1
|
Bréhat J, Issop L, Morin D. History of Tspo deletion and induction in vivo: Phenotypic outcomes under physiological and pathological situations. Biochimie 2024; 224:80-90. [PMID: 38432291 DOI: 10.1016/j.biochi.2024.03.001] [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: 12/21/2023] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/05/2024]
Abstract
The mitochondrial translocator protein (TSPO) is an outer mitochondrial protein membrane with high affinity for cholesterol. It is expressed in most tissues but is more particularly enriched in steroidogenic tissues. TSPO is involved in various biological mechanisms and TSPO regulation has been related to several diseases. However, despite a considerable number of published studies interested in TSPO over the past forty years, the precise function of the protein remains obscure. Most of the functions attributed to TSPO have been identified using pharmacological ligands of this protein, leading to much debate about the accuracy of these findings. In addition, research on the physiological role of TSPO has been hampered by the lack of in vivo deletion models. Studies to perform genetic deletion of Tspo in animal models have long been unsuccessful, which led to the conclusions that the deletion was deleterious and the gene essential to life. During the last decades, thanks to the significant technical advances allowing genome modification, several models of animal genetically modified for TSPO have been developed. These models have modified our view regarding TSPO and profoundly improved our fundamental knowledge on this protein. However, to date, they did not allow to elucidate the precise molecular function of TSPO and numerous questions persist concerning the physiological role of TSPO and its future as a therapeutic target. This article chronologically reviews the development of deletion and induction models of TSPO.
Collapse
Affiliation(s)
- Juliette Bréhat
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Leeyah Issop
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Didier Morin
- INSERM U955-IMRB, Team Ghaleh, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France.
| |
Collapse
|
2
|
El Chemali L, Boutary S, Liu S, Liu GJ, Middleton RJ, Banati RB, Bahrenberg G, Rupprecht R, Schumacher M, Massaad-Massade L. GRT-X Stimulates Dorsal Root Ganglia Axonal Growth in Culture via TSPO and Kv7.2/3 Potassium Channel Activation. Int J Mol Sci 2024; 25:7327. [PMID: 39000434 PMCID: PMC11242890 DOI: 10.3390/ijms25137327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
GRT-X, which targets both the mitochondrial translocator protein (TSPO) and the Kv7.2/3 (KCNQ2/3) potassium channels, has been shown to efficiently promote recovery from cervical spine injury. In the present work, we investigate the role of GRT-X and its two targets in the axonal growth of dorsal root ganglion (DRG) neurons. Neurite outgrowth was quantified in DRG explant cultures prepared from wild-type C57BL6/J and TSPO-KO mice. TSPO was pharmacologically targeted with the agonist XBD173 and the Kv7 channels with the activator ICA-27243 and the inhibitor XE991. GRT-X efficiently stimulated DRG axonal growth at 4 and 8 days after its single administration. XBD173 also promoted axonal elongation, but only after 8 days and its repeated administration. In contrast, both ICA27243 and XE991 tended to decrease axonal elongation. In dissociated DRG neuron/Schwann cell co-cultures, GRT-X upregulated the expression of genes associated with axonal growth and myelination. In the TSPO-KO DRG cultures, the stimulatory effect of GRT-X on axonal growth was completely lost. However, GRT-X and XBD173 activated neuronal and Schwann cell gene expression after TSPO knockout, indicating the presence of additional targets warranting further investigation. These findings uncover a key role of the dual mode of action of GRT-X in the axonal elongation of DRG neurons.
Collapse
Affiliation(s)
- Léa El Chemali
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Suzan Boutary
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Song Liu
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation (ANSTO), Kirrawee, NSW 2232, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organisation (ANSTO), Kirrawee, NSW 2232, Australia
| | - Richard B Banati
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - Gregor Bahrenberg
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078 Aachen, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, D-93053 Regensburg, Germany
| | - Michael Schumacher
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Liliane Massaad-Massade
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| |
Collapse
|
3
|
Sidhu SK, Mishra S. A cholesterol-centric outlook on steroidogenesis. VITAMINS AND HORMONES 2023; 124:405-428. [PMID: 38408806 DOI: 10.1016/bs.vh.2023.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Cholesterol, an essential and versatile lipid, is the precursor substrate for the biosynthesis of steroid hormones, and a key structural and functional component of organelle membranes in eukaryotic cells. Consequently, the framework of steroidogenesis across main steroidogenic cell types is built around cholesterol, including its cellular uptake, mobilization from intracellular storage, and finally, its transport to the mitochondria where steroidogenesis begins. This setup, which is controlled by different trophic hormones in their respective target tissues, allows steroidogenic cells to meet their steroidogenic need of cholesterol effectively without impinging on the basic need for organelle membranes and their functions. However, our understanding of the basal steroidogenesis (i.e., independent of trophic hormone stimulation), which is a cell-intrinsic trait, remains poor. Particularly, the role that cholesterol itself plays in the regulation of steroidogenic factors and events in steroid hormone-producing cells remains largely unexplored. This is likely because of challenges in selectively targeting the steroidogenic intracellular cholesterol pool in studies. New evidence suggests that cholesterol plays a role in steroidogenesis. These new findings have created new opportunities to advance our understanding in this field. In this book chapter, we will provide a cholesterol-centric view on steroidogenesis and emphasize the importance of the interplay between cholesterol and the mitochondria in steroidogenic cells. Moreover, we will discuss a novel mitochondrial player, prohibitin-1, in this context. The overall goal is to provide a stimulating perspective on cholesterol as an important regulator of steroidogenesis (i.e., more than just a substrate for steroid hormones) and present the mitochondria as a potential cell-intrinsic factor in regulating steroidogenic cholesterol homeostasis.
Collapse
Affiliation(s)
- Simarjit Kaur Sidhu
- Department of Physiology & Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Suresh Mishra
- Department of Physiology & Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Department of Internal Medicine, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
4
|
Liere P, Liu GJ, Pianos A, Middleton RJ, Banati RB, Akwa Y. The Comprehensive Steroidome in Complete TSPO/PBR Knockout Mice under Basal Conditions. Int J Mol Sci 2023; 24:ijms24032474. [PMID: 36768796 PMCID: PMC9916858 DOI: 10.3390/ijms24032474] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
The 18 kDa translocator protein (TSPO/PBR) is a multifunctional evolutionary highly conserved outer mitochondrial membrane protein. Decades of research has reported an obligatory role of TSPO/PBR in both mitochondrial cholesterol transport and, thus, steroid production. However, the strict dependency of steroidogenesis on TSPO/PBR has remained controversial. The aim of this study was to provide insight into the steroid profile in complete C57BL/6-Tspotm1GuWu(GuwiyangWurra)-knockout male mice (TSPO-KO) under basal conditions. The steroidome in the brain, adrenal glands, testes and plasma was measured by gas chromatography coupled to tandem mass spectrometry (GC-MS/MS). We found that steroids present in wild-type (WT) mice were also detected in TSPO-KO mice, including pregnenolone (PREG), progestogens, mineralo-glucocorticosteroids and androgens. The concentrations of PREG and most metabolites were similar between genotypes, except a significant decrease in the levels of the 5α-reduced metabolites of progesterone (PROG) in adrenal glands and plasma and of the 5α-reduced metabolites of corticosterone (B) in plasma in TSPO-KO compared to WT animals, suggesting other regulatory functions for the TSPO/PBR. The expression levels of the voltage-dependent anion-selective channel (VDAC-1), CYP11A1 and 5α-reductase were not significantly different between both groups. Thus, the complete deletion of the tspo gene in male mice does not impair de novo steroidogenesis in vivo.
Collapse
Affiliation(s)
- Philippe Liere
- Disease and Hormones of the Nervous System, U1195 Inserm-Université Paris Saclay, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation (ANSTO), Kirrawee, NSW 2232, Australia
- Faculty of Medicine and Health, Medical Imaging Sciences, Brain and Mind Centre, University of Sydney, Camperdown, NSW 2006, Australia
| | - Antoine Pianos
- Disease and Hormones of the Nervous System, U1195 Inserm-Université Paris Saclay, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France
| | - Ryan J. Middleton
- Australian Nuclear Science and Technology Organisation (ANSTO), Kirrawee, NSW 2232, Australia
| | - Richard B. Banati
- Australian Nuclear Science and Technology Organisation (ANSTO), Kirrawee, NSW 2232, Australia
- Faculty of Medicine and Health, Medical Imaging Sciences, Brain and Mind Centre, University of Sydney, Camperdown, NSW 2006, Australia
| | - Yvette Akwa
- Disease and Hormones of the Nervous System, U1195 Inserm-Université Paris Saclay, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France
- Correspondence: ; Tel.: +33-(0)1-49591878
| |
Collapse
|
5
|
Deeva OA, Yarkova MA, Mokrov GV, Gudasheva TA, Seredenin SB. Dipeptide Ligands of TSPO. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
Miura S, Oyanagi E, Watanabe C, Hamada H, Aoki T, Kremenik MJ, Yano H. Acetyl-L-carnitine attenuates Poly I: C- induced sickness behavior in mice. Biosci Biotechnol Biochem 2022; 86:1423-1430. [PMID: 35945649 DOI: 10.1093/bbb/zbac132] [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: 05/06/2022] [Accepted: 07/27/2022] [Indexed: 11/14/2022]
Abstract
Fatigue is accompanied by a decrease in physical activity or malaise, and might be reduced by acetyl-L-carnitine (ALC) administration. The purpose of this study was to investigate the preventive effects of ALC on Poly I: C-induced sickness behavior in mice. For the experiment, male C3H/HeN mice were used and treated with ALC for 5 days before Poly I: C administration. ALC administration attenuated the decrease in wheel behavior activity of mice at 24 h after Poly I: C administration, and ALC treated mice quickly recovered from the sickness behavior. The gene expression of brain-derived neurotrophic factor (BDNF) in the cerebrum and hippocampus, which is associated with physical activity, was higher in the ALC-treated group. Translocator protein 18kDa (TSPO), which has cytoprotective effects, was up-regulated in the cerebrum and hippocampus, suggesting that ALC suppressed the decrease in activity induced by Poly I: C treatment through enhancement of cytoprotective effects in the brain.
