1
|
McCarroll MN, Sisko E, Gong JH, Teng J, Taylor J, Myers-Turnbull D, Young D, Burley G, Pierce LX, Hibbs RE, Kokel D, Sello JK. A Multimodal, In Vivo Approach for Assessing Structurally and Phenotypically Related Neuroactive Molecules. ACS Chem Neurosci 2024. [PMID: 39287508 DOI: 10.1021/acschemneuro.4c00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
A recently reported behavioral screen in larval zebrafish for phenocopiers of known anesthetics and associated drugs yielded an isoflavone. Related isoflavones have also been reported as GABAA potentiators. From this, we synthesized a small library of isoflavones and incorporated an in vivo phenotypic approach to perform structure-behavior relationship studies of the screening hit and related analogs via behavioral profiling, patch-clamp experiments, and whole brain imaging. This revealed that analogs effect a range of behavioral responses, including sedation with and without enhancing the acoustic startle response. Interestingly, a subset of compounds effect sedation and enhancement of motor responses to both acoustic and light stimuli. Patch clamp recordings of cells with a human GABAA receptor confirmed that behavior-modulating isoflavones modify the GABA signaling. To better understand these molecules' nuanced effects on behavior, we performed whole brain imaging to reveal that analogs differentially effect neuronal activity. These studies demonstrate a multimodal approach to assessing activities of neuroactives.
Collapse
Affiliation(s)
- Matthew N McCarroll
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158, United States
| | - Elizabeth Sisko
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| | - Jung Ho Gong
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Jinfeng Teng
- Department of Neurobiology, University of California, San Diego, California 92093, United States
| | - Jack Taylor
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158, United States
- UCSF Weill Institute for Neurosciences Memory and Aging Center, University of California, San Francisco, California 94158, United States
| | - Douglas Myers-Turnbull
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158, United States
| | - Drew Young
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158, United States
| | - Grant Burley
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| | - Lain X Pierce
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| | - Ryan E Hibbs
- Department of Neurobiology, University of California, San Diego, California 92093, United States
| | - David Kokel
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California 94158, United States
| | - Jason K Sello
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
2
|
Jullian E, Russi M, Turki E, Bouvelot M, Tixier L, Middendorp S, Martin E, Monnier V. Glial overexpression of Tspo extends lifespan and protects against frataxin deficiency in Drosophila. Biochimie 2024; 224:71-79. [PMID: 38750879 DOI: 10.1016/j.biochi.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024]
Abstract
The translocator protein TSPO is an evolutionary conserved mitochondrial protein overexpressed in various contexts of neurodegeneration. Friedreich Ataxia (FA) is a neurodegenerative disease due to GAA expansions in the FXN gene leading to decreased expression of frataxin, a mitochondrial protein involved in the biosynthesis of iron-sulfur clusters. We previously reported that Tspo was overexpressed in a Drosophila model of this disease generated by CRISPR/Cas9 insertion of approximately 200 GAA in the intron of fh, the fly frataxin gene. Here, we describe a new Drosophila model of FA with 42 GAA repeats, called fh-GAAs. The smaller expansion size allowed to obtain adults exhibiting hallmarks of the FA disease, including short lifespan, locomotory defects and hypersensitivity to oxidative stress. The reduced lifespan was fully rescued by ubiquitous expression of human FXN, confirming that both frataxins share conserved functions. We observed that Tspo was overexpressed in heads and decreased in intestines of these fh-GAAs flies. Then, we further overexpressed Tspo specifically in glial cells and observed improved survival. Finally, we investigated the effects of Tspo overexpression in healthy flies. Increased longevity was conferred by glial-specific overexpression, with opposite effects in neurons. Overall, this study highlights protective effects of glial TSPO in Drosophila both in a neurodegenerative and a healthy context.
Collapse
Affiliation(s)
- Estelle Jullian
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| | - Maria Russi
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| | - Ema Turki
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| | - Margaux Bouvelot
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| | - Laura Tixier
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| | - Sandrine Middendorp
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| | - Elodie Martin
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| | - Véronique Monnier
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251, CNRS, F-75013, Paris, France.
| |
Collapse
|
3
|
Giladi M, Montgomery AP, Kassiou M, Danon JJ. Structure-based drug design for TSPO: Challenges and opportunities. Biochimie 2024; 224:41-50. [PMID: 38782353 DOI: 10.1016/j.biochi.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/27/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
The translocator protein 18 kDa (TSPO) is an evolutionarily conserved mitochondrial transmembrane protein implicated in various neuropathologies and inflammatory conditions, making it a longstanding diagnostic and therapeutic target of interest. Despite the development of various classes of TSPO ligand chemotypes, and the elucidation of bacterial and non-human mammalian experimental structures, many unknowns exist surrounding its differential structural and functional features in health and disease. There are several limitations associated with currently used computational methodologies for modelling the native structure and ligand-binding behaviour of this enigmatic protein. In this perspective, we provide a critical analysis of the developments in the uses of these methods, outlining their uses, inherent limitations, and continuing challenges. We offer suggestions of unexplored opportunities that exist in the use of computational methodologies which offer promise for enhancing our understanding of the TSPO.
Collapse
Affiliation(s)
- Mia Giladi
- School of Chemistry, The University of Sydney, 2050, Sydney, NSW, Australia
| | | | - Michael Kassiou
- School of Chemistry, The University of Sydney, 2050, Sydney, NSW, Australia.
| | - Jonathan J Danon
- School of Chemistry, The University of Sydney, 2050, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Firth W, Robb JL, Stewart D, Pye KR, Bamford R, Oguro-Ando A, Beall C, Ellacott KLJ. Regulation of astrocyte metabolism by mitochondrial translocator protein 18 kDa. J Neurochem 2024; 168:1374-1401. [PMID: 38482552 DOI: 10.1111/jnc.16089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 03/26/2024]
Abstract
The mitochondrial translocator protein 18 kDa (TSPO) has been linked to functions from steroidogenesis to regulation of cellular metabolism and is an attractive therapeutic target for chronic CNS inflammation. Studies in Leydig cells and microglia indicate that TSPO function may vary between cells depending on their specialized roles. Astrocytes are critical for providing trophic and metabolic support in the brain. Recent work has highlighted that TSPO expression increases in astrocytes under inflamed conditions and may drive astrocyte reactivity. Relatively little is known about the role TSPO plays in regulating astrocyte metabolism and whether this protein is involved in immunometabolic processes in these cells. Using TSPO-deficient (TSPO-/-) mouse primary astrocytes in vitro (MPAs) and a human astrocytoma cell line (U373 cells), we performed extracellular metabolic flux analyses. We found that TSPO deficiency reduced basal cellular respiration and attenuated the bioenergetic response to glucopenia. Fatty acid oxidation was increased, and lactate production was reduced in TSPO-/- MPAs and U373 cells. Co-immunoprecipitation studies revealed that TSPO forms a complex with carnitine palmitoyltransferase 1a in U373 and MPAs, presenting a mechanism wherein TSPO may regulate FAO in these cells. Compared to TSPO+/+ cells, in TSPO-/- MPAs we observed attenuated tumor necrosis factor release following 3 h lipopolysaccharide (LPS) stimulation, which was enhanced at 24 h post-LPS stimulation. Together these data suggest that while TSPO acts as a regulator of metabolic flexibility, TSPO deficiency does not appear to modulate the metabolic response of MPAs to inflammation, at least in response to the model used in this study.
Collapse
Affiliation(s)
- Wyn Firth
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Josephine L Robb
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Daisy Stewart
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Katherine R Pye
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Rosemary Bamford
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Asami Oguro-Ando
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Craig Beall
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kate L J Ellacott
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
5
|
Bäckström T, Doverskog M, Blackburn TP, Scharschmidt BF, Felipo V. Allopregnanolone and its antagonist modulate neuroinflammation and neurological impairment. Neurosci Biobehav Rev 2024; 161:105668. [PMID: 38608826 DOI: 10.1016/j.neubiorev.2024.105668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
Neuroinflammation accompanies several brain disorders, either as a secondary consequence or as a primary cause and may contribute importantly to disease pathogenesis. Neurosteroids which act as Positive Steroid Allosteric GABA-A receptor Modulators (Steroid-PAM) appear to modulate neuroinflammation and their levels in the brain may vary because of increased or decreased local production or import from the systemic circulation. The increased synthesis of steroid-PAMs is possibly due to increased expression of the mitochondrial cholesterol transporting protein (TSPO) in neuroinflammatory tissue, and reduced production may be due to changes in the enzymatic activity. Microglia and astrocytes play an important role in neuroinflammation, and their production of inflammatory mediators can be both activated and inhibited by steroid-PAMs and GABA. What is surprising is the finding that both allopregnanolone, a steroid-PAM, and golexanolone, a novel GABA-A receptor modulating steroid antagonist (GAMSA), can inhibit microglia and astrocyte activation and normalize their function. This review focuses on the role of steroid-PAMs in neuroinflammation and their importance in new therapeutic approaches to CNS and liver disease.
