerative disease of the upper motoneuron with still undefined pathomechanism. The intermittent mitochondrial permeability transition pore (mPTP) opening, called flickering, is an essential process that operates to maintain mitochondrial homeostasis by reducing intra-matrix Ca2+ and reactive oxygen species (ROS) concentration, and is critical for efficient synaptic function. Methods We use a fluorescence-based approach to measure mPTP flickering in living cells and biochemical and molecular biology techniques to dissect the pathogenic mechanism of SPG7. In the SPG7 animal model we evaluate the potential improvement of the motor defect, neuroinflammation and neurodegeneration by means of an mPTP inducer, the benzodiazepine Bz-423. Findings We demonstrate that paraplegin is required for efficient transient opening of the mPTP, that is impaired in both SPG7 patients-derived fibroblasts and primary neurons from Spg7−/− mice. We show that dysregulation of mPTP opening at the pre-synaptic terminal impairs neurotransmitter release leading to ineffective synaptic transmission. Lack of paraplegin impairs mPTP flickering by a mechanism involving increased expression and activity of sirtuin3, which promotes deacetylation of cyclophilin D, thus hampering mPTP opening. Pharmacological treatment with Bz-423, which bypasses the activity of CypD, normalizes synaptic transmission and rescues the motor impairment of the SPG7 mouse model. Interpretation mPTP targeting opens a new avenue for the potential therapy of this form of spastic paraplegia. Funding Telethon Foundation grant (TGMGCSBX16TT); Dept. of Defense, US Army, grant W81XWH-18–1–0001
Collapse
Affiliation(s)
- Irene Sambri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli-Naples, Italy
| | - Filomena Massa
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli-Naples, Italy
| | | | | | | | | | | | | | - Lorenzo Patanella
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli-Naples, Italy
| | - Annamaria Carissimo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli-Naples, Italy; Institute for Applied Mathematics 'Mauro Picone', National Research Council, Naples, Italy
| | - Antonella Iuliano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli-Naples, Italy
| | | | | | | | | | | | - Giorgio Casari
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli-Naples, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
36
|
Kikutani K, Giga H, Hosokawa K, Shime N, Aizawa H. Microglial translocator protein and stressor-related disorder. Neurochem Int 2020; 140:104855. [PMID: 32980493 DOI: 10.1016/j.neuint.2020.104855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023]
Abstract
Despite the prevalence of neuroinflammation in psychiatric disorders, molecular mechanism underlying it remains elusive. Translocator protein 18 kDa (TSPO), also known as peripheral benzodiazepine receptor, is a mitochondrial protein implicated in the synthesis of steroids in a variety of tissues. Multiple reports have shown increased expression of TSPO in the activated microglia in the CNS. Radioactive probes targeting TSPO have been developed and used for imaging assessment in neurological and psychiatric disorders to examine neuroinflammation. Recent studies revealed that the wide range of stressors ranging from psychological to physical insults induced TSPO in human, suggesting that this protein could be an important tool to explore the contribution of microglia in stressor-related disorders. In this review, we first overview the microglial activation with TSPO in a wide range of stressors in human and animal models to discuss prevalent roles of TSPO in response of CNS to stressors. With recent update of the signaling pathway revealing link connecting TSPO with neuroinflammatory effectors such as reactive oxygen species, we discuss TSPO as a therapeutic targeting tool for suppression of adverse effect of stressors on long-lasting changes in animal behaviors and activities. Targeting TSPO which mediates neuroinflammation under the stress might pave the way to develop therapeutic intervention and prophylaxis of stressor-related disorder.