Collapse
Affiliation(s)
- Suzuka Miura
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, 288 Matsushima, Kurashiki, Okayama, Japan
| | - Eri Oyanagi
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, 288 Matsushima, Kurashiki, Okayama, Japan
| | - Chihiro Watanabe
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, 288 Matsushima, Kurashiki, Okayama, Japan
| | - Hiroki Hamada
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, 288 Matsushima, Kurashiki, Okayama, Japan
| | - Takafumi Aoki
- Department of Clinical Nutrition, Kawasaki University of Medical Welfare, Kurashiki, 288 Matsushima, Kurashiki, Okayama, Japan
| | - Michael J Kremenik
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, 288 Matsushima, Kurashiki, Okayama, Japan
| | - Hiromi Yano
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, 288 Matsushima, Kurashiki, Okayama, Japan
| |
Collapse
|
7
|
Yue YY, Wang YC, Liao ZX, Hu FY, Liu QY, Dong J, Zhong M, Chen MH, Pan YM, Zhong H, Shang J. Peripheral benzodiazepine receptor TSPO needs to be reconsidered before using as a drug target for a pigmentary disorder. FASEB J 2022; 36:e22454. [PMID: 35839067 DOI: 10.1096/fj.202101746rr] [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: 11/17/2021] [Revised: 06/19/2022] [Accepted: 07/06/2022] [Indexed: 11/11/2022]
Abstract
The peripheral benzodiazepine receptor (TSPO/PBR) is highly conserved among different species but with perplexing biochemical functions. Multiple ligands of TSPO show commendable regulatory activities in lots of biological functions, such as neuro-protection, cholesterol transport, and so on. These researches support that TSPO may be a potential target for disease treatment and drug development. Previous studies have shown that its ligands benzodiazepines show a satisfactory effect on melanogenic promotion. However, the potential application of TSPO in drug development for pigmentary disorder needs further investigation. In this study, we confirmed the melanogenesis induction of TSPO ligand, Ro5-4864 in mouse melanoma cell lines, human skin tissue, and zebrafish embryos by inducing melanin synthesis and melanosome transport. Molecular genetics and pharmacological studies showed that TSPO deficiency did not affect melanin production in B16F10 cells and zebrafish embryos, nor did it affect the melanin promotion effect of Ro5-4864. Whether or not TSPO exists, the expression of lots of melanogenesis-related proteins, such as TYR, TRP-1, DCT, Mlph, and Rab27 was upregulated with the Ro5-4864 administration. These results indicated that Ro5-4864 induces melanogenesis in a TSPO-independent manner, which is inconsistent with previous research. This research is a reminder that we need to be very careful during target validation in drug development.
Collapse
Affiliation(s)
- Yun-Yun Yue
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yi-Chuan Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zi-Xian Liao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fang-Yuan Hu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiu-Yan Liu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jing Dong
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Min Zhong
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ming-Han Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu-Min Pan
- School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Hui Zhong
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jing Shang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, China.,NMPA Key Laboratory for Research and Evaluation of Cosmetics, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
8
|
Shi Y, Cui M, Ochs K, Brendel M, Strübing FL, Briel N, Eckenweber F, Zou C, Banati RB, Liu GJ, Middleton RJ, Rupprecht R, Rudolph U, Zeilhofer HU, Rammes G, Herms J, Dorostkar MM. Long-term diazepam treatment enhances microglial spine engulfment and impairs cognitive performance via the mitochondrial 18 kDa translocator protein (TSPO). Nat Neurosci 2022; 25:317-329. [PMID: 35228700 DOI: 10.1038/s41593-022-01013-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/14/2022] [Indexed: 01/08/2023]
Abstract
Benzodiazepines are widely administered drugs to treat anxiety and insomnia. In addition to tolerance development and abuse liability, their chronic use may cause cognitive impairment and increase the risk for dementia. However, the mechanism by which benzodiazepines might contribute to persistent cognitive decline remains unknown. Here we report that diazepam, a widely prescribed benzodiazepine, impairs the structural plasticity of dendritic spines, causing cognitive impairment in mice. Diazepam induces these deficits via the mitochondrial 18 kDa translocator protein (TSPO), rather than classical γ-aminobutyric acid type A receptors, which alters microglial morphology, and phagocytosis of synaptic material. Collectively, our findings demonstrate a mechanism by which TSPO ligands alter synaptic plasticity and, as a consequence, cause cognitive impairment.
Collapse
Affiliation(s)
- Yuan Shi
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Munich Medical Research School, Ludwig Maximilian University of Munich, Munich, Germany
| | - Mochen Cui
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Medical Research School, Ludwig Maximilian University of Munich, Munich, Germany
| | - Katharina Ochs
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Felix L Strübing
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Nils Briel
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Medical Research School, Ludwig Maximilian University of Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Chengyu Zou
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Richard B Banati
- Australian Nuclear Science and Technology Organisation (ANSTO), Sydney NSW, Australia.,Brain and Mind Centre, Medical Imaging Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney NSW, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation (ANSTO), Sydney NSW, Australia.,Brain and Mind Centre, Medical Imaging Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney NSW, Australia
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organisation (ANSTO), Sydney NSW, Australia
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Uwe Rudolph
- Department of Comparative Biosciences, College of Veterinary Medicine, and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Mario M Dorostkar
- Center for Neuropathology, Ludwig Maximilian University of Munich, Munich, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
9
|
Martins D, Giacomel A, Williams SCR, Turkheimer F, Dipasquale O, Veronese M. Imaging transcriptomics: Convergent cellular, transcriptomic, and molecular neuroimaging signatures in the healthy adult human brain. Cell Rep 2021; 37:110173. [PMID: 34965413 DOI: 10.1016/j.celrep.2021.110173] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/30/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
The integration of transcriptomic and neuroimaging data, "imaging transcriptomics," has recently emerged to generate hypotheses about potential biological pathways underlying regional variability in neuroimaging features. However, the validity of this approach is yet to be examined in depth. Here, we sought to bridge this gap by performing transcriptomic decoding of the regional distribution of well-known molecular markers spanning different elements of the biology of the healthy human brain. Imaging transcriptomics identifies biological and cell pathways that are consistent with the known biology of a wide range of molecular neuroimaging markers. The extent to which it can capture patterns of gene expression that align well with elements of the biology of the neuroinflammatory axis, at least in healthy controls without a proinflammatory challenge, is inconclusive. Imaging transcriptomics might constitute an interesting approach to improve our understanding of the biological pathways underlying regional variability in a wide range of neuroimaging phenotypes.
Collapse
Affiliation(s)
- Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK.
| | - Alessio Giacomel
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Steven C R Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK; Department of Information Engineering, University of Padua, Via Gradenigo, 6/b, 35131 Padova, Italy.
| | | |
Collapse
|
10
|
Abstract
It is becoming clearer that it might be a combination of different biological processes such as genetic, environmental, and psychological factors, together with immune system, stress response, brain neuroplasticity and the regulation of neurotransmitters, that leads to the development of major depressive disorder (MDD). A growing number of studies have tried to investigate the underlying mechanisms of MDD by analysing the expression levels of genes (mRNA) involved in such biological processes. In this review, I have highlighted a possible key role that gene expression might play in the treatment of MDD. This is critical because many patients do not respond to antidepressant treatment or can experience side effects, causing treatment to be interrupted. Unfortunately, selecting the best antidepressant for each individual is still largely a matter of making an informed guess.
Collapse
|
11
|
Zimmermann M, Reichert AS. Rapid metabolic and bioenergetic adaptations of astrocytes under hyperammonemia - a novel perspective on hepatic encephalopathy. Biol Chem 2021; 402:1103-1113. [PMID: 34331848 DOI: 10.1515/hsz-2021-0172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/18/2021] [Indexed: 12/17/2022]
Abstract
Hepatic encephalopathy (HE) is a well-studied, neurological syndrome caused by liver dysfunctions. Ammonia, the major toxin during HE pathogenesis, impairs many cellular processes within astrocytes. Yet, the molecular mechanisms causing HE are not fully understood. Here we will recapitulate possible underlying mechanisms with a clear focus on studies revealing a link between altered energy metabolism and HE in cellular models and in vivo. The role of the mitochondrial glutamate dehydrogenase and its role in metabolic rewiring of the TCA cycle will be discussed. We propose an updated model of ammonia-induced toxicity that may also be exploited for therapeutic strategies in the future.
Collapse
Affiliation(s)
- Marcel Zimmermann
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| |
Collapse
|
12
|
Veronese M, Tuosto M, Marques TR, Howes O, Pascual B, Yu M, Masdeu JC, Turkheimer F, Bertoldo A, Zanotti-Fregonara P. Parametric Mapping for TSPO PET Imaging with Spectral Analysis Impulsive Response Function. Mol Imaging Biol 2021; 23:560-571. [PMID: 33475944 PMCID: PMC8277653 DOI: 10.1007/s11307-020-01575-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to investigate the use of spectral analysis (SA) for voxel-wise analysis of TSPO PET imaging studies. TSPO PET quantification is methodologically complicated by the heterogeneity of TSPO expression and its cell-dependent modulation during neuroinflammatory response. Compartmental models to account for this complexity exist, but they are unreliable at the high noise typical of voxel data. On the contrary, SA is noise-robust for parametric mapping and provides useful information about tracer kinetics with a free compartmental structure. PROCEDURES SA impulse response function (IRF) calculated at 90 min after tracer injection was used as main parameter of interest in 3 independent PET imaging studies to investigate its sensitivity to (1) a TSPO genetic polymorphism (rs6971) known to affect tracer binding in a cross-sectional analysis of healthy controls scanned with [11C]PBR28 PET; (2) TSPO density with [11C]PBR28 in a competitive blocking study with a TSPO blocker, XBD173; and (3) the higher affinity of a second radiotracer for TSPO, by using data from a head-to-head comparison between [11C]PBR28 and [11C]ER176 scans. RESULTS SA-IRF produced parametric maps of visually good quality. These were sensitive to TSPO genotype (mean relative difference between high- and mixed-affinity binders = 25 %) and TSPO availability (mean signal displacement after 90 mg oral administration of XBD173 = 39 %). Regional averages of voxel-wise IRF estimates were strongly associated with regional total distribution volume (VT) estimated with a 2-tissue compartmental model with vascular compartment (Pearson's r = 0.86 ± 0.11) but less strongly with standard 2TCM-VT (Pearson's r = 0.76 ± 0.32). Finally, SA-IRF estimates for [11C]ER176 were significantly higher than [11C]PBR28 ones, consistent with the higher amount of specific binding of the former tracer. CONCLUSIONS SA-IRF can be used for voxel-wise quantification of TSPO PET data because it generates high-quality parametric maps, it is sensitive to TSPO availability and genotype, and it accounts for the complexity of TSPO tracer kinetics with no additional assumptions.
Collapse
Affiliation(s)
- Mattia Veronese
- Department of Neuroimaging, IoPPN, King's College London, London, UK.
| | - Marcello Tuosto
- Department of Information Engineering, Padova University, Padova, Italy
| | - Tiago Reis Marques
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
| | - Oliver Howes
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
- MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Belen Pascual
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Meixiang Yu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | | | - Alessandra Bertoldo
- Department of Information Engineering, Padova University, Padova, Italy
- Padova Neuroscience Centre, Padova University, Padova, Italy
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| |
Collapse
|
13
|
Frison M, Faccenda D, Abeti R, Rigon M, Strobbe D, England-Rendon BS, Cash D, Barnes K, Sadeghian M, Sajic M, Wells LA, Xia D, Giunti P, Smith K, Mortiboys H, Turkheimer FE, Campanella M. The translocator protein (TSPO) is prodromal to mitophagy loss in neurotoxicity. Mol Psychiatry 2021; 26:2721-2739. [PMID: 33664474 PMCID: PMC8505241 DOI: 10.1038/s41380-021-01050-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/13/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022]
Abstract
Dysfunctional mitochondria characterise Parkinson's Disease (PD). Uncovering etiological molecules, which harm the homeostasis of mitochondria in response to pathological cues, is therefore pivotal to inform early diagnosis and therapy in the condition, especially in its idiopathic forms. This study proposes the 18 kDa Translocator Protein (TSPO) to be one of those. Both in vitro and in vivo data show that neurotoxins, which phenotypically mimic PD, increase TSPO to enhance cellular redox-stress, susceptibility to dopamine-induced cell death, and repression of ubiquitin-dependent mitophagy. TSPO amplifies the extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) signalling, forming positive feedback, which represses the transcription factor EB (TFEB) and the controlled production of lysosomes. Finally, genetic variances in the transcriptome confirm that TSPO is required to alter the autophagy-lysosomal pathway during neurotoxicity.