Collapse
Affiliation(s)
| | | | | | | | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
6
|
Sandström A, Torrado-Carvajal A, Morrissey EJ, Kim M, Alshelh Z, Zhu Y, Li MD, Chang CY, Jarraya M, Akeju O, Schrepf A, Harris RE, Kwon YM, Bedair H, Chen AF, Mercaldo ND, Kettner N, Napadow V, Toschi N, Edwards RR, Loggia ML. [ 11 C]-PBR28 positron emission tomography signal as an imaging marker of joint inflammation in knee osteoarthritis. Pain 2024; 165:1121-1130. [PMID: 38015622 DOI: 10.1097/j.pain.0000000000003114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/24/2023] [Indexed: 11/30/2023]
Abstract
ABSTRACT Although inflammation is known to play a role in knee osteoarthritis (KOA), inflammation-specific imaging is not routinely performed. In this article, we evaluate the role of joint inflammation, measured using [ 11 C]-PBR28, a radioligand for the inflammatory marker 18-kDa translocator protein (TSPO), in KOA. Twenty-one KOA patients and 11 healthy controls (HC) underwent positron emission tomography/magnetic resonance imaging (PET/MRI) knee imaging with the TSPO ligand [ 11 C]-PBR28. Standardized uptake values were extracted from regions-of-interest (ROIs) semiautomatically segmented from MRI data, and compared across groups (HC, KOA) and subgroups (unilateral/bilateral KOA symptoms), across knees (most vs least painful), and against clinical variables (eg, pain and Kellgren-Lawrence [KL] grades). Overall, KOA patients demonstrated elevated [ 11 C]-PBR28 binding across all knee ROIs, compared with HC (all P 's < 0.005). Specifically, PET signal was significantly elevated in both knees in patients with bilateral KOA symptoms (both P 's < 0.01), and in the symptomatic knee ( P < 0.05), but not the asymptomatic knee ( P = 0.95) of patients with unilateral KOA symptoms. Positron emission tomography signal was higher in the most vs least painful knee ( P < 0.001), and the difference in pain ratings across knees was proportional to the difference in PET signal ( r = 0.74, P < 0.001). Kellgren-Lawrence grades neither correlated with PET signal (left knee r = 0.32, P = 0.19; right knee r = 0.18, P = 0.45) nor pain ( r = 0.39, P = 0.07). The current results support further exploration of [ 11 C]-PBR28 PET signal as an imaging marker candidate for KOA and a link between joint inflammation and osteoarthritis-related pain severity.
Collapse
Affiliation(s)
- Angelica Sandström
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Angel Torrado-Carvajal
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
- Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Madrid, Spain
| | - Erin J Morrissey
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Minhae Kim
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Zeynab Alshelh
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Yehui Zhu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Matthew D Li
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Connie Y Chang
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Mohamed Jarraya
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Andrew Schrepf
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States
| | - Richard E Harris
- Susan Samueli Integrative Health Institute, School of Medicine, University of California at Irvine, Irvine CA, United States
- Department of Anesthesiology and Perioperative Care, School of Medicine, University of California at Irvine, Irvine CA, United States
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States
| | - Young-Min Kwon
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Hany Bedair
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Antonia F Chen
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Nathaniel D Mercaldo
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Norman Kettner
- Department of Radiology, Logan University, Chesterfield, MO, United States
| | - Vitaly Napadow
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Nicola Toschi
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Biomedicine and Prevention, University of Rome, "Tor Vergata," Rome, Italy
| | - Robert R Edwards
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Marco L Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
7
|
Song J, Yu W, Chen S, Huang J, Zhou C, Liang H. Remimazolam attenuates inflammation and kidney fibrosis following folic acid injury. Eur J Pharmacol 2024; 966:176342. [PMID: 38290569 DOI: 10.1016/j.ejphar.2024.176342] [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: 09/10/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
The transition of acute kidney injury (AKI) to chronic kidney disease (CKD) is characterized by intense inflammation and progressive fibrosis. Remimazolam is widely used for procedural sedation in intensive care units, such as AKI patients. Remimazolam has been shown to possess anti-inflammatory and organ-protective properties. However, the role of remimazolam in inflammation and renal fibrosis following AKI remains unclear. Here, we explored the effects of remimazolam on the inflammatory response and kidney fibrogenesis of mice subjected to folic acid (FA) injury. Our results showed that remimazolam treatment alleviated kidney damage and dysfunction. Mice treated with remimazolam presented less collagen deposition in FA-injured kidneys compared with FA controls, which was accompanied by a reduction of extracellular matrix proteins accumulation and fibroblasts activation. Furthermore, remimazolam treatment reduced inflammatory cells infiltration into the kidneys of mice with FA injury and inhibited proinflammatory or profibrotic molecules expression. Finally, remimazolam treatment impaired the activation of bone marrow-derived fibroblasts and blunted the transformation of macrophages to myofibroblasts in FA nephropathy. Additionally, the benzodiazepine receptor antagonist PK-11195 partially reversed the protective effect of remimazolam on the FA-injured kidneys. Overall, remimazolam attenuates the inflammatory response and renal fibrosis development following FA-induced AKI, which may be related to the peripheral benzodiazepine receptor pathway.
Collapse
Affiliation(s)
- Jinfang Song
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, 528000, China; Zhuhai Campus, Zunyi Medical University, Zhuhai, 519041, China.
| | - Wenqiang Yu
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, 528000, China.
| | - Shuangquan Chen
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, 528000, China.
| | - Jiamin Huang
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, 528000, China.
| | - Chujun Zhou
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China.
| | - Hua Liang
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, 528000, China.
| |
Collapse
|
8
|
Yu M, Zhao S. Functional role of translocator protein and its ligands in ocular diseases (Review). Mol Med Rep 2024; 29:33. [PMID: 38186312 PMCID: PMC10804439 DOI: 10.3892/mmr.2024.13157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
The 18 kDa translocator protein (TSPO) is an essential outer mitochondrial membrane protein that is responsible for mitochondrial transport, maintenance of mitochondrial homeostasis and normal physiological cell function. The role of TSPO in the pathogenesis of ocular diseases is a growing area of interest. More notably, TSPO exerts positive effects in regulating various pathophysiological processes, such as the inflammatory response, oxidative stress, steroid synthesis and modulation of microglial function, in combination with a variety of specific ligands such as 1‑(2‑chlorophenyl‑N‑methylpropyl)‑3‑isoquinolinecarboxamide, 4'‑chlorodiazepam and XBD173. In the present review, the expression of TSPO in ocular tissues and the functional role of TSPO and its ligands in diverse ocular diseases was discussed.
Collapse
Affiliation(s)
- Mingyi Yu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 30384, P.R. China
| | - Shaozhen Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 30384, P.R. China
| |
Collapse
|
9
|
Zhang S, Deng Z, Qiu Y, Lu G, Wu J, Huang H. FGIN-1-27 Mitigates Radiation-induced Mitochondrial Hyperfunction and Cellular Hyperactivation in Cultured Astrocytes. Neuroscience 2023; 535:23-35. [PMID: 37913861 DOI: 10.1016/j.neuroscience.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
Radiation-induced brain injury (RBI) poses a significant challenge in the context of radiotherapy for intracranial tumors, necessitating a comprehensive understanding of the cellular and molecular mechanisms involved. While prior investigations have underscored the role of astrocyte activation and excessive vascular endothelial growth factor production in microvascular damage associated with RBI, there remains a scarcity of studies examining the impact of radiation on astrocytes, particularly regarding organelles such as mitochondria. Thus, our study aimed to elucidate alterations in astrocyte and mitochondrial functionality following radiation exposure, with a specific focus on evaluating the potential ameliorative effects of translocator protein 18 kDa(TSPO) ligands. In this study, cultured astrocytes were subjected to X-ray irradiation, and their cellular states and mitochondrial functions were examined and compared to control cells. Our findings revealed that radiation-induced astrocytic hyperactivation, transforming them into the neurotoxic A1-type, concomitant with reduced cell proliferation. Additionally, radiation triggered mitochondrial hyperfunction, heightened the mitochondrial membrane potential, and increased oxidative metabolite production. However, following treatment with FGIN-1-27, a TSPO ligand, we observed a restoration of mitochondrial function and a reduction in oxidative metabolite production. Moreover, this intervention mitigated astrocyte hyperactivity, decreased the number of A1-type astrocytes, and restored cell proliferative capacity. In conclusion, our study has unveiled additional manifestations of radiation-induced astrocyte dysfunction and validated that TSPO ligands may serve as a promising therapeutic strategy to mitigate this dysfunction. It has potential clinical implications for the treatment of RBI.