Collapse
Affiliation(s)
- Kazuya Kikutani
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Hiroshi Giga
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Koji Hosokawa
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Hidenori Aizawa
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan.
| |
Collapse
|
37
|
Avallone R, Lucchi C, Puja G, Codeluppi A, Filaferro M, Vitale G, Rustichelli C, Biagini G. BV-2 Microglial Cells Respond to Rotenone Toxic Insult by Modifying Pregnenolone, 5α-Dihydroprogesterone and Pregnanolone Levels. Cells 2020; 9:E2091. [PMID: 32933155 PMCID: PMC7563827 DOI: 10.3390/cells9092091] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 01/05/2023] Open
Abstract
Neuroinflammation, whose distinctive sign is the activation of microglia, is supposed to play a key role in the development and progression of neurodegenerative diseases. The aim of this investigation was to determine levels of neurosteroids produced by resting and injured BV-2 microglial cells. BV-2 cells were exposed to increasing concentrations of rotenone to progressively reduce their viability by increasing reactive oxygen species (ROS) production. BV-2 cell viability was significantly reduced 24, 48 and 72 h after rotenone (50-1000 nM) exposure. Concomitantly, rotenone (50-100 nM) determined a dose-independent augmentation of ROS production. Then, BV-2 cells were exposed to a single, threshold dose of rotenone (75 nM) to evaluate the overtime release of neurosteroids. In particular, pregnenolone, pregnenolone sulfate, progesterone, 5α-dihydroprogesterone (5α-DHP), allopregnanolone, and pregnanolone, were quantified in the culture medium by liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis. BV-2 cells synthesized all the investigated neurosteroids and, after exposure to rotenone, 5αDHP and pregnanolone production was remarkably increased. In conclusion, we found that BV-2 cells not only synthesize several neurosteroids, but further increase this production following oxidative damage. Pregnanolone and 5α-DHP may play a role in modifying the progression of neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rossella Avallone
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Chiara Lucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| | - Giulia Puja
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Alessandro Codeluppi
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Monica Filaferro
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| | - Giovanni Vitale
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Cecilia Rustichelli
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| |
Collapse
|
38
|
Ye C, Lin L, Zhang P, Chen Y, Huang J, Lin X. The protective effect of PK11195 on D-galactose-induced amnestic mild cognitive impairment in rats. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1190. [PMID: 33241039 PMCID: PMC7576013 DOI: 10.21037/atm-20-6157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background This study aimed to investigate the preventive effect of translocator protein 18kDa (TSPO) ligand PK11195 on amnestic mild cognitive impairment (aMCI), as well as its influence on astrocytes, in order to identify effective ways to prevent aMCI. Methods Male SD rats were randomly divided into control group (n=10), aMCI group (n=10), PK11195 group (n=10), PK11195 + D-gal group (n=10). The preventive effect of PK11195 on aMCI in rats was evaluated. The cognitive function of rats in four different treatment groups was determined using the Morris water maze (MWM), as well as whole-brain pathology and immunofluorescence of rat brain tissue. Results The results of the MWM behavioral test showed that rats pre-treated with PK11195 had improved escape latency and a higher number of platform crossings compared with the aMCI model rats. PK11195 was also shown to prevent the D-galactose (D-gal)-induced senescence of pyramidal cells in the hippocampal CA1 region and to inhibit the apoptosis of astrocytes. At the same time, compared with the aMCI model rats, the TSPO in the brain tissue of rats pretreated with PK11195 had a lower distribution density. Conclusions Our results prove that PK11195 can effectively prevent D-gal-induced decline of learning and memory function as well as inhibit abnormal changes of related cells.