Collapse
Affiliation(s)
- Michele Frison
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London, United Kingdom
- MRC Mitochondrial Biology Unit, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Danilo Faccenda
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London, United Kingdom
| | - Rosella Abeti
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square London, United Kingdom
| | - Manuel Rigon
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London, United Kingdom
- Department of Biology, University of Rome TorVergata, Via della Ricerca Scientifica, Rome, Italy
| | - Daniela Strobbe
- Department of Biology, University of Rome TorVergata, Via della Ricerca Scientifica, Rome, Italy
| | - Britannie S England-Rendon
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, Institute of Psychiatry, King's College London, Camberwell, United Kingdom
| | - Katy Barnes
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Mona Sadeghian
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Marija Sajic
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Lisa A Wells
- Imanova Limited, Centre for Imaging Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Dong Xia
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London, United Kingdom
| | - Paola Giunti
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square London, United Kingdom
| | - Kenneth Smith
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, King's College London, Camberwell, United Kingdom
| | - Michelangelo Campanella
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London, United Kingdom.
- Department of Biology, University of Rome TorVergata, Via della Ricerca Scientifica, Rome, Italy.
- University College London Consortium for Mitochondrial Research, London, United Kingdom.
| |
Collapse
|
14
|
Gene expression studies in Depression development and treatment: an overview of the underlying molecular mechanisms and biological processes to identify biomarkers. Transl Psychiatry 2021; 11:354. [PMID: 34103475 PMCID: PMC8187383 DOI: 10.1038/s41398-021-01469-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/29/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
A combination of different risk factors, such as genetic, environmental and psychological factors, together with immune system, stress response, brain neuroplasticity and the regulation of neurotransmitters, is thought to lead to the development of major depressive disorder (MDD). A growing number of studies have tried to investigate the underlying mechanisms of MDD by analysing the expression levels of genes involved in such biological processes. These studies have shown that MDD is not just a brain disorder, but also a body disorder, and this is mainly due to the interplay between the periphery and the Central Nervous System (CNS). To this purpose, most of the studies conducted so far have mainly dedicated to the analysis of the gene expression levels using postmortem brain tissue as well as peripheral blood samples of MDD patients. In this paper, we reviewed the current literature on candidate gene expression alterations and the few existing transcriptomics studies in MDD focusing on inflammation, neuroplasticity, neurotransmitters and stress-related genes. Moreover, we focused our attention on studies, which have investigated mRNA levels as biomarkers to predict therapy outcomes. This is important as many patients do not respond to antidepressant medication or could experience adverse side effects, leading to the interruption of treatment. Unfortunately, the right choice of antidepressant for each individual still remains largely a matter of taking an educated guess.
Collapse
|
15
|
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: 0.7] [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
|
16
|
Neuronal activity increases translocator protein (TSPO) levels. Mol Psychiatry 2021; 26:2025-2037. [PMID: 32398717 PMCID: PMC8440208 DOI: 10.1038/s41380-020-0745-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022]
Abstract
The mitochondrial protein, translocator protein (TSPO), is a widely used biomarker of neuroinflammation, but its non-selective cellular expression pattern implies roles beyond inflammatory processes. In the present study, we investigated whether neuronal activity modifies TSPO levels in the adult central nervous system. First, we used single-cell RNA sequencing to generate a cellular landscape of basal TSPO gene expression in the hippocampus of adult (12 weeks old) C57BL6/N mice, followed by confocal laser scanning microscopy to verify TSPO protein in neuronal and non-neuronal cell populations. We then quantified TSPO mRNA and protein levels after stimulating neuronal activity with distinct stimuli, including designer receptors exclusively activated by designer drugs (DREADDs), exposure to a novel environment and acute treatment with the psychostimulant drug, amphetamine. Single-cell RNA sequencing demonstrated a non-selective and multi-cellular gene expression pattern of TSPO at basal conditions in the adult mouse hippocampus. Confocal laser scanning microscopy confirmed that TSPO protein is present in neuronal and non-neuronal (astrocytes, microglia, vascular endothelial cells) cells of cortical (medial prefrontal cortex) and subcortical (hippocampus) brain regions. Stimulating neuronal activity through chemogenetic (DREADDs), physiological (novel environment exposure) or psychopharmacological (amphetamine treatment) approaches led to consistent increases in TSPO gene and protein levels in neurons, but not in microglia or astrocytes. Taken together, our findings show that neuronal activity has the potential to modify TSPO levels in the adult central nervous system. These findings challenge the general assumption that altered TSPO expression or binding unequivocally mirrors ongoing neuroinflammation and emphasize the need to consider non-inflammatory interpretations in some physiological or pathological contexts.
Collapse
|
17
|
Loth MK, Guariglia SR, Re DB, Perez J, de Paiva VN, Dziedzic JL, Chambers JW, Azzam DJ, Guilarte TR. A Novel Interaction of Translocator Protein 18 kDa (TSPO) with NADPH Oxidase in Microglia. Mol Neurobiol 2020; 57:4467-4487. [PMID: 32743737 PMCID: PMC7515859 DOI: 10.1007/s12035-020-02042-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
In the brain neuropil, translocator protein 18 kDa (TSPO) is a stress response protein that is upregulated in microglia and astrocytes in diverse central nervous system pathologies. TSPO is widely used as a biomarker of neuroinflammation in preclinical and clinical neuroimaging studies. However, there is a paucity of knowledge on the function(s) of TSPO in glial cells. In this study, we explored a putative interaction between TSPO and NADPH oxidase 2 (NOX2) in microglia. We found that TSPO associates with gp91phox and p22phox, the principal subunits of NOX2 in primary murine microglia. The association of TSPO with gp91phox and p22phox was observed using co-immunoprecipitation, confocal immunofluorescence imaging, and proximity ligation assay. We found that besides gp91phox and p22phox, voltage-dependent anion channel (VDAC) also co-immunoprecipitated with TSPO consistent with previous reports. When we compared lipopolysaccharide (LPS) stimulated microglia to vehicle control, we found that a lower amount of gp91phox and p22phox protein co-immunoprecipitated with TSPO suggesting a disruption of the TSPO-NOX2 subunits association. TSPO immuno-gold electron microscopy confirmed that TSPO is present in the outer mitochondrial membrane but it is also found in the endoplasmic reticulum (ER), mitochondria-associated ER membrane (MAM), and in the plasma membrane. TSPO localization at the MAM may represent a subcellular site where TSPO interacts with gp91phox and p22phox since the MAM is a point of communication between outer mitochondria membrane proteins (TSPO) and ER proteins (gp91phox and p22phox) where they mature and form the cytochrome b558 (Cytb558) heterodimer. We also found that an acute burst of reactive oxygen species (ROS) increased TSPO levels on the surface of microglia and this effect was abrogated by a ROS scavenger. These results suggest that ROS production may alter the subcellular distribution of TSPO. Collectively, our findings suggest that in microglia, TSPO is associated with the major NOX2 subunits gp91phox and p22phox. We hypothesize that this interaction may regulate Cytb558 formation and modulate NOX2 levels, ROS production, and redox homeostasis in microglia.
Collapse
Affiliation(s)
- Meredith K Loth
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Sara R Guariglia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Diane B Re
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Juan Perez
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, 33199, USA
| | - Vanessa Nunes de Paiva
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jennifer L Dziedzic
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jeremy W Chambers
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, 33199, USA
| | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, 33199, USA
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA.
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
18
|
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: 43] [Impact Index Per Article: 10.8] [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
|
19
|
Qiu ZK, Liu X, Chen Y, Wu RJ, Guan SF, Pan YY, Wang QB, Tang D, Zhu T, Chen JS. Translocator protein 18 kDa: a potential therapeutic biomarker for post traumatic stress disorder. Metab Brain Dis 2020; 35:695-707. [PMID: 32172519 DOI: 10.1007/s11011-020-00548-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 02/10/2020] [Indexed: 11/11/2022]
Abstract
Post traumatic stress disorder (PTSD) is widely regarded as a stress-related and trauma disorder. The symptoms of PTSD are characterized as a spectrum of vulnerabilities after the exposure to an extremely traumatic stressor. Considering as one of complex mental disorders, little progress has been made toward its diagnostic biomarkers, despite the involvement of PTSD has been studied. Many studies into the underlying neurobiology of PTSD implicated the dysfunction of neurosteroids biosynthesis and neuorinflammatory processes. Translocator protein 18 kDa (TSPO) has been considered as one of the promising therapeutic biomarkers for neurological stress disorders (like PTSD, depression, anxiety, et al) without the benzodiazepine-like side effects. This protein participates in the formation of neurosteroids and modulation of neuroinflammation. The review outlines current knowledge involving the role of TSPO in the neuropathology of PTSD and the anti-PTSD-like effects of TSPO ligands.
Collapse
Affiliation(s)
- Zhi-Kun Qiu
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Xu Liu
- Pharmacy Department of Medical Supplies Center of General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, People's Republic of China
| | - Yong Chen
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Rong-Jia Wu
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Shi-Feng Guan
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Yun-Yun Pan
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Qian-Bo Wang
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Dan Tang
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Tao Zhu
- Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Ji-Sheng Chen
- Pharmaceutical Department of The First Affiliated Hospital of Guangdong Pharmaceutical University, Clinical Pharmacy Department of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
20
|
Tong J, Williams B, Rusjan PM, Mizrahi R, Lacapère JJ, McCluskey T, Furukawa Y, Guttman M, Ang LC, Boileau I, Meyer JH, Kish SJ. Concentration, distribution, and influence of aging on the 18 kDa translocator protein in human brain: Implications for brain imaging studies. J Cereb Blood Flow Metab 2020; 40:1061-1076. [PMID: 31220997 PMCID: PMC7181090 DOI: 10.1177/0271678x19858003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Positron emission tomography (PET) imaging of the translocator protein (TSPO) is widely used as a biomarker of microglial activation. However, TSPO protein concentration in human brain has not been optimally quantified nor has its regional distribution been compared to TSPO binding. We determined TSPO protein concentration, change with age, and regional distribution by quantitative immunoblotting in autopsied human brain. Brain TSPO protein concentration (>0.1 ng/µg protein) was higher than those reported by in vitro binding assays by at least 2 to 70 fold. TSPO protein distributed widely in both gray and white matter regions, with distribution in major gray matter areas ranked generally similar to that of PET binding in second-generation radiotracer studies. TSPO protein concentration in frontal cortex was high at birth, declined precipitously during the first three months, and increased modestly during adulthood/senescence (10%/decade; vs. 30% for comparison astrocytic marker GFAP). As expected, TSPO protein levels were significantly increased (+114%) in degenerating putamen in multiple system atrophy, providing further circumstantial support for TSPO as a gliosis marker. Overall, findings show some similarities between TSPO protein and PET binding characteristics in the human brain but also suggest that part of the TSPO protein pool might be less available for radioligand binding.