Collapse
Affiliation(s)
- Shifeng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Zhezhi Deng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yuemin Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Gengxin Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Junyu Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Haiwei Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China.
| |
Collapse
|
10
|
Tuominen S, Nissi L, Kukkula A, Routila J, Huusko T, Leivo I, Minn H, Irjala H, Löyttyniemi E, Ventelä S, Sundvall M, Grönroos TJ. TSPO is a potential independent prognostic factor associated with cellular respiration and p16 in head and neck squamous cell carcinoma. Front Oncol 2023; 13:1298333. [PMID: 38162485 PMCID: PMC10755888 DOI: 10.3389/fonc.2023.1298333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Background Treatment resistance and relapse are common problems in head and neck squamous cell carcinoma (HNSCC). Except for p16, no clinically accepted prognostic biomarkers are available for HNSCC. New biomarkers predictive of recurrence and survival are crucial for optimal treatment planning and patient outcome. High translocator protein (TSPO) levels have been associated with poor survival in cancer, but the role of TSPO has not been extensively evaluated in HNSCC. Materials and methods TSPO expression was determined in a large population-based tissue microarray cohort including 611 patients with HNSCC and evaluated for survival in several clinicopathological subgroups. A TCGA HNSCC cohort was used to further analyze the role of TSPO in HNSCC. Results TSPO expression was downregulated in more aggressive tumors. Low TSPO expression associated with worse 5-year survival and was an independent prognostic factor for disease-specific survival. Subgroup analyses showed that low TSPO expression associated with worse survival particularly in p16-positive oropharyngeal cancer. In silico analyses supported the prognostic role of TSPO. Cellular respiration had the highest significance in pathway analyses for genes expressed positively with TSPO. Conclusion Decreased TSPO expression associates with poor prognosis in HNSCC. TSPO is a prognostic biomarker in HNSCC to potentially guide treatment stratification especially in p16-positive oropharyngeal cancer.
Collapse
Affiliation(s)
- Sanni Tuominen
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
- Cancer Research Unit, Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Research Laboratory, Turku University Hospital and University of Turku, Turku, Finland
- Medicity Research Laboratory, Faculty of Medicine, University of Turku, Turku, Finland
| | - Linda Nissi
- Department of Clinical Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Antti Kukkula
- Cancer Research Unit, Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Research Laboratory, Turku University Hospital and University of Turku, Turku, Finland
| | - Johannes Routila
- Department of Otorhinolaryngology – Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Teemu Huusko
- Department of Otorhinolaryngology – Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Ilmo Leivo
- Department of Pathology, Turku University Hospital and University of Turku, Turku, Finland
| | - Heikki Minn
- Department of Clinical Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Heikki Irjala
- Department of Otorhinolaryngology – Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, Turku University Hospital and University of Turku, Turku, Finland
| | - Sami Ventelä
- Department of Otorhinolaryngology – Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Maria Sundvall
- Cancer Research Unit, Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Research Laboratory, Turku University Hospital and University of Turku, Turku, Finland
- Department of Clinical Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Tove J. Grönroos
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
- Medicity Research Laboratory, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Clinical Oncology, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
11
|
Firth W, Robb JL, Stewart D, Pye KR, Bamford R, Oguro-Ando A, Beall C, Ellacott KLJ. Regulation of astrocyte metabolism by mitochondrial translocator protein 18kDa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560159. [PMID: 37873215 PMCID: PMC10592862 DOI: 10.1101/2023.09.29.560159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The mitochondrial translocator protein 18kDa (TSPO) has been linked to a variety of functions from steroidogenesis to regulation of cellular metabolism and is an attractive therapeutic target for chronic CNS inflammation. Studies in the periphery using Leydig cells and hepatocytes, as well as work in microglia, indicate that the function of TSPO may vary between cells depending on their specialised roles. Astrocytes are critical for providing trophic and metabolic support in the brain as part of their role in maintaining brain homeostasis. Recent work has highlighted that TSPO expression increases in astrocytes under inflamed conditions and may drive astrocyte reactivity. However, relatively little is known about the role TSPO plays in regulating astrocyte metabolism and whether this protein is involved in immunometabolic processes in these cells. Using TSPO-deficient (TSPO-/-) mouse primary astrocytes in vitro (MPAs) and a human astrocytoma cell line (U373 cells), we performed metabolic flux analyses. We found that loss of TSPO reduced basal astrocyte respiration and increased the bioenergetic response to glucose reintroduction following glucopenia, while increasing fatty acid oxidation (FAO). Lactate production was significantly reduced in TSPO-/- astrocytes. Co-immunoprecipitation studies in U373 cells revealed that TSPO forms a complex with carnitine palmitoyltransferase 1a, which presents a mechanism wherein TSPO may regulate FAO in astrocytes. Compared to TSPO+/+ cells, inflammation induced by 3h lipopolysaccharide (LPS) stimulation of TSPO-/- MPAs revealed attenuated tumour necrosis factor release, which was enhanced in TSPO-/- MPAs at 24h LPS stimulation. Together these data suggest that while TSPO acts as a regulator of metabolic flexibility in astrocytes, loss of TSPO does not appear to modulate the metabolic response of astrocytes to inflammation, at least in response to the stimulus/time course used in this study.
Collapse
Affiliation(s)
- Wyn Firth
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Josephine L Robb
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Daisy Stewart
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Katherine R Pye
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Rosemary Bamford
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Asami Oguro-Ando
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Craig Beall
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kate LJ Ellacott
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
12
|
Montégut L, Abdellatif M, Motiño O, Madeo F, Martins I, Quesada V, López‐Otín C, Kroemer G. Acyl coenzyme A binding protein (ACBP): An aging- and disease-relevant "autophagy checkpoint". Aging Cell 2023; 22:e13910. [PMID: 37357988 PMCID: PMC10497816 DOI: 10.1111/acel.13910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023] Open
Abstract
Acyl coenzyme A binding protein (ACBP), also known as diazepam-binding inhibitor (DBI), is a phylogenetically ancient protein present in some eubacteria and the entire eukaryotic radiation. In several eukaryotic phyla, ACBP/DBI transcends its intracellular function in fatty acid metabolism because it can be released into the extracellular space. This ACBP/DBI secretion usually occurs in response to nutrient scarcity through an autophagy-dependent pathway. ACBP/DBI and its peptide fragments then act on a range of distinct receptors that diverge among phyla, namely metabotropic G protein-coupled receptor in yeast (and likely in the mammalian central nervous system), a histidine receptor kinase in slime molds, and ionotropic gamma-aminobutyric acid (GABA)A receptors in mammals. Genetic or antibody-mediated inhibition of ACBP/DBI orthologs interferes with nutrient stress-induced adaptations such as sporulation or increased food intake in multiple species, as it enhances lifespan or healthspan in yeast, plant leaves, nematodes, and multiple mouse models. These lifespan and healthspan-extending effects of ACBP/DBI suppression are coupled to the induction of autophagy. Altogether, it appears that neutralization of extracellular ACBP/DBI results in "autophagy checkpoint inhibition" to unleash the anti-aging potential of autophagy. Of note, in humans, ACBP/DBI levels increase in various tissues, as well as in the plasma, in the context of aging, obesity, uncontrolled infection or cardiovascular, inflammatory, neurodegenerative, and malignant diseases.