Collapse
Affiliation(s)
- Chen Ye
- Department of Anesthesiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Institute of Anesthesiology, Fujian Medical University, Fuzhou, China
| | - Lanying Lin
- Department of Anesthesiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Institute of Anesthesiology, Fujian Medical University, Fuzhou, China
| | - Peiling Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Institute of Anesthesiology, Fujian Medical University, Fuzhou, China
| | - Yi Chen
- Department of Anesthesiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Institute of Anesthesiology, Fujian Medical University, Fuzhou, China
| | - Jinghao Huang
- Department of Anesthesiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Institute of Anesthesiology, Fujian Medical University, Fuzhou, China
| | - Xianzhong Lin
- Department of Anesthesiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Institute of Anesthesiology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
39
|
Baburina Y, Odinokova I, Krestinina O. The Effects of PK11195 and Protoporphyrin IX Can Modulate Chronic Alcohol Intoxication in Rat Liver Mitochondria under the Opening of the Mitochondrial Permeability Transition Pore. Cells 2020; 9:cells9081774. [PMID: 32722345 PMCID: PMC7463720 DOI: 10.3390/cells9081774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Decades of active research have shown that mitochondrial dysfunction, the associated oxidative stress, impaired anti-stress defense mechanisms, and the activation of the proapoptotic signaling pathways underlie pathological changes in organs and tissues. Pathologies caused by alcohol primarily affect the liver. Alcohol abuse is the cause of many liver diseases, such as steatosis, alcoholic steatohepatitis, fibrosis, cirrhosis, and, potentially, hepatocellular cancer. In this study, the effect of chronic alcohol exposure on rat liver mitochondria was investigated. We observed an ethanol-induced increase in sensitivity to calcium, changes in the level of protein kinase Akt and GSK-3β phosphorylation, an induction of the mitochondrial permeability transition pore (mPTP), and strong alterations in the expression of mPTP regulators. Moreover, we also showed an enhanced effect of PK11195 and PPIX, on the parameters of the mPTP opening in rat liver mitochondria (RLM) isolated from ethanol-treated rats compared to the RLM from control rats. We suggest that the results of this study could help elucidate the mechanisms of chronic ethanol action on the mitochondria and contribute to the development of new therapeutic strategies for treating the effects of ethanol-related diseases.
Collapse
|
40
|
Inflammation: major denominator of obesity, Type 2 diabetes and Alzheimer's disease-like pathology? Clin Sci (Lond) 2020; 134:547-570. [PMID: 32167154 DOI: 10.1042/cs20191313] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/08/2023]
Abstract
Adipose tissue is an active metabolic organ that contributes to processes such as energy storage and utilization and to the production of a number of metabolic agents, such as adipokines, which play a role in inflammation. In this review, we try to elucidate the connections between peripheral inflammation at obesity and Type 2 diabetes and the central inflammatory process. Multiple lines of evidence highlight the importance of peripheral inflammation and its link to neuroinflammation, which can lead to neurodegenerative diseases such as dementia, Alzheimer's disease (AD) and Parkinson's disease. In addition to the accumulation of misfolded amyloid beta (Aβ) peptide and the formation of the neurofibrillary tangles of hyperphosphorylated tau protein in the brain, activated microglia and reactive astrocytes are the main indicators of AD progression. They were found close to Aβ plaques in the brains of both AD patients and rodent models of Alzheimer's disease-like pathology. Cytokines are key players in pro- and anti-inflammatory processes and are also produced by microglia and astrocytes. The interplay of seemingly unrelated pathways between the periphery and the brain could, in fact, have a common denominator, with inflammation in general being a key factor affecting neuronal processes in the brain. An increased amount of white adipose tissue throughout the body seems to be an important player in pro-inflammatory processes. Nevertheless, other important factors should be studied to elucidate the pathological processes of and the relationship among obesity, Type 2 diabetes and neurodegenerative diseases.
Collapse
|
41
|
Yao R, Pan R, Shang C, Li X, Cheng J, Xu J, Li Y. Translocator Protein 18 kDa (TSPO) Deficiency Inhibits Microglial Activation and Impairs Mitochondrial Function. Front Pharmacol 2020; 11:986. [PMID: 32695005 PMCID: PMC7339871 DOI: 10.3389/fphar.2020.00986] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
TSPO is mainly expressed in the mitochondrial outer membrane of microglia in the central nervous system, and its expression is greatly increased when microglia are activated. However, the role and mechanism of this protein in microglial activation is not well characterized. In this study, we investigated the role of TSPO in microglial activation by isolating primary microglia from TSPO knockout mice and constructing TSPO-knockdown microglial cell line. We found that TSPO deficiency significantly inhibited microglial activation induced by LPS or IL-4. Mechanistically, TSPO deficiency greatly decreased the mitochondrial membrane potential and ATP production. Moreover, an analysis of cellular energy metabolism showed that TSPO deficiency suppressed mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis, resulting in microglial overall metabolic deficits. Together, our results reveal a crucial role of TSPO in microglial activation through the regulation of mitochondrial metabolism, thus providing a potential therapeutic target for neuroinflammation-related diseases of the central nervous system.