Collapse
Affiliation(s)
- Junchao Tong
- Preclinical Imaging, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
- Junchao Tong, Preclinical Imaging, Centre
for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8,
Canada.
| | - Belinda Williams
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Addiction Imaging Research Group,
Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario,
Canada
| | - Pablo M. Rusjan
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Romina Mizrahi
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Jean-Jacques Lacapère
- Sorbonne Universités-UPMC University of
Paris 06, Département de Chimie, École Normale Supérieure-PSL Research University,
Paris, France
| | - Tina McCluskey
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Yoshiaki Furukawa
- Department of Neurology, Juntendo Tokyo
Koto Geriatric Medical Center, and Faculty of Medicine, University & Post
Graduate University of Juntendo, Tokyo, Japan
| | - Mark Guttman
- Centre for Movement Disorders, Toronto,
Ontario, Canada
| | - Lee-Cyn Ang
- Division of Neuropathology, London
Health Science Centre, University of Western Ontario, London, Ontario, Canada
| | - Isabelle Boileau
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
- Addiction Imaging Research Group,
Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario,
Canada
| | - Jeffrey H Meyer
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Stephen J Kish
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| |
Collapse
|
21
|
Microglial activation in schizophrenia: Is translocator 18 kDa protein (TSPO) the right marker? Schizophr Res 2020; 215:167-172. [PMID: 31699629 DOI: 10.1016/j.schres.2019.10.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022]
Abstract
Positron emission tomography (PET) with translocator 18 kDa protein (TSPO) radioligands has frequently been used to investigate microglial activation in schizophrenia in vivo. However, the specificity of this marker is increasingly debated. Here we show that TSPO expression is 1) not increased in postmortem brain tissue of schizophrenia patients; 2) not correlated with expression of microglial activation markers; 3) not restricted to microglia; and 4) not upregulated in ex vivo activated human primary microglia. Our data are in line with recent reports showing that TSPO expression is not increased in schizophrenia and that it is not a specific marker for activated microglia. This study emphasizes the need for further development of tracers to study the role of microglial activation in schizophrenia and other diseases.
Collapse
|
22
|
Characterisation of the ligand binding sites in the translocator protein TSPO using the chimeric bacterial-mammalian constructs. Protein Expr Purif 2019; 164:105456. [DOI: 10.1016/j.pep.2019.105456] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/18/2019] [Accepted: 07/12/2019] [Indexed: 01/18/2023]
|
23
|
Dahoun T, Calcia MA, Veronese M, Bloomfield P, Reis Marques T, Turkheimer F, Howes OD. The association of psychosocial risk factors for mental health with a brain marker altered by inflammation: A translocator protein (TSPO) PET imaging study. Brain Behav Immun 2019; 80:742-750. [PMID: 31112791 DOI: 10.1016/j.bbi.2019.05.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 12/11/2022] Open
Abstract
Psychiatric disorders associated with psychosocial risk factors, including depression and psychosis, have been shown to demonstrate increased microglia activity. Whilst preclinical studies indicate that psychosocial stress leads to increased levels of microglia in the frontal cortex, no study has yet been performed in humans. This study aimed at investigating whether psychosocial risk factors for depression and/or psychosis would be associated with alterations in a brain marker expressed by microglia, the translocator specific protein (TSPO) in humans. We used [11C]-PBR28 Positron Emission Tomography on healthy subjects exposed to childhood and adulthood psychosocial risk factors (high-risk group, N = 12) and age- and sex-matched healthy controls not exposed to childhood and adulthood psychosocial risk factors (low-risk group, N = 12). The [11C]-PBR28 volume of distribution (VT) and Distribution Volume Ratio (DVR) were measured in the total gray matter, and frontal, parietal, temporal, occipital lobes. Levels of childhood trauma, anxiety and depression were measured using respectively the Childhood Trauma Questionnaire, State-anxiety questionnaire and Beck Depression Inventory. Compared to the low-risk group, the high-risk group did not exhibit significant differences in the mean [11C]-PBR28 VT (F(1,20) = 1.619, p = 0.218) or DVR (F(1,22) = 0.952, p = 0.340) on any region. There were no significant correlations between the [11C]-PBR28 VT and DVRs in total gray matter and frontal lobe and measures of childhood trauma, anxiety and depression. Psychosocial risk factors for depression and/or psychosis are unlikely to be associated with alterations in [11C]-PBR28 binding, indicating that alterations in TSPO expression reported in these disorders is unlikely to be caused by psychosocial risk factors alone.
Collapse
Affiliation(s)
- Tarik Dahoun
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX37 JX, UK
| | - Marilia A Calcia
- Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Peter Bloomfield
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK
| | - Tiago Reis Marques
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Oliver D Howes
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK.
| |
Collapse
|
24
|
Transcriptional regulation of Translocator protein (18 kDa) (TSPO) in microglia requires Pu.1, Ap1 and Sp factors. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:1119-1133. [PMID: 30412797 DOI: 10.1016/j.bbagrm.2018.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/12/2018] [Accepted: 10/31/2018] [Indexed: 11/21/2022]
Abstract
Mitochondrial Translocator protein (18 kDa) (TSPO) is strongly expressed in reactive microglia and serves as a therapeutic target for alleviation of neuronal degeneration. However, little is known about TSPO's transcriptional regulation in microglia. The aim of this study was to identify genetic elements and transcription factors required for basal and inducible TSPO expression in microglia. Murine Tspo promoter was cloned into the pGL4.10 luciferase vector and functionally characterized in BV-2 cells. Deletion mutagenesis indicated that -845 bases upstream were sufficient to reconstitute near maximal promoter activity in BV-2. Deletion of -593 to -520 sequences, which harbour an Ap1, Ets.2 and Nkx3.1 site which also serves as a non-canonical binding site for Sp1-family transcription factors, led to a dramatic decrease in both basal and LPS induced promoter activity. Further deletion of -168 to -39 sequences, which contains four GC boxes, also led to a significant decrease in promoter activity. Targeted mutations of Ap1, Ets.2, Nkx3.1/Sp1/3/4 and the GC boxes led to significant decreases in promoter activity. ChIP-qPCR revealed that Pu.1, Ap1, Stat3, Sp1, Sp3 and Sp4 bind to the endogenous Tspo promoter. Notably, binding of these factors, with the exception of Stat3, was significantly enhanced upon LPS treatment. RNAi silencing of Pu.1, cJun, cFos, Sp1, Sp3, Sp4 and Stat3 strongly lowered Tspo promoter activity while Ap1 silencing inhibited LPS induced increase in Tspo protein levels. These findings demonstrate that consensus binding sequences for Ap1, Ets.2, distal as well as proximal Sp1/3/4 sites regulate basal and LPS induced Tspo promoter activity in microglia.
Collapse
|
25
|
Betlazar C, Harrison-Brown M, Middleton RJ, Banati R, Liu GJ. Cellular Sources and Regional Variations in the Expression of the Neuroinflammatory Marker Translocator Protein (TSPO) in the Normal Brain. Int J Mol Sci 2018; 19:ijms19092707. [PMID: 30208620 PMCID: PMC6163555 DOI: 10.3390/ijms19092707] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/09/2018] [Accepted: 09/09/2018] [Indexed: 02/07/2023] Open
Abstract
The inducible expression of the mitochondrial translocator protein 18 kDa (TSPO) by activated microglia is a prominent, regular feature of acute and chronic-progressive brain pathology. This expression is also the rationale for the continual development of new TSPO binding molecules for the diagnosis of "neuroinflammation" by molecular imaging. However, there is in the normal brain an ill-defined, low-level constitutive expression of TSPO. Taking advantage of healthy TSPO knockout mouse brain tissue to validate TSPO antibody specificity, this study uses immunohistochemistry to determine the regional distribution and cellular sources of TSPO in the normal mouse brain. Fluorescence microscopy revealed punctate TSPO immunostaining in vascular endothelial cells throughout the brain. In the olfactory nerve layers and glomeruli of the olfactory bulb, choroid plexus and ependymal layers, we confirm constitutive TSPO expression levels similar to peripheral organs, while some low TSPO expression is present in regions of known neurogenesis, as well as cerebellar Purkinje cells. The distributed-sparse expression of TSPO in endothelial mitochondria throughout the normal brain can be expected to give rise to a low baseline signal in TSPO molecular imaging studies. Finally, our study emphasises the need for valid and methodologically robust verification of the selectivity of TSPO ligands through the use of TSPO knockout tissues.
Collapse
Affiliation(s)
- Calina Betlazar
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- 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.
| | - Meredith Harrison-Brown
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- 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.
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
| | - Richard Banati
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- 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
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- 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.
| |
Collapse
|
26
|
TSPO in diverse CNS pathologies and psychiatric disease: A critical review and a way forward. Pharmacol Ther 2018; 194:44-58. [PMID: 30189290 DOI: 10.1016/j.pharmthera.2018.09.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The use of Translocator Protein 18 kDa (TSPO) as a clinical neuroimaging biomarker of brain injury and neuroinflammation has increased exponentially in the last decade. There has been a furious pace in the development of new radiotracers for TSPO positron emission tomography (PET) imaging and its use has now been extensively described in many neurological and mental disorders. This fast pace of research and the ever-increasing number of new laboratories entering the field often times lack an appreciation of the historical perspective of the field and introduce dogmatic, but unproven facts, related to the underlying neurobiology of the TSPO response to brain injury and neuroinflammation. Paradoxically, while in neurodegenerative disorders and in all types of CNS pathologies brain TSPO levels increase, a new observation in psychiatric disorders such as schizophrenia is decreased brain levels of TSPO measured by PET. The neurobiological bases for this new finding is currently not known, but rigorous experimental design using multiple experimental approaches and careful interpretation of results is critically important to provide the methodological and/or biological underpinnings to this new observation. This review provides a perspective of the early history of validating TSPO as a biomarker of brain injury and neuroinflammation and a critical analysis of controversial topics in the literature related to the cellular sources of the TSPO response. The latter is important in order to provide the correct interpretation of PET studies in neurodegenerative and psychiatric disorders. Furthermore, this review proposes some yet to be explored explanations to new findings in psychiatric disorders and new approaches to quantitatively assess the glial sources of the TSPO response in order to move the field forward.
Collapse
|
27
|
Selvaraj V, Stocco DM, Clark BJ. Current knowledge on the acute regulation of steroidogenesis. Biol Reprod 2018; 99:13-26. [PMID: 29718098 PMCID: PMC6044331 DOI: 10.1093/biolre/ioy102] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/23/2018] [Accepted: 04/26/2018] [Indexed: 12/31/2022] Open
Abstract
How rapid induction of steroid hormone biosynthesis occurs in response to trophic hormone stimulation of steroidogenic cells has been a subject of intensive investigation for approximately six decades. A key observation made very early was that acute regulation of steroid biosynthesis required swift and timely synthesis of a new protein whose role appeared to be involved in the delivery of the substrate for all steroid hormones, cholesterol, from the outer to the inner mitochondrial membrane where the process of steroidogenesis begins. It was quickly learned that this transfer of cholesterol to the inner mitochondrial membrane was the regulated and rate-limiting step in steroidogenesis. Following this observation, the quest for this putative regulator protein(s) began in earnest in the late 1950s. This review provides a history of this quest, the candidate proteins that arose over the years and facts surrounding their rise or decline. Only two have persisted-translocator protein (TSPO) and the steroidogenic acute regulatory protein (StAR). We present a detailed summary of the work that has been published for each of these two proteins, the specific data that has appeared in support of their role in cholesterol transport and steroidogenesis, and the ensuing observations that have arisen in recent years that have refuted the role of TSPO in this process. We believe that the only viable candidate that has been shown to be indispensable is the StAR protein. Lastly, we provide our view on what may be the most important questions concerning the acute regulation of steroidogenesis that need to be asked in future.