Collapse
Affiliation(s)
- Léa Montégut
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Inserm U1138Université Paris Cité, Sorbonne UniversitéParisFrance
- Metabolomics and Cell Biology PlatformsGustave Roussy InstitutVillejuifFrance
- Faculté de MédecineUniversité de Paris SaclayParisFrance
| | - Mahmoud Abdellatif
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Inserm U1138Université Paris Cité, Sorbonne UniversitéParisFrance
- Metabolomics and Cell Biology PlatformsGustave Roussy InstitutVillejuifFrance
- Department of CardiologyMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| | - Omar Motiño
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Inserm U1138Université Paris Cité, Sorbonne UniversitéParisFrance
- Metabolomics and Cell Biology PlatformsGustave Roussy InstitutVillejuifFrance
| | - Frank Madeo
- BioTechMed‐GrazGrazAustria
- Institute of Molecular Biosciences, NAWI GrazUniversity of GrazGrazAustria
- Field of Excellence BioHealthUniversity of GrazGrazAustria
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Inserm U1138Université Paris Cité, Sorbonne UniversitéParisFrance
- Metabolomics and Cell Biology PlatformsGustave Roussy InstitutVillejuifFrance
| | - Victor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología del Principado de Asturias (IUOPA)Universidad de OviedoOviedoSpain
| | - Carlos López‐Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Inserm U1138Université Paris Cité, Sorbonne UniversitéParisFrance
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología del Principado de Asturias (IUOPA)Universidad de OviedoOviedoSpain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Inserm U1138Université Paris Cité, Sorbonne UniversitéParisFrance
- Metabolomics and Cell Biology PlatformsGustave Roussy InstitutVillejuifFrance
- Institut du Cancer Paris CARPEM, Department of BiologyHôpital Européen Georges Pompidou, AP‐HPParisFrance
| |
Collapse
|
13
|
Yu C, Deng XJ, Xu D. Microglia in epilepsy. Neurobiol Dis 2023; 185:106249. [PMID: 37536386 DOI: 10.1016/j.nbd.2023.106249] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/07/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
Epilepsy is one of most common chronic neurological disorders, and the antiseizure medications developed by targeting neurocentric mechanisms have not effectively reduced the proportion of patients with drug-resistant epilepsy. Further exploration of the cellular or molecular mechanism of epilepsy is expected to provide new options for treatment. Recently, more and more researches focus on brain network components other than neurons, among which microglia have attracted much attention for their diverse biological functions. As the resident immune cells of the central nervous system, microglia have highly plastic transcription, morphology and functional characteristics, which can change dynamically in a context-dependent manner during the progression of epilepsy. In the pathogenesis of epilepsy, highly reactive microglia interact with other components in the epileptogenic network by performing crucial functions such as secretion of soluble factors and phagocytosis, thus continuously reshaping the landscape of the epileptic brain microenvironment. Indeed, microglia appear to be both pro-epileptic and anti-epileptic under the different spatiotemporal contexts of disease, rendering interventions targeting microglia biologically complex and challenging. This comprehensive review critically summarizes the pathophysiological role of microglia in epileptic brain homeostasis alterations and explores potential therapeutic or modulatory targets for epilepsy targeting microglia.
Collapse
Affiliation(s)
- Cheng Yu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Xue-Jun Deng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Da Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| |
Collapse
|
14
|
Fogliano C, Carotenuto R, Rusciano G, Sasso A, Motta CM, Agnisola C, Avallone B. Structural and functional damage to the retina and skeletal muscle in Xenopus laevis embryos exposed to the commonly used psychotropic benzodiazepine delorazepam. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104235. [PMID: 37481049 DOI: 10.1016/j.etap.2023.104235] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/03/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023]
Abstract
Benzodiazepines, psychotropic drugs, are among the most frequently found pharmaceuticals in aquatic matrices. An increasing number of studies are reporting their harmful effects on adults' behaviour and physiology, while little information is available regarding developing organisms exposed since early stages. Improper activation of GABA receptors during embryonic development is likely to induce relevant consequences on the morphogenesis and, at later stages, on behaviour. This study investigated the negative effects of three increasing concentrations of delorazepam on Xenopus laevis retinal and skeletal muscle morphogenesis. Morphological and ultrastructural investigations were correlated with gene expression, while Raman spectroscopy highlighted the main biochemical components affected. Conventional phototactic response and orientation in the magnetic field were assessed as indicators of proper interaction between sensory organs and the nervous system. Results confirm the profound impact of delorazepam on development and return an alarming picture of the amphibians' survival potentialities in a benzodiazepine-contaminated environment.
Collapse
Affiliation(s)
- Chiara Fogliano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Rosa Carotenuto
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Giulia Rusciano
- Department of Physics, University of Naples Federico II, Naples, Italy
| | - Antonio Sasso
- Department of Physics, University of Naples Federico II, Naples, Italy
| | | | - Claudio Agnisola
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Bice Avallone
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
15
|
Liu Y, Wang S, Yang W. Inhibiting the Proliferation of Colorectal Cancer Cells by Reducing TSPO/VDAC Expression. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:1378-1389. [PMID: 37593520 PMCID: PMC10430413 DOI: 10.18502/ijph.v52i7.13239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/13/2023] [Indexed: 08/19/2023]
Abstract
Background We aimed to explore the mechanism of the effect of remimazolam (Rem) on the proliferation of colorectal cancer (CRC) cells with CRC as a disease context. Methods Translocation protein (TSPO) expression in CRC was determined by Western blotting and qRT-PCR in the Second Affiliated Hospital of Qiqihar Medical University from March 2019 to February 2022. TSPO-interacting proteins were predicted through string database. The proliferation was measured by CCK-8 and 5-ethynyl-2-deoxyuridine (EDU). The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT) and clonal colony on cells were formed to screen for the optimal concentration of Rem and to detect the viability. The expression of apoptosis-related proteins, Bcl-2 and P53, was determined by qRT-PCR and Western blotting. The effect of Rem on the expression of tumor markers, CEA and CA19-9, in CRC was examined through ELISA. Results TSPO expression in CRC tissues and cells was higher than that in ANT samples and normal intestinal epithelial cells. Over-expression of TSPO promoted the proliferation of HCT116 and the expression of tumor markers CEA and CA19-9 and inhibited the apoptosis of HCT116. Interference with TSPO inhibited the proliferation of HCT116 and the expression of CEA and CA19-9 and promoted the apoptosis of HCT116. 1 μg/mL Rem could inhibit the viability of HCT116, the proliferation of HCT116 and the expression of CEA and CA19-9, and improve the apoptosis of HCT116. TSPO could interact with VDAC and affect its protein expression, and Rem could inhibit the proliferation and the expression of CEA and CA19-9 through the TSPO/VDAC pathway, to promote its apoptosis. Conclusion Rem affects the proliferation of CRC cells by inhibiting the TSPO/VDAC pathway.
Collapse
Affiliation(s)
- Yang Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Shuyue Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Weining Yang
- Operating Room, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| |
Collapse
|
16
|
Nevárez-Ramírez AJ, Guzmán-Ortiz AL, Cortes-Reynosa P, Perez-Salazar E, Jaimes-Ortega GA, Valle-Rios R, Marín-Hernández Á, Rodríguez-Zavala JS, Ruiz-May E, Castrejón-Flores JL, Quezada H. Shotgun Proteomics of Co-Cultured Leukemic and Bone Marrow Stromal Cells from Different Species as a Preliminary Approach to Detect Intercellular Protein Transfer. Proteomes 2023; 11:proteomes11020015. [PMID: 37092456 PMCID: PMC10123657 DOI: 10.3390/proteomes11020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/25/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Cellular interactions within the bone marrow microenvironment modulate the properties of subsets of leukemic cells leading to the development of drug-resistant phenotypes. The intercellular transfer of proteins and organelles contributes to this process but the set of transferred proteins and their effects in the receiving cells remain unclear. This study aimed to detect the intercellular protein transfer from mouse bone marrow stromal cells (OP9 cell line) to human T-lymphoblasts (CCRF-CEM cell line) using nanoLC-MS/MS-based shotgun proteomics in a 3D co-culture system. After 24 h of co-culture, 1513 and 67 proteins from human and mouse origin, respectively, were identified in CCRF-CEM cells. The presence of mouse proteins in the human cell line, detected by analyzing the differences in amino acid sequences of orthologous peptides, was interpreted as the result of intercellular transfer. The transferred proteins might have contributed to the observed resistance to vincristine, methotrexate, and hydrogen peroxide in the co-cultured leukemic cells. Our results suggest that shotgun proteomic analyses of co-cultured cells from different species could be a simple option to get a preliminary survey of the proteins exchanged among interacting cells.
Collapse
Affiliation(s)
- Abraham Josué Nevárez-Ramírez
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
- Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Av. Acueducto s/n, Barrio La Laguna, Mexico City 07340, Mexico
| | - Ana Laura Guzmán-Ortiz
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
| | - Pedro Cortes-Reynosa
- Departamento de Biología Celular, CINVESTAV-IPN, Av Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Eduardo Perez-Salazar
- Departamento de Biología Celular, CINVESTAV-IPN, Av Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Gustavo Alberto Jaimes-Ortega
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Circuito interior, Av. Universidad 3000, Ciudad Universitaria, Coyoacán, Mexico City 04510, Mexico
| | - Ricardo Valle-Rios
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Circuito interior, Av. Universidad 3000, Ciudad Universitaria, Coyoacán, Mexico City 04510, Mexico
| | - Álvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Belisario Domínguez—Sección XVI, Mexico City 14080, Mexico
| | - José S. Rodríguez-Zavala
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Belisario Domínguez—Sección XVI, Mexico City 14080, Mexico
| | - Eliel Ruiz-May
- Red de Estudios Moleculares Avanzados, Clúster Científico y Tecnológico BioMimic®, Instituto de Ecología A.C. (INECOL), Carretera Antigua a Coatepec 351, El Haya, Xalapa 91073, Mexico
| | - José Luis Castrejón-Flores
- Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Av. Acueducto s/n, Barrio La Laguna, Mexico City 07340, Mexico
| | - Héctor Quezada
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
| |
Collapse
|
17
|
Raval NR, Wetherill RR, Wiers CE, Dubroff JG, Hillmer AT. Positron Emission Tomography of Neuroimmune Responses in Humans: Insights and Intricacies. Semin Nucl Med 2023; 53:213-229. [PMID: 36270830 PMCID: PMC11261531 DOI: 10.1053/j.semnuclmed.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/06/2022]
Abstract
The brain's immune system plays a critical role in responding to immune challenges and maintaining homeostasis. However, dysregulated neuroimmune function contributes to neurodegenerative disease and neuropsychiatric conditions. In vivo positron emission tomography (PET) imaging of the neuroimmune system has facilitated a greater understanding of its physiology and the pathology of some neuropsychiatric conditions. This review presents an in-depth look at PET findings from human neuroimmune function studies, highlighting their importance in current neuropsychiatric research. Although the majority of human PET studies feature radiotracers targeting the translocator protein 18 kDa (TSPO), this review also considers studies with other neuroimmune targets, including monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, nitric oxide synthase, and the purinergic P2X7 receptor. Promising new targets, such as colony-stimulating factor 1, Sphingosine-1-phosphate receptor 1, and the purinergic P2Y12 receptor, are also discussed. The significance of validating neuroimmune targets and understanding their function and expression is emphasized in this review to better identify and interpret PET results.