Collapse
Affiliation(s)
- Rumeng Yao
- Department of Neuropharmacology and Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ruiyuan Pan
- The State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chao Shang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xiaoheng Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jinbo Cheng
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, China.,Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
| | - Jiangping Xu
- Department of Neuropharmacology and Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Central Laboratory, Southern Medical University, Guangzhou, China
| | - Yunfeng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
42
|
Wilton DK, Stevens B. The contribution of glial cells to Huntington's disease pathogenesis. Neurobiol Dis 2020; 143:104963. [PMID: 32593752 DOI: 10.1016/j.nbd.2020.104963] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/07/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
Glial cells play critical roles in the normal development and function of neural circuits, but in many neurodegenerative diseases, they become dysregulated and may contribute to the development of brain pathology. In Huntington's disease (HD), glial cells both lose normal functions and gain neuropathic phenotypes. In addition, cell-autonomous dysfunction elicited by mutant huntingtin (mHTT) expression in specific glial cell types is sufficient to induce both pathology and Huntington's disease-related impairments in motor and cognitive performance, suggesting that these cells may drive the development of certain aspects of Huntington's disease pathogenesis. In support of this imaging studies in pre-symptomatic HD patients and work on mouse models have suggested that glial cell dysfunction occurs at a very early stage of the disease, prior to the onset of motor and cognitive deficits. Furthermore, selectively ablating mHTT from specific glial cells or correcting for HD-induced changes in their transcriptional profile rescues some HD-related phenotypes, demonstrating the potential of targeting these cells for therapeutic intervention. Here we review emerging research focused on understanding the involvement of different glial cell types in specific aspects of HD pathogenesis. This work is providing new insight into how HD impacts biological functions of glial cells in the healthy brain as well as how HD induced dysfunction in these cells might change the way they integrate into biological circuits.
Collapse
Affiliation(s)
- Daniel K Wilton
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center, Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Wolf A, Herb M, Schramm M, Langmann T. The TSPO-NOX1 axis controls phagocyte-triggered pathological angiogenesis in the eye. Nat Commun 2020; 11:2709. [PMID: 32483169 PMCID: PMC7264151 DOI: 10.1038/s41467-020-16400-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Aberrant immune responses including reactive phagocytes are implicated in the etiology of age-related macular degeneration (AMD), a major cause of blindness in the elderly. The translocator protein (18 kDa) (TSPO) is described as a biomarker for reactive gliosis, but its biological functions in retinal diseases remain elusive. Here, we report that tamoxifen-induced conditional deletion of TSPO in resident microglia using Cx3cr1CreERT2:TSPOfl/fl mice or targeting the protein with the synthetic ligand XBD173 prevents reactivity of phagocytes in the laser-induced mouse model of neovascular AMD. Concomitantly, the subsequent neoangiogenesis and vascular leakage are prevented by TSPO knockout or XBD173 treatment. Using different NADPH oxidase-deficient mice, we show that TSPO is a key regulator of NOX1-dependent neurotoxic ROS production in the retina. These data define a distinct role for TSPO in retinal phagocyte reactivity and highlight the protein as a drug target for immunomodulatory and antioxidant therapies for AMD.
Collapse
Affiliation(s)
- Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, D-50931, Cologne, Germany
| | - Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, D-50931, Cologne, Germany
| | - Michael Schramm
- Institute for Medical Microbiology, Immunology and Hygiene, D-50931, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, D-50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, D-50931, Cologne, Germany.