Collapse
Affiliation(s)
- Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | - Douglas M Stocco
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Barbara J Clark
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
28
|
Fukudome D, Hayes LN, Faust TE, Foss CA, Kondo MA, Lee BJ, Saito A, Kano SI, Coughlin JM, Kamiya A, Pomper MG, Sawa A, Niwa M. Translocator protein (TSPO) and stress cascades in mouse models of psychosis with inflammatory disturbances. Schizophr Res 2018; 197:492-497. [PMID: 29398205 PMCID: PMC6470041 DOI: 10.1016/j.schres.2018.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 12/12/2017] [Accepted: 01/17/2018] [Indexed: 11/28/2022]
Abstract
Changes in inflammatory cascades have been implicated in the underlying pathophysiology of psychosis. Translocator protein 18 kDa (TSPO) has been used to assess neuroinflammatory processes in psychotic disorders. Nonetheless, it is unclear whether TSPO, a mitochondrial protein, can be interpreted as a general marker for inflammation in diseases involving psychosis. To address this question, we investigated TSPO signaling in representative mouse models for psychosis with inflammatory disturbances. The maternal immune activation and cuprizone short-term exposure models show different TSPO signaling. Furthermore, we observed similarities and differences in their respective stress pathways including stress hormone signaling and oxidative stress that are functionally interconnected with the inflammatory responses. We propose that more careful studies of TSPO distribution in neuroinflammation and other stress cascades associated with psychotic symptoms will allow us to understand the biological mechanisms underlying psychosis-related behaviors.
Collapse
Affiliation(s)
- Daisuke Fukudome
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Lindsay N. Hayes
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Travis E. Faust
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Catherine A. Foss
- Russell H Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21231, USA
| | - Mari A. Kondo
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Brian J. Lee
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Atsushi Saito
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Shin-ichi Kano
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Jennifer M. Coughlin
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Atsushi Kamiya
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Martin G. Pomper
- Russell H Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21231, USA
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA.
| | - Minae Niwa
- Department of Psychiatry, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA.
| |
Collapse
|
29
|
Jaipuria G, Ukmar-Godec T, Zweckstetter M. Challenges and approaches to understand cholesterol-binding impact on membrane protein function: an NMR view. Cell Mol Life Sci 2018; 75:2137-2151. [PMID: 29520423 PMCID: PMC11105689 DOI: 10.1007/s00018-018-2789-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/16/2018] [Accepted: 02/27/2018] [Indexed: 01/27/2023]
Abstract
Experimental evidence for a direct role of lipids in determining the structure, dynamics, and function of membrane proteins leads to the term 'functional lipids'. In particular, the sterol molecule cholesterol modulates the activity of many membrane proteins. The precise nature of cholesterol-binding sites and the consequences of modulation of local membrane micro-viscosity by cholesterol, however, is often unknown. Here, we review the current knowledge of the interaction of cholesterol with transmembrane proteins, with a special focus on structural aspects of the interaction derived from nuclear magnetic resonance approaches. We highlight examples of the importance of cholesterol modulation of membrane protein function, discuss the specificity of cholesterol binding, and review the proposed binding motifs from a molecular perspective. We conclude with a short perspective on what could be future trends in research efforts targeted towards a better understanding of cholesterol/membrane protein interactions.
Collapse
Affiliation(s)
- Garima Jaipuria
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
| | - Tina Ukmar-Godec
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany.
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
30
|
Letter to the Editor re: Increased Expression of Translocator Protein (TSPO) Marks Pro-inflammatory Microglia but Does Not Predict Neurodegeneration. Mol Imaging Biol 2018; 20:352-353. [PMID: 29427254 DOI: 10.1007/s11307-018-1172-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
31
|
Notter T, Coughlin JM, Gschwind T, Weber-Stadlbauer U, Wang Y, Kassiou M, Vernon AC, Benke D, Pomper MG, Sawa A, Meyer U. Translational evaluation of translocator protein as a marker of neuroinflammation in schizophrenia. Mol Psychiatry 2018; 23:323-334. [PMID: 28093569 DOI: 10.1038/mp.2016.248] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/14/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) imaging with radiotracers that target translocator protein 18 kDa (TSPO) has become a popular approach to assess putative neuroinflammatory processes and associated microglia activation in psychotic illnesses. It remains unclear, however, whether TSPO imaging can accurately capture low-grade inflammatory processes such as those present in schizophrenia and related disorders. Therefore, we evaluated the validity of TSPO as a disease-relevant marker of inflammation using a translational approach, which combined neurodevelopmental and neurodegenerative mouse models with PET imaging in patients with recent-onset schizophrenia and matched controls. Using an infection-mediated neurodevelopmental mouse model, we show that schizophrenia-relevant behavioral abnormalities and increased inflammatory cytokine expression are associated with reduced prefrontal TSPO levels. On the other hand, TSPO was markedly upregulated in a mouse model of acute neurodegeneration and reactive gliosis, which was induced by intrahippocampal injection of kainic acid. In both models, the changes in TSPO levels were not restricted to microglia but emerged in various cell types, including microglia, astrocytes and vascular endothelial cells. Human PET imaging using the second-generation TSPO radiotracer [11C]DPA-713 revealed a strong trend towards reduced TSPO binding in the middle frontal gyrus of patients with recent-onset schizophrenia, who were previously shown to display increased levels of inflammatory cytokines in peripheral and central tissues. Together, our findings challenge the common assumption that central low-grade inflammation in schizophrenia is mirrored by increased TSPO expression or ligand binding. Our study further underscores the need to interpret altered TSPO binding in schizophrenia with caution, especially when measures of TSPO are not complemented with other markers of inflammation. Unless more selective microglial markers are available for PET imaging, quantification of cytokines and other inflammatory biomarkers, along with their molecular signaling pathways, may be more accurate in attempts to characterize inflammatory profiles in schizophrenia and other mental disorders that lack robust reactive gliosis.
Collapse
Affiliation(s)
- T Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - J M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - T Gschwind
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - U Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Y Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - M Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW, Australia
- Discipline of Medical Radiation Sciences, The University of Sydney, Sydney, NSW, Australia
| | - A C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - D Benke
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - M G Pomper
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - U Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Liang JJ, Rasmusson AM. Overview of the Molecular Steps in Steroidogenesis of the GABAergic Neurosteroids Allopregnanolone and Pregnanolone. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2018; 2:2470547018818555. [PMID: 32440589 PMCID: PMC7219929 DOI: 10.1177/2470547018818555] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/19/2018] [Indexed: 12/23/2022]
Abstract
Allopregnanolone and pregnanolone-neurosteroids synthesized from progesterone in the brain, adrenal gland, ovary and testis-have been implicated in a range of neuropsychiatric conditions including seizure disorders, post-traumatic stress disorder, major depression, post-partum depression, pre-menstrual dysphoric disorder, chronic pain, Parkinson's disease, Alzheimer's disease, neurotrauma, and stroke. Allopregnanolone and pregnanolone equipotently facilitate the effects of gamma-amino-butyric acid (GABA) at GABAA receptors, and when sulfated, antagonize N-methyl-D-aspartate receptors. They play myriad roles in neurophysiological homeostasis and adaptation to stress while exerting anxiolytic, antidepressant, anti-nociceptive, anticonvulsant, anti-inflammatory, sleep promoting, memory stabilizing, neuroprotective, pro-myelinating, and neurogenic effects. Given that these neurosteroids are synthesized de novo on demand, this review details the molecular steps involved in the biochemical conversion of cholesterol to allopregnanolone and pregnanolone within steroidogenic cells. Although much is known about the early steps in neurosteroidogenesis, less is known about transcriptional, translational, and post-translational processes in allopregnanolone- and pregnanolone-specific synthesis. Further research to elucidate these mechanisms as well as to optimize the timing and dose of interventions aimed at altering the synthesis or levels of these neurosteroids is much needed. This should include the development of novel therapeutics for the many neuropsychiatric conditions to which dysregulation of these neurosteroids contributes.
Collapse
Affiliation(s)
| | - Ann M. Rasmusson
- Boston
University School of Medicine, Boston, MA,
USA
- National Center for PTSD, Women’s Health
Science Division, Department of Veterans Affairs, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA,
USA
| |
Collapse
|
33
|
Notter T, Coughlin JM, Sawa A, Meyer U. Reconceptualization of translocator protein as a biomarker of neuroinflammation in psychiatry. Mol Psychiatry 2018; 23:36-47. [PMID: 29203847 DOI: 10.1038/mp.2017.232] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/05/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
A great deal of interest in psychiatric research is currently centered upon the pathogenic role of inflammatory processes. Positron emission tomography (PET) using radiolabeled ligands selective for the 18 kDa translocator protein (TSPO) has become the most widely used technique to assess putative neuroimmune abnormalities in vivo. Originally used to detect discrete neurotoxic damages, TSPO has generally turned into a biomarker of 'neuroinflammation' or 'microglial activation'. Psychiatric research has mostly accepted these denotations of TSPO, even if they may be inadequate and misleading under many pathological conditions. A reliable and neurobiologically meaningful diagnosis of 'neuroinflammation' or 'microglial activation' is unlikely to be achieved by the sole use of TSPO PET imaging. It is also very likely that the pathological meanings of altered TSPO binding or expression are disease-specific, and therefore, not easily generalizable across different neuropathologies or inflammatory conditions. This difficulty is intricately linked to the varying (and still ill-defined) physiological functions and cellular expression patterns of TSPO in health and disease. While altered TSPO binding or expression may indeed mirror ongoing neuroinflammatory processes in some cases, it may reflect other pathophysiological processes such as abnormalities in cell metabolism, energy production and oxidative stress in others. Hence, the increasing popularity of TSPO PET imaging has paradoxically introduced substantial uncertainty regarding the nature and meaning of neuroinflammatory processes and microglial activation in psychiatry, and likely in other neuropathological conditions as well. The ambiguity of conceiving TSPO simply as a biomarker of 'neuroinflammation' or 'microglial activation' calls for alternative interpretations and complimentary approaches. Without the latter, the ongoing scientific efforts and excitement surrounding the role of the neuroimmune system in psychiatry may not turn into therapeutic hope for affected individuals.
Collapse
Affiliation(s)
- T Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - J M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - U Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Abstract
A compelling need in the field of neurodegenerative diseases is the development and validation of biomarkers for early identification and differential diagnosis. The availability of positron emission tomography (PET) neuroimaging tools for the assessment of molecular biology and neuropathology has opened new venues in the diagnostic design and the conduction of new clinical trials. PET techniques, allowing the in vivo assessment of brain function and pathology changes, are increasingly showing great potential in supporting clinical diagnosis also in the early and even preclinical phases of dementia. This review will summarize the most recent evidence on fluorine-18 fluorodeoxyglucose-, amyloid -, tau -, and neuroinflammation - PET tools, highlighting strengths and limitations and possible new perspectives in research and clinical applications. Appropriate use of PET tools is crucial for a prompt diagnosis and target evaluation of new developed drugs aimed at slowing or preventing dementia.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Arianna Sala
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Paola Caminiti
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
35
|
Fernández Hurst N, Zanetti SR, Báez NS, Bibolini MJ, Bouzat C, Roth GA. Diazepam treatment reduces inflammatory cells and mediators in the central nervous system of rats with experimental autoimmune encephalomyelitis. J Neuroimmunol 2017; 313:145-151. [PMID: 28992974 DOI: 10.1016/j.jneuroim.2017.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/21/2017] [Accepted: 09/28/2017] [Indexed: 12/23/2022]
Abstract
Benzodiazepines are psychoactive drugs and some of them also affect immune cells. We here characterized the inflammatory and infiltrating immune cells in the central nervous system (CNS) during the acute phase of experimental autoimmune encephalomyelitis (EAE) in animals treated with Diazepam. Also, we evaluated the expression of Translocator Protein (18kDa) (TSPO), which is a biomarker of neuroinflammatory diseases. The results indicate that Diazepam exerts protective effects on EAE development, decreasing the incidence of the disease and reducing the number of inflammatory cells in CNS, with a concomitant decrease of TSPO levels in brain tissue and CNS inflammatory CD11b+ cells.