Collapse
Affiliation(s)
- Nakul R Raval
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT
| | - Reagan R Wetherill
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT; Department of Psychiatry, Yale University, New Haven, CT.
| |
Collapse
|
18
|
The GABA and GABA-Receptor System in Inflammation, Anti-Tumor Immune Responses, and COVID-19. Biomedicines 2023; 11:biomedicines11020254. [PMID: 36830790 PMCID: PMC9953446 DOI: 10.3390/biomedicines11020254] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
GABA and GABAA-receptors (GABAA-Rs) play major roles in neurodevelopment and neurotransmission in the central nervous system (CNS). There has been a growing appreciation that GABAA-Rs are also present on most immune cells. Studies in the fields of autoimmune disease, cancer, parasitology, and virology have observed that GABA-R ligands have anti-inflammatory actions on T cells and antigen-presenting cells (APCs), while also enhancing regulatory T cell (Treg) responses and shifting APCs toward anti-inflammatory phenotypes. These actions have enabled GABAA-R ligands to ameliorate autoimmune diseases, such as type 1 diabetes (T1D), multiple sclerosis (MS), and rheumatoid arthritis, as well as type 2 diabetes (T2D)-associated inflammation in preclinical models. Conversely, antagonism of GABAA-R activity promotes the pro-inflammatory responses of T cells and APCs, enhancing anti-tumor responses and reducing tumor burden in models of solid tumors. Lung epithelial cells also express GABA-Rs, whose activation helps maintain fluid homeostasis and promote recovery from injury. The ability of GABAA-R agonists to limit both excessive immune responses and lung epithelial cell injury may underlie recent findings that GABAA-R agonists reduce the severity of disease in mice infected with highly lethal coronaviruses (SARS-CoV-2 and MHV-1). These observations suggest that GABAA-R agonists may provide off-the-shelf therapies for COVID-19 caused by new SARS-CoV-2 variants, as well as novel beta-coronaviruses, which evade vaccine-induced immune responses and antiviral medications. We review these findings and further advance the notions that (1) immune cells possess GABAA-Rs to limit inflammation in the CNS, and (2) this natural "braking system" on inflammatory responses may be pharmacologically engaged to slow the progression of autoimmune diseases, reduce the severity of COVID-19, and perhaps limit neuroinflammation associated with long COVID.
Collapse
|
19
|
Proteomics analysis in myocardium of spontaneously hypertensive rats. Sci Rep 2023; 13:276. [PMID: 36609626 PMCID: PMC9822958 DOI: 10.1038/s41598-023-27590-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023] Open
Abstract
Hypertension-related left ventricular hypertrophy is recognized as a good predictor of adverse cardiovascular events. However, the underlying mechanism of left ventricular hypertrophy is still not fully understood. This study employed liquid chromatography coupled with tandem mass spectrometry to investigate global changes in protein profile in myocardium of spontaneously hypertensive rat, a classical animal model of essential hypertension. There were 369 differentially expressed proteins in myocardium between spontaneously hypertensive rats and normotensive rats. Xenobiotic catabolic process, cholesterol binding and mitochondrial proton-transporting ATP synthase were found to be the most significantly enriched biological process, molecular function and cellular component terms of Gene Ontology, respectively. Drug metabolism-cytochrome P450 was revealed to be the most significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways. FYN proto-oncogene, Src family tyrosine kinase was found to have the most interactions with other proteins. Differentially expressed proteins involved in xenobiotic catabolic process, lipid transport and metabolism, mitochondrial function might be targets for further study of hypertension-related left ventricular hypertrophy.
Collapse
|
20
|
Janisch N, Levendosky K, Budell WC, Quadri LEN. Genetic Underpinnings of Carotenogenesis and Light-Induced Transcriptome Remodeling in the Opportunistic Pathogen Mycobacterium kansasii. Pathogens 2023; 12:86. [PMID: 36678434 PMCID: PMC9861118 DOI: 10.3390/pathogens12010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Mycobacterium kansasii (Mk) causes opportunistic pulmonary infections with tuberculosis-like features. The bacterium is well known for its photochromogenicity, i.e., the production of carotenoid pigments in response to light. The genetics defining the photochromogenic phenotype of Mk has not been investigated and defined pigmentation mutants to facilitate studies on the role of carotenes in the bacterium's biology are not available thus far. In this study, we set out to identify genetic determinants involved in Mk photochromogenicity. We screened a library of ~150,000 transposon mutants for colonies with pigmentation abnormalities. The screen rendered a collection of ~200 mutants. Each of these mutants could be assigned to one of four distinct phenotypic groups. The insertion sites in the mutant collection clustered in three chromosomal regions. A combination of phenotypic analysis, sequence bioinformatics, and gene expression studies linked these regions to carotene biosynthesis, carotene degradation, and monounsaturated fatty acid biosynthesis. Furthermore, introduction of the identified carotenoid biosynthetic gene cluster into non-pigmented Mycobacterium smegmatis endowed the bacterium with photochromogenicity. The studies also led to identification of MarR-type and TetR/AcrR-type regulators controlling photochromogenicity and carotenoid breakdown, respectively. Lastly, the work presented also provides a first insight into the Mk transcriptome changes in response to light.
Collapse
Affiliation(s)
- Niklas Janisch
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Keith Levendosky
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - William C. Budell
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Luis E. N. Quadri
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- Biochemistry Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
21
|
Singh P, kumari N, Kaul A, Srivastava A, Singh VK, Srivastava K, Tiwari AK. Acetamidobenzoxazolone conjugated DOTA system for assessing 18 kDa translocator protein during pulmonary inflammation. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
22
|
Martinez-Orengo N, Tahmazian S, Lai J, Wang Z, Sinharay S, Schreiber-Stainthorp W, Basuli F, Maric D, Reid W, Shah S, Hammoud DA. Assessing organ-level immunoreactivity in a rat model of sepsis using TSPO PET imaging. Front Immunol 2022; 13:1010263. [PMID: 36439175 PMCID: PMC9685400 DOI: 10.3389/fimmu.2022.1010263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
There is current need for new approaches to assess/measure organ-level immunoreactivity and ensuing dysfunction in systemic inflammatory response syndrome (SIRS) and sepsis, in order to protect or recover organ function. Using a rat model of systemic sterile inflammatory shock (intravenous LPS administration), we performed PET imaging with a translocator protein (TSPO) tracer, [18F]DPA-714, as a biomarker for reactive immunoreactive changes in the brain and peripheral organs. In vivo dynamic PET/CT scans showed increased [18F]DPA-714 binding in the brain, lungs, liver and bone marrow, 4 hours after LPS injection. Post-LPS mean standard uptake values (SUVmean) at equilibrium were significantly higher in those organs compared to baseline. Changes in spleen [18F]DPA-714 binding were variable but generally decreased after LPS. SUVmean values in all organs, except the spleen, positively correlated with several serum cytokines/chemokines. In vitro measures of TSPO expression and immunofluorescent staining validated the imaging results. Noninvasive molecular imaging with [18F]DPA-714 PET in a rat model of systemic sterile inflammatory shock, along with in vitro measures of TSPO expression, showed brain, liver and lung inflammation, spleen monocytic efflux/lymphocytic activation and suggested increased bone marrow hematopoiesis. TSPO PET imaging can potentially be used to quantify SIRS and sepsis-associated organ-level immunoreactivity and assess the effectiveness of therapeutic and preventative approaches for associated organ failures, in vivo.