| |
Collapse
|
44
|
Lee Y, Park Y, Nam H, Lee JW, Yu SW. Translocator protein (TSPO): the new story of the old protein in neuroinflammation. BMB Rep 2020. [PMID: 31818362 PMCID: PMC6999824 DOI: 10.5483/bmbrep.2020.53.1.273] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Translocator protein (TSPO), also known as peripheral benzodiazepine receptor, is a transmembrane protein located on the outer mitochondria membrane (OMM) and mainly expressed in glial cells in the brain. Because of the close correlation of its expression level with neuropathology and therapeutic efficacies of several TSPO binding ligands under many neurological conditions, TSPO has been regarded as both biomarker and therapeutic target, and the biological functions of TSPO have been a major research focus. However, recent genetic studies with animal and cellular models revealed unexpected results contrary to the anticipated biological importance of TSPO and cast doubt on the action modes of the TSPO-binding drugs. In this review, we summarize recent controversial findings on the discrepancy between pharmacological and genetic studies of TSPO and suggest some future direction to understand this old and mysterious protein.
Collapse
Affiliation(s)
- Younghwan Lee
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Youngjin Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Hyeri Nam
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Ji-Won Lee
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Seong-Woon Yu
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
45
|
18-kDa translocator protein association complexes in the brain: From structure to function. Biochem Pharmacol 2020; 177:114015. [PMID: 32387458 DOI: 10.1016/j.bcp.2020.114015] [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: 02/17/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
The outer mitochondrial membrane 18-kDa translocator protein (TSPO) is highly conserved in organisms of different species and ubiquitously expressed throughout tissues, including the nervous system. In the healthy adult brain, TSPO expression levels are low and promptly modulated under different pathological conditions, such as cancer, inflammatory states, and neurological and psychiatric disorders. Not surprisingly, several endogenous and synthetic molecules capable of binding TSPO have been proposed as drugs or diagnostic tools for brain diseases. The most studied biochemical function of TSPO is cholesterol translocation into mitochondria, which in turn affects the synthesis of steroids in the periphery and neurosteroids in the brain. In the last 30 years, roles for TSPO have also been suggested in other cellular processes, such as heme synthesis, apoptosis, autophagy, calcium signalling and reactive oxygen species production. Herein, we provide an overview of TSPO associations with different proteins, focusing particular attention on their related functions. Furthermore, recent TSPO-targeted therapeutic interventions are explored and discussed as prospect for innovative treatments in mental and brain diseases.
Collapse
|
46
|
Dimitrova-Shumkovska J, Krstanoski L, Veenman L. Diagnostic and Therapeutic Potential of TSPO Studies Regarding Neurodegenerative Diseases, Psychiatric Disorders, Alcohol Use Disorders, Traumatic Brain Injury, and Stroke: An Update. Cells 2020; 9:cells9040870. [PMID: 32252470 PMCID: PMC7226777 DOI: 10.3390/cells9040870] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation and cell death are among the common symptoms of many central nervous system diseases and injuries. Neuroinflammation and programmed cell death of the various cell types in the brain appear to be part of these disorders, and characteristic for each cell type, including neurons and glia cells. Concerning the effects of 18-kDa translocator protein (TSPO) on glial activation, as well as being associated with neuronal cell death, as a response mechanism to oxidative stress, the changes of its expression assayed with the aid of TSPO-specific positron emission tomography (PET) tracers' uptake could also offer evidence for following the pathogenesis of these disorders. This could potentially increase the number of diagnostic tests to accurately establish the stadium and development of the disease in question. Nonetheless, the differences in results regarding TSPO PET signals of first and second generations of tracers measured in patients with neurological disorders versus healthy controls indicate that we still have to understand more regarding TSPO characteristics. Expanding on investigations regarding the neuroprotective and healing effects of TSPO ligands could also contribute to a better understanding of the therapeutic potential of TSPO activity for brain damage due to brain injury and disease. Studies so far have directed attention to the effects on neurons and glia, and processes, such as death, inflammation, and regeneration. It is definitely worthwhile to drive such studies forward. From recent research it also appears that TSPO ligands, such as PK11195, Etifoxine, Emapunil, and 2-Cl-MGV-1, demonstrate the potential of targeting TSPO for treatments of brain diseases and disorders.