Collapse
Affiliation(s)
- Nicolás Fernández Hurst
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, CONICET-UNC), Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
| | - Samanta R Zanetti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB, CONICET-UNS), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - Natalia S Báez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI, UNCCONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
| | - Mario J Bibolini
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, CONICET-UNC), Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
| | - Cecilia Bouzat
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB, CONICET-UNS), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - German A Roth
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, CONICET-UNC), Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina.
| |
Collapse
|
36
|
Differential effects of the 18-kDa translocator protein (TSPO) ligand etifoxine on steroidogenesis in rat brain, plasma and steroidogenic glands: Pharmacodynamic studies. Psychoneuroendocrinology 2017; 83:122-134. [PMID: 28609670 DOI: 10.1016/j.psyneuen.2017.05.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/21/2017] [Accepted: 05/23/2017] [Indexed: 11/20/2022]
Abstract
Etifoxine is indicated in humans for treating anxiety. In rodents, besides its anxiolytic-like properties, it has recently shown neuroprotective and neuroregenerative activities. It acts by enhancing GABAA receptor function and by stimulating acute steroid biosynthesis via the activation of the 18-kDa translocator protein. However, the regulatory action of etifoxine on steroid production is not well characterized. In this work, we performed dose-response, acute and chronic time-course experiments on the effects of intraperitoneal injections of etifoxine on steroid levels in adult male rat brain and plasma analyzed by gas chromatography-mass spectrometry. Concentrations of pregnenolone, progesterone and its 5α-reduced metabolites were significantly increased in both tissues in response to 25 and 50mg/kg of etifoxine, as compared with vehicle controls, and reached maximal values at 0.5-1h post-injection. Daily injections of etifoxine (50mg/kg, 15days) kept them increased at day 15. Comparisons between steroidogenic tissues revealed that 1h after 50mg/kg of etifoxine treatment, levels of pregnenolone, progesterone and corticosterone were highest in adrenal glands and markedly increased together with their reduced metabolites. They were also increased by etifoxine in brain and plasma, but not in testis except for corticosterone and its metabolites. In contrast, testosterone level was significantly decreased in testis while with its 5α-reduced metabolites, it was unchanged in brain. Results demonstrate that the modulation of steroid concentrations by etifoxine is dependent on the type of steroid and on the steroidogenic organ. They further suggest that adrenal steroids upregulated by etifoxine make an important contribution to the steroids present in brain. This work provides a precise and complete view of steroids regulated by etifoxine that could be useful in therapeutic research.
Collapse
|
37
|
|
38
|
Classical and Novel TSPO Ligands for the Mitochondrial TSPO Can Modulate Nuclear Gene Expression: Implications for Mitochondrial Retrograde Signaling. Int J Mol Sci 2017; 18:ijms18040786. [PMID: 28387723 PMCID: PMC5412370 DOI: 10.3390/ijms18040786] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
It is known that knockdown of the mitochondrial 18 kDa translocator protein (TSPO) as well as TSPO ligands modulate various functions, including functions related to cancer. To study the ability of TSPO to regulate gene expression regarding such functions, we applied microarray analysis of gene expression to U118MG glioblastoma cells. Within 15 min, the classical TSPO ligand PK 11195 induced changes in expression of immediate early genes and transcription factors. These changes also included gene products that are part of the canonical pathway serving to modulate general gene expression. These changes are in accord with real-time, reverse transcriptase (RT) PCR. At the time points of 15, 30, 45, and 60 min, as well as 3 and 24 h of PK 11195 exposure, the functions associated with the changes in gene expression in these glioblastoma cells covered well known TSPO functions. These functions included cell viability, proliferation, differentiation, adhesion, migration, tumorigenesis, and angiogenesis. This was corroborated microscopically for cell migration, cell accumulation, adhesion, and neuronal differentiation. Changes in gene expression at 24 h of PK 11195 exposure were related to downregulation of tumorigenesis and upregulation of programmed cell death. In the vehicle treated as well as PK 11195 exposed cell cultures, our triple labeling showed intense TSPO labeling in the mitochondria but no TSPO signal in the cell nuclei. Thus, mitochondrial TSPO appears to be part of the mitochondria-to-nucleus signaling pathway for modulation of nuclear gene expression. The novel TSPO ligand 2-Cl-MGV-1 appeared to be very specific regarding modulation of gene expression of immediate early genes and transcription factors.
Collapse
|
39
|
Jaipuria G, Leonov A, Giller K, Vasa SK, Jaremko Ł, Jaremko M, Linser R, Becker S, Zweckstetter M. Cholesterol-mediated allosteric regulation of the mitochondrial translocator protein structure. Nat Commun 2017; 8:14893. [PMID: 28358007 PMCID: PMC5379104 DOI: 10.1038/ncomms14893] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/08/2017] [Indexed: 12/02/2022] Open
Abstract
Cholesterol is an important regulator of membrane protein function. However, the exact mechanisms involved in this process are still not fully understood. Here we study how the tertiary and quaternary structure of the mitochondrial translocator protein TSPO, which binds cholesterol with nanomolar affinity, is affected by this sterol. Residue-specific analysis of TSPO by solid-state NMR spectroscopy reveals a dynamic monomer–dimer equilibrium of TSPO in the membrane. Binding of cholesterol to TSPO's cholesterol-recognition motif leads to structural changes across the protein that shifts the dynamic equilibrium towards the translocator monomer. Consistent with an allosteric mechanism, a mutation within the oligomerization interface perturbs transmembrane regions located up to 35 Å away from the interface, reaching TSPO's cholesterol-binding motif. The lower structural stability of the intervening transmembrane regions provides a mechanistic basis for signal transmission. Our study thus reveals an allosteric signal pathway that connects membrane protein tertiary and quaternary structure with cholesterol binding. The outer mitochondrial membrane translocator protein (TSPO) mediates several mitochondrial functions and binds cholesterol with a high affinity. Here the authors use solid-state NMR to show that cholesterol binding to TSPO results in allosteric changes that modulate TSPO oligomerization.
Collapse
Affiliation(s)
- Garima Jaipuria
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Strasse 3a, 37075 Göttingen, Germany
| | - Andrei Leonov
- Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany
| | - Karin Giller
- Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany
| | - Suresh Kumar Vasa
- Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany
| | - Łukasz Jaremko
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Strasse 3a, 37075 Göttingen, Germany
| | - Mariusz Jaremko
- Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany
| | - Rasmus Linser
- Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany
| | - Stefan Becker
- Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany
| | - Markus Zweckstetter
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Strasse 3a, 37075 Göttingen, Germany.,Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany.,Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073 Göttingen, Germany
| |
Collapse
|
40
|
Liu GJ, Middleton RJ, Banati RB. Subcellular distribution of the 18kDa translocator protein and transcript variant PBR-S in human cells. Gene 2017; 613:45-56. [PMID: 28263860 DOI: 10.1016/j.gene.2017.02.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/22/2017] [Accepted: 02/28/2017] [Indexed: 10/20/2022]
Abstract
Despite continued interest in the 18kDa translocator protein (PBR/TSPO) as a biomarker and a therapeutic target for a range of diseases, its functional role, such as in the steroid synthesis pathway and energy metabolism has either become contentious or remains to be described more precisely. The PBR/TSPO gene consists of four exons, while a shorter isoform termed PBR-S lacks exon 2. The PBR-S 102-codon open reading frame differs to that of PBR/TSPO, resulting in a protein that is completely unrelated to PBR/TSPO. To our knowledge, PBR-S protein has never been described and has no known or proposed function. To obtain possible clues on the role of this uncharacterised protein, we compared the subcellular distribution of PBR-S to that of PBR/TSPO. By expressing fluorescently tagged PBR/TSPO, we confirmed that full-length PBR/TSPO co-localises with mitochondria in HeLa, HEK-293, MDA-MB-231, BJ and U87-MG human cell lines. Unlike the strictly mitochondrial localisation of PBR/TSPO, PBR-S has a punctate distribution throughout the cytosol that co-localises with lysosomes in HeLa, HEK-293, MDA-MB-231, BJ and U87-MG cells. In summary, within the cell lines examined we confirm mitochondria rather than occasionally reported other localisations, such as the cell nucleus, to be the only site where PBR/TSPO resides. Due to the lack of any shared protein sequences and the different subcellular locations, we suggest that the previously uncharacterised PBR-S protein variant of the PBR/TSPO gene is likely to serve a different yet to be discovered function compared to PBR/TSPO.
Collapse
Affiliation(s)
- Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW 2234, Australia; National Imaging Facility, Brain and Mind Centre and Faculty of Health Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW 2234, Australia
| | - Richard B Banati
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW 2234, Australia; National Imaging Facility, Brain and Mind Centre and Faculty of Health Sciences, University of Sydney, Camperdown, NSW 2050, Australia.
| |
Collapse
|
41
|
Liu GJ, Middleton RJ, Kam WWY, Chin DY, Hatty CR, Chan RHY, Banati RB. Functional gains in energy and cell metabolism after TSPO gene insertion. Cell Cycle 2017; 16:436-447. [PMID: 28103132 PMCID: PMC5351937 DOI: 10.1080/15384101.2017.1281477] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent loss-of-function studies in tissue-specific as well as global Tspo (Translocator Protein 18 kDa) knockout mice have not confirmed its long assumed indispensability for the translocation of cholesterol across the mitochondrial inter-membrane space, a rate-limiting step in steroid biosynthesis. Instead, recent studies in global Tspo knockout mice indicate that TSPO may play a more fundamental role in cellular bioenergetics, which may include the indirect down-stream regulation of transport or metabolic functions. To examine whether overexpression of the TSPO protein alters the cellular bioenergetic profile, Jurkat cells with low to absent endogenous expression were transfected with a TSPO construct to create a stable cell line with de novo expression of exogenous TSPO protein. Expression of TSPO was confirmed by RT-qPCR, radioligand binding with [3H]PK11195 and immunocytochemistry with a TSPO antibody. We demonstrate that TSPO gene insertion causes increased transcription of genes involved in the mitochondrial electron transport chain. Furthermore, TSPO insertion increased mitochondrial ATP production as well as cell excitability, reflected in a decrease in patch clamp recorded rectified K channel currents. These functional changes were accompanied by an increase in cell proliferation and motility, which were inhibited by PK11195, a selective ligand for TSPO. We suggest that TSPO may serve a range of functions that can be viewed as downstream regulatory effects of its primary, evolutionary conserved role in cell metabolism and energy production.