Collapse
Affiliation(s)
- Neysha Martinez-Orengo
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sarine Tahmazian
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jianhao Lai
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Zeping Wang
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sanhita Sinharay
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - William Schreiber-Stainthorp
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - William Reid
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Swati Shah
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Dima A. Hammoud,
| |
Collapse
|
23
|
Fogliano C, Motta CM, Venditti P, Fasciolo G, Napolitano G, Avallone B, Carotenuto R. Environmental concentrations of a delorazepam-based drug impact on embryonic development of non-target Xenopus laevis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 250:106244. [PMID: 35878487 DOI: 10.1016/j.aquatox.2022.106244] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Benzodiazepines, psychotropics drugs used for treating sleep disorders, anxiety and epilepsy, represent a major class of emerging water pollutants. As occurs for other pharmaceutical residues, they are not efficiently degraded during sewage treatment and persist in effluent waters. Bioaccumulation is already reported in fish and small crustaceans, but the impact and consequences on other "non-target" aquatic species are still unclear and nowadays of great interest. In this study, we investigated the effects of a pharmaceutical preparation containing the benzodiazepine delorazepam on the embryogenesis of Xenopus laevis, amphibian model species, taxa at high risk of exposure to water contaminants. Environmental (1 μg/L) and two higher (5 and 10 μg/L) concentrations were tested on tadpoles up to stage 45/46. Results demonstrate that delorazepam interferes with embryo development and that the effects are prevalently dose-dependent. Delorazepam reduces vitality by decreasing heart rate and motility, induces marked cephalic and abdominal edema, as well as intestinal and retinal defects. At the molecular level, delorazepam increases ROS production, modifies the expression of some master developmental genes and pro-inflammatory cytokines. The resulting stress condition significantly affects embryos' development and threatens their survival. Similar effects should be expected as well in embryos belonging to other aquatic species that have not been yet considered targets for these pharmaceutical residues.
Collapse
Affiliation(s)
- Chiara Fogliano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | - Paola Venditti
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Gianluca Fasciolo
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Gaetana Napolitano
- Department of Science and Technology, University of Naples Parthenope, Naples, Italy
| | - Bice Avallone
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | - Rosa Carotenuto
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
24
|
Wongso H. Recent progress on the development of fluorescent probes targeting the translocator protein 18 kDa (TSPO). Anal Biochem 2022; 655:114854. [PMID: 35963341 DOI: 10.1016/j.ab.2022.114854] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/06/2022] [Indexed: 11/01/2022]
Abstract
The translocator protein 18 kDa (TSPO) was first identified in 1997, and has now become one of the appealing subcellular targets in medicinal chemistry and its related fields. TSPO involves in a variety of diseases, covering neurodegenerative diseases, psychiatric disorders, cancers, and so on. To date, various high-affinity TSPO ligands labelled with single-photon emission computed tomography (SPECT)/positron emission tomography (PET) radionuclides have been reported, with some third-generation radioligands advanced to clinical trials. On the other hand, only a few number of TSPO ligands have been labelled with fluorophores for disease diagnosis. It is noteworthy that the majority of the TSPO fluorescent probes synthesised to date are based on visible fluorophores, suggesting that their applications are limited to in vitro studies, such as in vitro imaging of cancer cells, post-mortem analysis, and tissue biopsies examinations. In this context, the potential application of TSPO ligands can be broadened for in vivo investigations of human diseases by labelling with near-infrared (NIR)-fluorophores or substituting visible fluorophores with NIR-fluorophores on the currently developed fluorescent probes. In this review article, recent progress on fluorescent probes targeting the TSPO are summarised, with an emphasis on development trend in recent years and application prospects in the future.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia.
| |
Collapse
|
25
|
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
|
26
|
The mitochondrial translocator protein (TSPO): a key multifunctional molecule in the nervous system. Biochem J 2022; 479:1455-1466. [PMID: 35819398 DOI: 10.1042/bcj20220050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022]
Abstract
Translocator protein (TSPO, 18 kDa), formerly known as peripheral benzodiazepine receptor, is an evolutionary well-conserved protein located on the outer mitochondrial membrane. TSPO is involved in a variety of fundamental physiological functions and cellular processes. Its expression levels are regulated under many pathological conditions, therefore, TSPO has been proposed as a tool for diagnostic imaging and an attractive therapeutic drug target in the nervous system. Several synthetic TSPO ligands have thus been explored as agonists and antagonists for innovative treatments as neuroprotective and regenerative agents. In this review, we provide state-of-the-art knowledge of TSPO functions in the brain and peripheral nervous system. Particular emphasis is placed on its contribution to important physiological functions such as mitochondrial homeostasis, energy metabolism and steroidogenesis. We also report how it is involved in neuroinflammation, brain injury and diseases of the nervous system.
Collapse
|
27
|
Singh P, Adhikari A, Singh D, Gond C, Tiwari AK. The 18-kDa Translocator Protein PET Tracers as a Diagnostic Marker for Neuroinflammation: Development and Current Standing. ACS OMEGA 2022; 7:14412-14429. [PMID: 35557664 PMCID: PMC9089361 DOI: 10.1021/acsomega.2c00588] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/05/2022] [Indexed: 05/13/2023]
Abstract
Translocator protein (TSPO, 18 kDa) is an evolutionary, well-preserved, and tryptophan-rich 169-amino-acid protein which localizes on the contact sites between the outer and inner mitochondrial membranes of steroid-synthesizing cells. This mitochondrial protein is implicated in an extensive range of cellular activities, including steroid synthesis, cholesterol transport, apoptosis, mitochondrial respiration, and cell proliferation. The upregulation of TSPO is well documented in diverse disease conditions including neuroinflammation, cancer, brain injury, and inflammation in peripheral organs. On the basis of these outcomes, TSPO has been assumed to be a fascinating subcellular target for early stage imaging of the diseased state and for therapeutic purposes. The main outline of this Review is to give an update on dealing with the advances made in TSPO PET tracers for neuroinflammation, synchronously emphasizing the approaches applied for the design and advancement of new tracers with reference to their structure-activity relationship (SAR).
Collapse
Affiliation(s)
- Priya Singh
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Anupriya Adhikari
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Deepika Singh
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Chandraprakash Gond
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - Anjani Kumar Tiwari
- Department
of Chemistry, Babasaheb Bhimrao Ambedkar
University (A Central University), Lucknow, 226025, Uttar Pradesh, India
- Address:
Department of Chemistry,
Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh. Tel.: +91-7503381343. Fax: +91-522-2440821. E-mail:
| |
Collapse
|
28
|
Translocator Protein Ligand PIGA1138 Reduces Disease Symptoms and Severity in Experimental Autoimmune Encephalomyelitis Model of Primary Progressive Multiple Sclerosis. Mol Neurobiol 2022; 59:1744-1765. [PMID: 35018577 DOI: 10.1007/s12035-022-02737-2] [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: 10/30/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system (CNS) caused by CNS infiltration of peripheral immune cells, immune-mediated attack of the myelin sheath, neuroinflammation, and/or axonal/neuronal dysfunctions. Some drugs are available to cope with relapsing-remitting MS (RRMS) but there is no therapy for the primary progressive MS (PPMS). Because growing evidence supports a regulatory role of the translocator protein (TSPO) in neuroinflammatory, demyelinating, and neurodegenerative processes, we investigated the therapeutic potential of phenylindolyilglyoxylamydes (PIGAs) TSPO ligands in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) mice mimicking the human PPMS. MOG-EAE C57Bl/6-mice were treated by TSPO ligands PIGA839, PIGA1138, or the vehicle. Several methods were combined to evaluate PIGAs-TSPO ligand effects on MOG-EAE symptoms, CNS infiltration by immune cells, demyelination, and axonal damages. PIGA1138 (15 mg/kg) drastically reduced MOG-EAE mice clinical scores, ameliorated motor dysfunctions assessed with the Catwalk device, and counteracted MOG-EAE-induced demyelination by preserving Myelin basic protein (MBP) expression in the CNS. Furthermore, PIGA1138-treatment prevented EAE-evoked decreased neurofilament-200 expression in spinal and cerebellar axons. Moreover, PIGA1138 inhibited peripheral immune-CD45 + cell infiltration in the CNS, suggesting that it may control inflammatory mechanisms involved in PPMS. Concordantly, PIGA1138 enhanced anti-inflammatory interleukin-10 serum level in MOG-EAE mice. PIGA1138-treatment, which increased neurosteroid allopregnanolone production, ameliorated all pathological biomarkers, while PIGA839, unable to activate neurosteroidogenesis in vivo, exerted only moderate/partial effects in MOG-EAE mice. Altogether, our results suggest that PIGA1138-based treatment may represent an interesting possibility to be explored for the innovation of effective therapies against PPMS.