Collapse
Affiliation(s)
- Jasmina Dimitrova-Shumkovska
- Department of Experimental Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University Ss Cyril and Methodius, Arhimedova 3, P.O. Box 162, 1000 Skopje, Republic of North Macedonia;
- Correspondence: (J.D.-S.); (L.V.)
| | - Ljupcho Krstanoski
- Department of Experimental Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University Ss Cyril and Methodius, Arhimedova 3, P.O. Box 162, 1000 Skopje, Republic of North Macedonia;
| | - Leo Veenman
- Technion-Israel Institute of Technology, Faculty of Medicine, Rappaport Institute of Medical Research, 1 Efron Street, P.O. Box 9697, Haifa 31096, Israel
- Correspondence: (J.D.-S.); (L.V.)
| |
Collapse
|
47
|
Betlazar C, Middleton RJ, Banati R, Liu GJ. The Translocator Protein (TSPO) in Mitochondrial Bioenergetics and Immune Processes. Cells 2020; 9:cells9020512. [PMID: 32102369 PMCID: PMC7072813 DOI: 10.3390/cells9020512] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
The translocator protein (TSPO) is an outer mitochondrial membrane protein that is widely used as a biomarker of neuroinflammation, being markedly upregulated in activated microglia in a range of brain pathologies. Despite its extensive use as a target in molecular imaging studies, the exact cellular functions of this protein remain in question. The long-held view that TSPO plays a fundamental role in the translocation of cholesterol through the mitochondrial membranes, and thus, steroidogenesis, has been disputed by several groups with the advent of TSPO knockout mouse models. Instead, much evidence is emerging that TSPO plays a fundamental role in cellular bioenergetics and associated mitochondrial functions, also part of a greater role in the innate immune processes of microglia. In this review, we examine the more direct experimental literature surrounding the immunomodulatory effects of TSPO. We also review studies which highlight a more central role for TSPO in mitochondrial processes, from energy metabolism, to the propagation of inflammatory responses through reactive oxygen species (ROS) modulation. In this way, we highlight a paradigm shift in approaches to TSPO functioning.
Collapse
Affiliation(s)
- Calina Betlazar
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia
- Correspondence: (C.B.); (G-J.L.)
| | - Ryan J. Middleton
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
| | - Richard Banati
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia
| | - Guo-Jun Liu
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia
- Correspondence: (C.B.); (G-J.L.)
| |
Collapse
|
48
|
Da Pozzo E, Tremolanti C, Costa B, Giacomelli C, Milenkovic VM, Bader S, Wetzel CH, Rupprecht R, Taliani S, Da Settimo F, Martini C. Microglial Pro-Inflammatory and Anti-Inflammatory Phenotypes Are Modulated by Translocator Protein Activation. Int J Mol Sci 2019; 20:ijms20184467. [PMID: 31510070 PMCID: PMC6770267 DOI: 10.3390/ijms20184467] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
A key role of the mitochondrial Translocator Protein 18 KDa (TSPO) in neuroinflammation has been recently proposed. However, little is known about TSPO-activated pathways underlying the modulation of reactive microglia. In the present work, the TSPO activation was explored in an in vitro human primary microglia model (immortalized C20 cells) under inflammatory stimulus. Two different approaches were used with the aim to (i) pharmacologically amplify or (ii) silence, by the lentiviral short hairpin RNA, the TSPO physiological function. In the TSPO pharmacological stimulation model, the synthetic steroidogenic selective ligand XBD-173 attenuated the activation of microglia. Indeed, it reduces and increases the release of pro-inflammatory and anti-inflammatory cytokines, respectively. Such ligand-induced effects were abolished when C20 cells were treated with the steroidogenesis inhibitor aminoglutethimide. This suggests a role for neurosteroids in modulating the interleukin production. The highly steroidogenic ligand XBD-173 attenuated the neuroinflammatory response more effectively than the poorly steroidogenic ones, which suggests that the observed modulation on the cytokine release may be influenced by the levels of produced neurosteroids. In the TSPO silencing model, the reduction of TSPO caused a more inflamed phenotype with respect to scrambled cells. Similarly, during the inflammatory response, the TSPO silencing increased and reduced the release of pro-inflammatory and anti-inflammatory cytokines, respectively. In conclusion, the obtained results are in favor of a homeostatic role for TSPO in the context of dynamic balance between anti-inflammatory and pro-inflammatory mediators in the human microglia-mediated inflammatory response. Interestingly, our preliminary results propose that the TSPO expression could be stimulated by NF-κB during activation of the inflammatory response.