Collapse
Affiliation(s)
- Guo-Jun Liu
- a Australian Nuclear Science and Technology Organisation , Lucas Heights , NSW , Australia.,b Faculty of Health Science and Brain and Mind Centre, University of Sydney , NSW , Australia
| | - Ryan J Middleton
- a Australian Nuclear Science and Technology Organisation , Lucas Heights , NSW , Australia
| | - Winnie Wai-Ying Kam
- a Australian Nuclear Science and Technology Organisation , Lucas Heights , NSW , Australia.,c Department of Health Technology and Informatics , Hong Kong Polytechnic University , Hung Hom, Hong Kong , China
| | - David Y Chin
- d NCRIS Biologics Facility, Australian Institute for Bioengineering and Nanotechnology, University of Queensland , QLD , Australia
| | - Claire R Hatty
- a Australian Nuclear Science and Technology Organisation , Lucas Heights , NSW , Australia.,b Faculty of Health Science and Brain and Mind Centre, University of Sydney , NSW , Australia
| | - Ronald H Y Chan
- a Australian Nuclear Science and Technology Organisation , Lucas Heights , NSW , Australia.,b Faculty of Health Science and Brain and Mind Centre, University of Sydney , NSW , Australia
| | - Richard B Banati
- a Australian Nuclear Science and Technology Organisation , Lucas Heights , NSW , Australia.,b Faculty of Health Science and Brain and Mind Centre, University of Sydney , NSW , Australia
| |
Collapse
|
42
|
Miller WL. Disorders in the initial steps of steroid hormone synthesis. J Steroid Biochem Mol Biol 2017; 165:18-37. [PMID: 26960203 DOI: 10.1016/j.jsbmb.2016.03.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/01/2016] [Accepted: 03/03/2016] [Indexed: 12/29/2022]
Abstract
Steroidogenesis begins with cellular internalization of low-density lipoprotein particles and subsequent intracellular processing of cholesterol. Disorders in these steps include Adrenoleukodystrophy, Wolman Disease and its milder variant Cholesterol Ester Storage Disease, and Niemann-Pick Type C Disease, all of which may present with adrenal insufficiency. The means by which cholesterol is directed to steroidogenic mitochondria remains incompletely understood. Once cholesterol reaches the outer mitochondrial membrane, its delivery to the inner mitochondrial membrane is regulated by the steroidogenic acute regulatory protein (StAR). Severe StAR mutations cause classic congenital lipoid adrenal hyperplasia, characterized by lipid accumulation in the adrenal, adrenal insufficiency, and disordered sexual development in 46,XY individuals. The lipoid CAH phenotype, including spontaneous puberty in 46,XX females, is explained by a two-hit model. StAR mutations that retain partial function cause a milder, non-classic disease characterized by glucocorticoid deficiency, with lesser disorders of mineralocorticoid and sex steroid synthesis. Once inside the mitochondria, cholesterol is converted to pregnenolone by the cholesterol side-chain cleavage enzyme, P450scc, encoded by the CYP11A1 gene. Rare patients with mutations of P450scc are clinically and hormonally indistinguishable from those with lipoid CAH, and may also present as milder non-classic disease. Patients with P450scc defects do not have the massive adrenal hyperplasia that characterizes lipoid CAH, but adrenal imaging may occasionally fail to distinguish these, necessitating DNA sequencing.
Collapse
Affiliation(s)
- Walter L Miller
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143-0556, United States.
| |
Collapse
|
43
|
Busch AWU, WareJoncas Z, Montgomery BL. Tryptophan-Rich Sensory Protein/Translocator Protein (TSPO) from Cyanobacterium Fremyella diplosiphon Binds a Broad Range of Functionally Relevant Tetrapyrroles. Biochemistry 2016; 56:73-84. [PMID: 27990801 DOI: 10.1021/acs.biochem.6b01019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tryptophan-rich sensory protein/translocator protein (TSPO) is a membrane protein involved in stress adaptation in the cyanobacterium Fremyella diplosiphon. Characterized mammalian and proteobacterial TSPO homologues bind tetrapyrroles and cholesterol ligands. We investigated the ligand binding properties of TSPO from F. diplosiphon (FdTSPO1), which was functionally characterized in prior genetic studies. Two additional TSPO proteins (FdTSPO2 and FdTSPO3) are present in F. diplosiphon; they are similar in size to reported bacterial TSPOs and smaller than FdTSPO1. The longer cyanobacterial TSPO1 is found almost exclusively in filamentous cyanobacteria and has a relatively low degree of homology to bacterial and mammalian TSPO homologues with confirmed tetrapyrrole binding. To probe distinctions of long-form TSPOs, we tested the binding of porphyrin and bilin to FdTSPO1 and measured binding affinities in the low micromolar range, with the highest binding affinity detected for heme. Although tetrapyrrole ligands bound FdTSPO1 with affinities similar to those previously reported for proteobacterial TSPO, binding of cholesterol to FdTSPO1 was particularly poor and was not improved by introducing an amino acid motif known to enhance cholesterol binding in other bacterial TSPO homologues. Additionally, we detected limited binding of bacterial hopanoids to FdTSPO1. Cyanobacterial TSPO1 from the oxygenic photosynthetic F. diplosiphon, thus, binds a range of tetrapyrroles of functional relevance with efficiencies similar to those of mammalian and proteobacterial homologues, but the level of cholesterol binding is greatly reduced compared to that of mammalian TSPO. Furthermore, the ΔFdTSPO1 mutant exhibits altered growth in the presence of biliverdin compared to that of wild-type cells under green light. Together, these results suggest that TSPO molecules may play roles in bilin homeostasis or trafficking in cyanobacteria.
Collapse
Affiliation(s)
- Andrea W U Busch
- Plant Research Laboratory, Department of Energy, Michigan State University , East Lansing, Michigan 48824, United States
| | - Zachary WareJoncas
- Plant Research Laboratory, Department of Energy, Michigan State University , East Lansing, Michigan 48824, United States
| | - Beronda L Montgomery
- Plant Research Laboratory, Department of Energy, Michigan State University , East Lansing, Michigan 48824, United States.,Department of Biochemistry and Molecular Biology, Michigan State University , East Lansing, Michigan 48824, United States.,Department of Microbiology & Molecular Genetics, Michigan State University , East Lansing, Michigan 48824, United States
| |
Collapse
|
44
|
Tetrapyrroles as Endogenous TSPO Ligands in Eukaryotes and Prokaryotes: Comparisons with Synthetic Ligands. Int J Mol Sci 2016; 17:ijms17060880. [PMID: 27271616 PMCID: PMC4926414 DOI: 10.3390/ijms17060880] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 05/17/2016] [Accepted: 05/19/2016] [Indexed: 12/26/2022] Open
Abstract
The 18 kDa translocator protein (TSPO) is highly 0conserved in eukaryotes and prokaryotes. Since its discovery in 1977, numerous studies established the TSPO’s importance for life essential functions. For these studies, synthetic TSPO ligands typically are applied. Tetrapyrroles present endogenous ligands for the TSPO. Tetrapyrroles are also evolutionarily conserved and regulate multiple functions. TSPO and tetrapyrroles regulate each other. In animals TSPO-tetrapyrrole interactions range from effects on embryonic development to metabolism, programmed cell death, response to stress, injury and disease, and even to life span extension. In animals TSPOs are primarily located in mitochondria. In plants TSPOs are also present in plastids, the nuclear fraction, the endoplasmic reticulum, and Golgi stacks. This may contribute to translocation of tetrapyrrole intermediates across organelles’ membranes. As in animals, plant TSPO binds heme and protoporphyrin IX. TSPO-tetrapyrrole interactions in plants appear to relate to development as well as stress conditions, including salt tolerance, abscisic acid-induced stress, reactive oxygen species homeostasis, and finally cell death regulation. In bacteria, TSPO is important for switching from aerobic to anaerobic metabolism, including the regulation of photosynthesis. As in mitochondria, in bacteria TSPO is located in the outer membrane. TSPO-tetrapyrrole interactions may be part of the establishment of the bacterial-eukaryote relationships, i.e., mitochondrial-eukaryote and plastid-plant endosymbiotic relationships.
Collapse
|
45
|
Li F, Liu J, Liu N, Kuhn LA, Garavito RM, Ferguson-Miller S. Translocator Protein 18 kDa (TSPO): An Old Protein with New Functions? Biochemistry 2016; 55:2821-31. [PMID: 27074410 DOI: 10.1021/acs.biochem.6b00142] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Translocator protein 18 kDa (TSPO) was previously known as the peripheral benzodiazepine receptor (PBR) in eukaryotes, where it is mainly localized to the mitochondrial outer membrane. Considerable evidence indicates that it plays regulatory roles in steroidogenesis and apoptosis and is involved in various human diseases, such as metastatic cancer, Alzheimer's and Parkinson's disease, inflammation, and anxiety disorders. Ligands of TSPO are widely used as diagnostic tools and treatment options, despite there being no clear understanding of the function of TSPO. An ortholog in the photosynthetic bacterium Rhodobacter was independently discovered as the tryptophan-rich sensory protein (TspO) and found to play a role in the response to changes in oxygen and light conditions that regulate photosynthesis and respiration. As part of this highly conserved protein family found in all three kingdoms, the rat TSPO is able to rescue the knockout phenotype in Rhodobacter, indicating functional as well as structural conservation. Recently, a major breakthrough in the field was achieved: the determination of atomic-resolution structures of TSPO from different species by several independent groups. This now allows us to reexamine the function of TSPO with a molecular perspective. In this review, we focus on recently determined structures of TSPO and their implications for potential functions of this ubiquitous multifaceted protein. We suggest that TSPO is an ancient bacterial receptor/stress sensor that has developed additional interactions, partners, and roles in its mitochondrial outer membrane environment in eukaryotes.
Collapse
Affiliation(s)
- Fei Li
- Department of Biochemistry and Molecular Biology, Michigan State University , East Lansing, Michigan 48824, United States
| | - Jian Liu
- Department of Biochemistry and Molecular Biology, Michigan State University , East Lansing, Michigan 48824, United States
| | - Nan Liu
- Department of Biochemistry and Molecular Biology, Michigan State University , East Lansing, Michigan 48824, United States.,Department of Computer Science and Engineering, Michigan State University , East Lansing, Michigan 48824-1319, United States.,Department of Chemistry, Michigan State University , East Lansing, Michigan 48824-1319, United States
| | - Leslie A Kuhn
- Department of Biochemistry and Molecular Biology, Michigan State University , East Lansing, Michigan 48824, United States.,Department of Computer Science and Engineering, Michigan State University , East Lansing, Michigan 48824-1319, United States
| | - R Michael Garavito
- Department of Biochemistry and Molecular Biology, Michigan State University , East Lansing, Michigan 48824, United States
| | - Shelagh Ferguson-Miller
- Department of Biochemistry and Molecular Biology, Michigan State University , East Lansing, Michigan 48824, United States
| |
Collapse
|
46
|
Ge L, Villinger S, Mari SA, Giller K, Griesinger C, Becker S, Müller DJ, Zweckstetter M. Molecular Plasticity of the Human Voltage-Dependent Anion Channel Embedded Into a Membrane. Structure 2016; 24:585-594. [PMID: 27021164 PMCID: PMC5654509 DOI: 10.1016/j.str.2016.02.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/12/2016] [Accepted: 02/22/2016] [Indexed: 12/28/2022]
Abstract
The voltage-dependent anion channel (VDAC) regulates the flux of metabolites and ions across the outer mitochondrial membrane. Regulation of ion flow involves conformational transitions in VDAC, but the nature of these changes has not been resolved to date. By combining single-molecule force spectroscopy with nuclear magnetic resonance spectroscopy we show that the β barrel of human VDAC embedded into a membrane is highly flexible. Its mechanical flexibility exceeds by up to one order of magnitude that determined for β strands of other membrane proteins and is largest in the N-terminal part of the β barrel. Interaction with Ca(2+), a key regulator of metabolism and apoptosis, considerably decreases the barrel's conformational variability and kinetic free energy in the membrane. The combined data suggest that physiological VDAC function depends on the molecular plasticity of its channel.