Collapse
|
29
|
The Neuro-Protective Effects of the TSPO Ligands CB86 and CB204 on 6-OHDA-Induced PC12 Cell Death as an In Vitro Model for Parkinson’s Disease. BIOLOGY 2021; 10:biology10111183. [PMID: 34827176 PMCID: PMC8615274 DOI: 10.3390/biology10111183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 11/21/2022]
Abstract
Simple Summary Aims and objectives: For this study, we hypothesized that the two TSPO ligands CB86 and CB204 can inhibit cellular apoptosis and necrosis by in in vitro cellular PD model of undifferentiated PC12 cells exposed to 6-hydroxydopamine (6-OHDA, 80 µM). The two TSPO ligands CB86 and CB204 seem to suppress cell death of PC12 induced by 6-OHDA. The results may be relevant to the use of these two TSPO ligands as therapeutic options for neurodegenerative diseases like Parkinson disease (PD). Results: The two ligands normalized significantly (57% and 52%, respectively, from 44%; whereas the control was 68%) cell proliferation at different time points from 0–24 h. As compared to control, the red count was increased up to 57-fold whereas CB86 and CB204 inhibited to 2.7-fold and 3.2-fold, respectively. CB86 and CB204 inhibited also normalized the cell viability up to 1.8-fold after the exposure to 6-OHDA, as assessed by XTT assay. The two TSPO ligands also inhibited apoptosis significantly (1.3-fold for both) as assessed by apopxin green staining. Conclusion: It appears that CB86, CB204, and maybe other TSPO ligands are able to slow the progression of neurodegenerative diseases like PD. Abstract Parkinson’s disease (PD) is a progressive neurodegenerative disorder which is characterized by the degeneration of dopaminergic neurons in substantia nigra (SN). Oxidative stress or reactive oxygen species (ROS) generation was suggested to play a role in this specific type of neurodegeneration. Therapeutic options which can target and counteract ROS generation may be of benefit. TSPO ligands are known to counteract with neuro-inflammation, ROS generation, apoptosis, and necrosis. In the current study, we investigated an in vitro cellular PD model by the assessment of 6-hydroxydopamine (6-OHDA, 80 µM)-induced PC12 neurotoxicity. Simultaneously to the exposure of the cells to 6-OHDA, we added the TSPO ligands CB86 and CB204 (25 µM each) and assessed the impact on several markers of cell death. The two ligands normalized significantly (57% and 52% respectively, from 44%; whereas the control was 68%) cell proliferation at different time points from 0–24 h. Additionally, we evaluated the effect of these two TSPO ligands on necrosis using propidium iodide (PI) staining and found that the ligands inhibited significantly the 6-OHDA-induced necrosis. As compared to control, the red count was increased up to 57-fold whereas CB86 and CB204 inhibited to 2.7-fold and 3.2-fold respectively. Necrosis was also analyzed by LDH assay which showed significant effect. Both assays demonstrated similar potent anti-necrotic effect of the two TSPO ligands. Reactive oxygen species (ROS) generation induced by 6-OHDA was also inhibited by the two TSPO ligand up to 1.3 and 1.5-fold respectively, as compared to 6-OHDA group. CB86 and CB204 inhibited also normalized the cell viability up to 1.8-fold after the exposure to 6-OHDA, as assessed by XTT assay. The two TSPO ligands also inhibited apoptosis significantly (1.3-fold for both) as assessed by apopxin green staining. In summary, it appears that the two TSPO ligands CB86 and CB204 can suppress cell death of PC12 induced by 6-OHDA. The results may be relevant to the use of these two TSPO ligands as therapeutic option neurodegenerative diseases like PD.
Collapse
|
30
|
Keskin E, Can EY, Aydın HA, Işık E, Özgen U, Şimşek K, Cengil O, Başar C, Kalaycı M. The preventative effect of of Ro5-4864 (peripheral benzodiazepine receptor agonist) on spinal epidural fibrosis after laminectomy in a rat model. Neurol Res 2021; 43:1107-1115. [PMID: 34461817 DOI: 10.1080/01616412.2021.1949689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To investigate the histopathological effects of a peripheral benzodiazepine receptor agonist (Ro5-4864) on epidural fibrosis (EF) in an experimental study model (post-laminectomy) in rats. METHODS A total of 32 albino Wistar rats were randomly divided into four equal groups (n = 8). In Group 1, no treatment was applied after laminectomy (control group). In Group 2, hemostasis was achieved after Laminectomy, and the surgical procedure was terminated by placing a 2-mm absorbable gelatin sponge dipped in saline into the epidural space. In Group 3, low-dose (4 mg/kg) Ro5-4864 was administered 30 minutes before the surgery. In Group 4, high-dose (8 mg/kg) Ro5-4864 was administered 30 minutes before the surgery. A histopathological examination was performed to evaluate arachnoidal invasion and EF. RESULTS Our data revealed the EF was significantly reduced in rats treated with high-dose Ro5-4864 (Group 4) compared to the control and saline-soaked Spongostan groups (p = 0.000 and p = 0.006, respectively). There was no significant difference between the groups treated with high- and low-dose Ro5-4864. Arachnoidal invasion was not seen in any of the rats in the high-dose R05-4864 group. However, the arachnoidal invasion results did not significantly differ between the study groups (p = 0.052 = 0.05). CONCLUSIONS Our study showed that Ro5-4864 could be effective in reducing EF in rats after.
Collapse
Affiliation(s)
- Emrah Keskin
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Emine Yılmaz Can
- Department of Pharmacology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Hasan Ali Aydın
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Emre Işık
- Department of Pathology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Utku Özgen
- Department of Neurosurgery, Atatürk State Hospital, Zonguldak, Turkey
| | - Kenan Şimşek
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Osman Cengil
- Department of Experimental Animal Research Laboratory, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Cansu Başar
- Insurance Information and Monitoring Center, Istanbul, Turkey
| | - Murat Kalaycı
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
31
|
Giordani A, Menziani MC, Moresco RM, Matarrese M, Paolino M, Saletti M, Giuliani G, Anzini M, Cappelli A. Exploring Translocator Protein (TSPO) Medicinal Chemistry: An Approach for Targeting Radionuclides and Boron Atoms to Mitochondria. J Med Chem 2021; 64:9649-9676. [PMID: 34254805 DOI: 10.1021/acs.jmedchem.1c00379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Translocator protein 18 kDa [TSPO or peripheral-type benzodiazepine receptor (PBR)] was identified in the search of binding sites for benzodiazepine anxiolytic drugs in peripheral regions. In these areas, binding sites for TSPO ligands were recognized in steroid-producing tissues. TSPO plays an important role in many cellular functions, and its coding sequence is highly conserved across species. TSPO is located predominantly on the membrane of mitochondria and is overexpressed in several solid cancers. TSPO basal expression in the CNS is low, but it becomes high in neurodegenerative conditions. Thus, TSPO constitutes not only as an outstanding drug target but also as a valuable marker for the diagnosis of a number of diseases. The aim of the present article is to show the lesson we have learned from our activity in TSPO medicinal chemistry and in approaching the targeted delivery to mitochondria by means of TSPO ligands.
Collapse
Affiliation(s)
- Antonio Giordani
- Rottapharm Biotech S.p.A., Via Valosa di Sopra 9, 20900 Monza, Italy
| | - Maria Cristina Menziani
- Dipartimento di Scienze Chimiche e Geologiche, Università di Modena e Reggio Emilia, Via Campi 103, 41121 Modena, Italy
| | - Rosa Maria Moresco
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Mario Matarrese
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Germano Giuliani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Maurizio Anzini
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
32
|
Scott MC, Bedi SS, Olson SD, Sears CM, Cox CS. Microglia as therapeutic targets after neurological injury: strategy for cell therapy. Expert Opin Ther Targets 2021; 25:365-380. [PMID: 34029505 DOI: 10.1080/14728222.2021.1934447] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Microglia is the resident tissue macrophages of the central nervous system. Prolonged microglial activation often occurs after traumatic brain injury and is associated with deteriorating neurocognitive outcomes. Resolution of microglial activation is associated with limited tissue loss and improved neurocognitive outcomes. Limiting the prolonged pro-inflammatory response and the associated secondary tissue injury provides the rationale and scientific premise for considering microglia as a therapeutic target. AREAS COVERED In this review, we discuss markers of microglial activation, such as immunophenotype and microglial response to injury, including cytokine/chemokine release, free radical formation, morphology, phagocytosis, and metabolic shifts. We compare the origin and role in neuroinflammation of microglia and monocytes/macrophages. We review potential therapeutic targets to shift microglial polarization. Finally, we review the effect of cell therapy on microglia. EXPERT OPINION Dysregulated microglial activation after neurologic injury, such as traumatic brain injury, can worsen tissue damage and functional outcomes. There are potential targets in microglia to attenuate this activation, such as proteins and molecules that regulate microglia polarization. Cellular therapeutics that limit, but do not eliminate, the inflammatory response have improved outcomes in animal models by reducing pro-inflammatory microglial activation via secondary signaling. These findings have been replicated in early phase clinical trials.