Collapse
Affiliation(s)
- Eleonora Da Pozzo
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.D.P.); (C.T.); (C.G.); (S.T.); (F.D.S.); (C.M.)
| | - Chiara Tremolanti
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.D.P.); (C.T.); (C.G.); (S.T.); (F.D.S.); (C.M.)
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.D.P.); (C.T.); (C.G.); (S.T.); (F.D.S.); (C.M.)
- Correspondence:
| | - Chiara Giacomelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.D.P.); (C.T.); (C.G.); (S.T.); (F.D.S.); (C.M.)
| | - Vladimir M. Milenkovic
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, 93059 Regensburg, Germany; (V.M.M.); (S.B.); (C.H.W.); (R.R.)
| | - Stefanie Bader
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, 93059 Regensburg, Germany; (V.M.M.); (S.B.); (C.H.W.); (R.R.)
| | - Christian H. Wetzel
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, 93059 Regensburg, Germany; (V.M.M.); (S.B.); (C.H.W.); (R.R.)
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, 93059 Regensburg, Germany; (V.M.M.); (S.B.); (C.H.W.); (R.R.)
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.D.P.); (C.T.); (C.G.); (S.T.); (F.D.S.); (C.M.)
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.D.P.); (C.T.); (C.G.); (S.T.); (F.D.S.); (C.M.)
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.D.P.); (C.T.); (C.G.); (S.T.); (F.D.S.); (C.M.)
| |
Collapse
|
49
|
Milenkovic VM, Slim D, Bader S, Koch V, Heinl ES, Alvarez-Carbonell D, Nothdurfter C, Rupprecht R, Wetzel CH. CRISPR-Cas9 Mediated TSPO Gene Knockout alters Respiration and Cellular Metabolism in Human Primary Microglia Cells. Int J Mol Sci 2019; 20:ijms20133359. [PMID: 31323920 PMCID: PMC6651328 DOI: 10.3390/ijms20133359] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/04/2019] [Accepted: 07/07/2019] [Indexed: 02/06/2023] Open
Abstract
The 18 kDa translocator protein (TSPO) is an evolutionary conserved cholesterol binding protein localized in the outer mitochondrial membrane. It has been implicated in the regulation of various cellular processes including oxidative stress, proliferation, apoptosis, and steroid hormone biosynthesis. Since the expression of TSPO in activated microglia is upregulated in various neuroinflammatory and neurodegenerative disorders, we set out to examine the role of TSPO in an immortalized human microglia C20 cell line. To this end, we performed a dual approach and used (i) lentiviral shRNA silencing to reduce TSPO expression, and (ii) the CRISPR/Cas9 technology to generate complete TSPO knockout microglia cell lines. Functional characterization of control and TSPO knockdown as well as knockout cells, revealed only low de novo steroidogenesis in C20 cells, which was not dependent on the level of TSPO expression or influenced by the treatment with TSPO-specific ligands. In contrast to TSPO knockdown C20 cells, which did not show altered mitochondrial function, the TSPO deficient knockout cells displayed a significantly decreased mitochondrial membrane potential and cytosolic Ca2+ levels, as well as reduced respiratory function. Performing the rescue experiment by lentiviral overexpression of TSPO in knockout cells, increased oxygen consumption and restored respiratory function. Our study provides further evidence for a significant role of TSPO in cellular and mitochondrial metabolism and demonstrates that different phenotypes of mitochondrial function are dependent on the level of TSPO expression.
Collapse
Affiliation(s)
- Vladimir M Milenkovic
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Dounia Slim
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Stefanie Bader
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Victoria Koch
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Elena-Sofia Heinl
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - David Alvarez-Carbonell
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Caroline Nothdurfter
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H Wetzel
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|