Collapse
Affiliation(s)
- Lin Ge
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Saskia Villinger
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Stefania A Mari
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Karin Giller
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Christian Griesinger
- 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
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zürich, Mattenstrasse 26, 4058 Basel, Switzerland.
| | - Markus Zweckstetter
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany; Structural Biology in Dementia, German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Strasse 3a, 37075 Göttingen, Germany; Department of Neurology, University Medical Center Göttingen, University of Göttingen, Am Waldweg 33, 37073 Göttingen, Germany.
| |
Collapse
|
47
|
Porcu P, Barron AM, Frye CA, Walf AA, Yang SY, He XY, Morrow AL, Panzica GC, Melcangi RC. Neurosteroidogenesis Today: Novel Targets for Neuroactive Steroid Synthesis and Action and Their Relevance for Translational Research. J Neuroendocrinol 2016; 28:12351. [PMID: 26681259 PMCID: PMC4769676 DOI: 10.1111/jne.12351] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 12/12/2015] [Accepted: 12/12/2015] [Indexed: 12/19/2022]
Abstract
Neuroactive steroids are endogenous neuromodulators synthesised in the brain that rapidly alter neuronal excitability by binding to membrane receptors, in addition to the regulation of gene expression via intracellular steroid receptors. Neuroactive steroids induce potent anxiolytic, antidepressant, anticonvulsant, sedative, analgesic and amnesic effects, mainly through interaction with the GABAA receptor. They also exert neuroprotective, neurotrophic and antiapoptotic effects in several animal models of neurodegenerative diseases. Neuroactive steroids regulate many physiological functions, such as the stress response, puberty, the ovarian cycle, pregnancy and reward. Their levels are altered in several neuropsychiatric and neurological diseases and both preclinical and clinical studies emphasise a therapeutic potential of neuroactive steroids for these diseases, whereby symptomatology ameliorates upon restoration of neuroactive steroid concentrations. However, direct administration of neuroactive steroids has several challenges, including pharmacokinetics, low bioavailability, addiction potential, safety and tolerability, which limit its therapeutic use. Therefore, modulation of neurosteroidogenesis to restore the altered endogenous neuroactive steroid tone may represent a better therapeutic approach. This review summarises recent approaches that target the neuroactive steroid biosynthetic pathway at different levels aiming to promote neurosteroidogenesis. These include modulation of neurosteroidogenesis through ligands of the translocator protein 18 kDa and the pregnane xenobiotic receptor, as well as targeting of specific neurosteroidogenic enzymes such as 17β-hydroxysteroid dehydrogenase type 10 or P450 side chain cleavage. Enhanced neurosteroidogenesis through these targets may be beneficial not only for neurodegenerative diseases, such as Alzheimer's disease and age-related dementia, but also for neuropsychiatric diseases, including alcohol use disorders.
Collapse
Affiliation(s)
- Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Anna M. Barron
- Molecular Imaging Center, National Institute of Radiological Sciences, Anagawa, Inage-ku, Chiba, Japan
| | - Cheryl Anne Frye
- Institute of Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, AK, USA
- The University at Albany, Albany, NY, USA
| | - Alicia A. Walf
- Institute of Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, AK, USA
- The University at Albany, Albany, NY, USA
- Department of Cognitive Science, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Song-Yu Yang
- Department of Developmental Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Xue-Ying He
- Department of Developmental Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - A. Leslie Morrow
- Departments of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Gian Carlo Panzica
- Department of Neuroscience, University of Turin, and NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Roberto C. Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
48
|
Insight into the Molecular Imaging of Alzheimer's Disease. Int J Biomed Imaging 2016; 2016:7462014. [PMID: 26880871 PMCID: PMC4736963 DOI: 10.1155/2016/7462014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/16/2015] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease is a complex neurodegenerative disease affecting millions of individuals worldwide. Earlier it was diagnosed only via clinical assessments and confirmed by postmortem brain histopathology. The development of validated biomarkers for Alzheimer's disease has given impetus to improve diagnostics and accelerate the development of new therapies. Functional imaging like positron emission tomography (PET), single photon emission computed tomography (SPECT), functional magnetic resonance imaging (fMRI), and proton magnetic resonance spectroscopy provides a means of detecting and characterising the regional changes in brain blood flow, metabolism, and receptor binding sites that are associated with Alzheimer's disease. Multimodal neuroimaging techniques have indicated changes in brain structure and metabolic activity, and an array of neurochemical variations that are associated with neurodegenerative diseases. Radiotracer-based PET and SPECT potentially provide sensitive, accurate methods for the early detection of disease. This paper presents a review of neuroimaging modalities like PET, SPECT, and selected imaging biomarkers/tracers used for the early diagnosis of AD. Neuroimaging with such biomarkers and tracers could achieve a much higher diagnostic accuracy for AD and related disorders in the future.
Collapse
|
49
|
Busch AWU, Montgomery BL. The Tryptophan-Rich Sensory Protein (TSPO) is Involved in Stress-Related and Light-Dependent Processes in the Cyanobacterium Fremyella diplosiphon. Front Microbiol 2015; 6:1393. [PMID: 26696996 PMCID: PMC4677103 DOI: 10.3389/fmicb.2015.01393] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/23/2015] [Indexed: 11/20/2022] Open
Abstract
The tryptophan-rich sensory protein (TSPO) is a membrane protein, which is a member of the 18 kDa translocator protein/peripheral-type benzodiazepine receptor (MBR) family of proteins that is present in most organisms and is also referred to as Translocator protein 18 kDa. Although TSPO is associated with stress- and disease-related processes in organisms from bacteria to mammals, full elucidation of the functional role of the TSPO protein is lacking for most organisms in which it is found. In this study, we describe the regulation and function of a TSPO homolog in the cyanobacterium Fremyella diplosiphon, designated FdTSPO. Accumulation of the FdTSPO transcript is upregulated by green light and in response to nutrient deficiency and stress. A F. diplosiphon TSPO deletion mutant (i.e., ΔFdTSPO) showed altered responses compared to the wild type (WT) strain under stress conditions, including salt treatment, osmotic stress, and induced oxidative stress. Under salt stress, the FdTSPO transcript is upregulated and a ΔFdTSPO mutant accumulates lower levels of reactive oxygen species (ROS) and displays increased growth compared to WT. In response to osmotic stress, FdTSPO transcript levels are upregulated and ΔFdTSPO mutant cells exhibit impaired growth compared to the WT. By comparison, methyl viologen-induced oxidative stress results in higher ROS levels in the ΔFdTSPO mutant compared to the WT strain. Taken together, our results provide support for the involvement of membrane-localized FdTSPO in mediating cellular responses to stress in F. diplosiphon and represent detailed functional analysis of a cyanobacterial TSPO. This study advances our understanding of the functional roles of TSPO homologs in vivo.
Collapse
Affiliation(s)
- Andrea W. U. Busch
- Department of Energy – Plant Research Laboratory, Michigan State University, East LansingMI, USA
| | - Beronda L. Montgomery
- Department of Energy – Plant Research Laboratory, Michigan State University, East LansingMI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East LansingMI, USA
| |
Collapse
|
50
|
Scholz R, Caramoy A, Bhuckory MB, Rashid K, Chen M, Xu H, Grimm C, Langmann T. Targeting translocator protein (18 kDa) (TSPO) dampens pro-inflammatory microglia reactivity in the retina and protects from degeneration. J Neuroinflammation 2015; 12:201. [PMID: 26527153 PMCID: PMC4630900 DOI: 10.1186/s12974-015-0422-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/26/2015] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Reactive microglia are commonly seen in retinal degenerative diseases, and neurotoxic microglia responses can contribute to photoreceptor cell death. We and others have previously shown that translocator protein (18 kDa) (TSPO) is highly induced in retinal degenerations and that the selective TSPO ligand XBD173 (AC-5216, emapunil) exerts strong anti-inflammatory effects on microglia in vitro and ex vivo. Here, we investigated whether targeting TSPO with XBD173 has immuno-modulatory and neuroprotective functions in two mouse models of acute retinal degeneration using bright white light exposure. METHODS BALB/cJ and Cx3cr1(GFP/+) mice received intraperitoneal injections of 10 mg/kg XBD173 or vehicle for five consecutive days, starting 1 day prior to exposure to either 15,000 lux white light for 1 h or 50,000 lux focal light for 10 min, respectively. The effects of XBD173 treatment on microglia and Müller cell reactivity were analyzed by immuno-stainings of retinal sections and flat mounts, fluorescence-activated cell sorting (FACS) analysis, and mRNA expression of microglia markers using quantitative real-time PCR (qRT-PCR). Optical coherence tomography (OCT), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) stainings, and morphometric analyses were used to quantify the extent of retinal degeneration and photoreceptor apoptosis. RESULTS Four days after the mice were challenged with bright white light, a large number of amoeboid-shaped alerted microglia appeared in the degenerating outer retina, which was nearly completely prevented by treatment with XBD173. This treatment also down-regulated the expression of TSPO protein in microglia but did not change the TSPO levels in the retinal pigment epithelium (RPE). RT-PCR analysis showed that the microglia/macrophage markers Cd68 and activated microglia/macrophage whey acidic protein (Amwap) as well as the pro-inflammatory genes Ccl2 and Il6 were reduced after XBD173 treatment. Light-induced degeneration of the outer retina was nearly fully blocked by XBD173 treatment. We further confirmed these findings in an independent mouse model of focal light damage. Retinas of animals receiving XBD173 therapy displayed significantly more ramified non-reactive microglia and more viable arrestin-positive cone photoreceptors than vehicle controls. CONCLUSIONS Targeting TSPO with XBD173 effectively counter-regulates microgliosis and ameliorates light-induced retinal damage, highlighting a new pharmacological concept for the treatment of retinal degenerations.
Collapse
Affiliation(s)
- Rebecca Scholz
- Department of Ophthalmology, Laboratory for Experimental Immunology of the Eye, University of Cologne, 50931, Cologne, Germany.
| | - Albert Caramoy
- Department of Ophthalmology, Laboratory for Experimental Immunology of the Eye, University of Cologne, 50931, Cologne, Germany.
| | - Mohajeet B Bhuckory
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT12 6BA, UK.
| | - Khalid Rashid
- Department of Ophthalmology, Laboratory for Experimental Immunology of the Eye, University of Cologne, 50931, Cologne, Germany.
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT12 6BA, UK.
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT12 6BA, UK.
| | - Christian Grimm
- Department of Ophthalmology, Lab for Retinal Cell Biology, University of Zürich, 8057, Zürich, Switzerland.
| | - Thomas Langmann
- Department of Ophthalmology, Laboratory for Experimental Immunology of the Eye, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|