Collapse
Affiliation(s)
- M Collins Scott
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston (Uthealth), USA
| | - Supinder S Bedi
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Candice M Sears
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| |
Collapse
|
33
|
Sparrow EL, James S, Hussain K, Beers SA, Cragg MS, Bogdanov YD. Activation of GABA(A) receptors inhibits T cell proliferation. PLoS One 2021; 16:e0251632. [PMID: 34014994 PMCID: PMC8136847 DOI: 10.1371/journal.pone.0251632] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 04/29/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The major sites for fast synaptic inhibition in the central nervous system (CNS) are ion channels activated by γ-aminobutyric acid (GABA). These receptors are referred as GABA(A) receptors (GABA(A)R). Recent evidence indicates a role of GABA(A)R in modulating the immune response. This work aimed to discern the role of GABA and GABA(A)Rs in human and mouse T cell activity. METHODS Mouse splenocytes or human peripheral blood mononuclear cells (PBMCs) were activated with anti-CD3 antibodies and the proliferation of both CD8+ and CD4+ T cells assessed through flow cytometry. Subsequently, the effects on T cell proliferation of either GABA(A)R modulation by diazepam that is also capable of activating mitochondrial based translocator protein (TSPO), alprazolam and allopregnanolone or inhibition by bicucculine methiodide (BMI) and (1,2,5,6-Tetrahydropyridin-4-yl)methylphosphinic acid (TPMPA) were assessed. RESULTS Positive modulation of GABA(A)Rs either by benzodiazepines or the neurosteroid allopregnanolone inhibits both mouse and human T cell proliferation. GABAergic inhibition of T cell proliferation by benzodiazepines could be rescued by GABA(A)R blocking. Our data suggest that benzodiazepines influence T cell proliferation through both TSPO and GABA(A)Rs activation. CONCLUSIONS We conclude that activation of GABA(A)Rs provides immunosuppression by inhibiting T cell proliferation.
Collapse
Affiliation(s)
- Emma L. Sparrow
- Antibody and Vaccine Group, Centre for Cancer Immunology, MP127, University of Southampton Faculty of Medicine, Southampton, Hants, United Kingdom
| | - Sonya James
- Antibody and Vaccine Group, Centre for Cancer Immunology, MP127, University of Southampton Faculty of Medicine, Southampton, Hants, United Kingdom
| | - Khiyam Hussain
- Antibody and Vaccine Group, Centre for Cancer Immunology, MP127, University of Southampton Faculty of Medicine, Southampton, Hants, United Kingdom
| | - Stephen A. Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, MP127, University of Southampton Faculty of Medicine, Southampton, Hants, United Kingdom
| | - Mark S. Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, MP127, University of Southampton Faculty of Medicine, Southampton, Hants, United Kingdom
| | - Yury D. Bogdanov
- Antibody and Vaccine Group, Centre for Cancer Immunology, MP127, University of Southampton Faculty of Medicine, Southampton, Hants, United Kingdom
| |
Collapse
|
34
|
Chen S, Gao L, Li X, Ye Y. Allopregnanolone in mood disorders: Mechanism and therapeutic development. Pharmacol Res 2021; 169:105682. [PMID: 34019980 DOI: 10.1016/j.phrs.2021.105682] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 01/23/2023]
Abstract
The neuroactive steroid allopregnanolone (ALLO) is an endogenous positive allosteric modulator of GABA type A receptor (GABAAR), and the down-regulation of its biosynthesis have been attributed to the development of mood disorders, such as depression, anxiety and post-traumatic stress disorder (PTSD). ALLO mediated depression/anxiety involves GABAergic mechanisms and appears to be related to brain-derived neurotrophic factor (BDNF), dopamine receptor, glutamate neurotransmission, and Ca2+ channel. In the clinical, brexanolone, as a newly developed intravenous ALLO preparation, has been approved for the treatment of postpartum depression (PPD). In addition, traditional antidepressants such as selective serotonin reuptake inhibitor (SSRI) could reverse ALLO decline. Recently, the translocation protein (TSPO, 18 kDa), which involves in the speed-limiting step of ALLO synthesis, and ALLO derivatization have been identified as new directions for antidepressant therapy. This review provides an overview of ALLO researches in animal model and patients, discusses its role in the development and treatment of depression/anxiety, and directs its therapeutic potential in future.
Collapse
Affiliation(s)
- Shiyi Chen
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Lijuan Gao
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Xiaoyu Li
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Yiping Ye
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
35
|
Zürcher NR, Loggia ML, Mullett JE, Tseng C, Bhanot A, Richey L, Hightower BG, Wu C, Parmar AJ, Butterfield RI, Dubois JM, Chonde DB, Izquierdo-Garcia D, Wey HY, Catana C, Hadjikhani N, McDougle CJ, Hooker JM. [ 11C]PBR28 MR-PET imaging reveals lower regional brain expression of translocator protein (TSPO) in young adult males with autism spectrum disorder. Mol Psychiatry 2021; 26:1659-1669. [PMID: 32076115 PMCID: PMC8159742 DOI: 10.1038/s41380-020-0682-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 01/12/2020] [Accepted: 02/06/2020] [Indexed: 12/19/2022]
Abstract
Mechanisms of neuroimmune and mitochondrial dysfunction have been repeatedly implicated in autism spectrum disorder (ASD). To examine these mechanisms in ASD individuals, we measured the in vivo expression of the 18 kDa translocator protein (TSPO), an activated glial marker expressed on mitochondrial membranes. Participants underwent scanning on a simultaneous magnetic resonance-positron emission tomography (MR-PET) scanner with the second-generation TSPO radiotracer [11C]PBR28. By comparing TSPO in 15 young adult males with ASD with 18 age- and sex-matched controls, we showed that individuals with ASD exhibited lower regional TSPO expression in several brain regions, including the bilateral insular cortex, bilateral precuneus/posterior cingulate cortex, and bilateral temporal, angular, and supramarginal gyri, which have previously been implicated in autism in functional MR imaging studies. No brain region exhibited higher regional TSPO expression in the ASD group compared with the control group. A subset of participants underwent a second MR-PET scan after a median interscan interval of 3.6 months, and we determined that TSPO expression over this period of time was stable and replicable. Furthermore, voxelwise analysis confirmed lower regional TSPO expression in ASD at this later time point. Lower TSPO expression in ASD could reflect abnormalities in neuroimmune processes or mitochondrial dysfunction.
Collapse
Affiliation(s)
- N R Zürcher
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - M L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - J E Mullett
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - C Tseng
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - A Bhanot
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - L Richey
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - B G Hightower
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - C Wu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - A J Parmar
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - R I Butterfield
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - J M Dubois
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - D B Chonde
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - D Izquierdo-Garcia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - H Y Wey
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - C Catana
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - N Hadjikhani
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Gillberg Neuropsychiatry Center, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden
| | - C J McDougle
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - J M Hooker
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Georges E, Sottas C, Li Y, Papadopoulos V. Direct and specific binding of cholesterol to the mitochondrial translocator protein (TSPO) using PhotoClick cholesterol analogue. J Biochem 2021; 170:239-243. [PMID: 33846725 DOI: 10.1093/jb/mvab031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/18/2021] [Indexed: 11/14/2022] Open
Abstract
The translocator protein (TSPO) is a five-helix transmembrane protein localized to the outer mitochondria membrane. Radioligand binding assays and chemical crosslinking showed TSPO to be a high affinity cholesterol-binding protein. In this report, we show that TSPO in mitochondrial fractions from MA-10 mouse tumour Leydig cells can interact directly and competitively with the clickable photoreactive cholesterol analogue. PhotoClick cholesterol showed saturable photoaffinity labelling of TSPO that could be specifically immunoprecipitated with anti-TSPO antibody, following the click reaction with the fluorescent-azide probe, tetramethylrhodamine (TAMRA)-azide. Moreover, excess cholesterol reduced the photolabelling of both total mitochondrial proteins and TSPO. Together, the results of this study demonstrated direct binding of PhotoClick cholesterol to TSPO and that this interaction occurs at physiologically relevant site(s).
Collapse
Affiliation(s)
- Elias Georges
- Institute of Parasitology, McGill University, Montreal, Quebec H9X1C0, Canada.,Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Chantal Sottas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuchang Li
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
37
|
Vaht M. Variation rs6971 in the Translocator Protein Gene ( TSPO) is Associated with Aggressiveness and Impulsivity but Not with Anxiety in a Population-Representative Sample of Young Adults. The Journal of Genetic Psychology 2021; 182:149-162. [PMID: 33769215 DOI: 10.1080/00221325.2021.1896470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Expression of the 18-kDa translocator protein (TSPO), originally identified as a peripheral benzodiazepine receptor, has been found to be altered in several psychiatric disorders. A common single nucleotide polymorphism (rs6971) in the TSPO gene leads to an amino acid substitution, Ala147Thr, which dramatically alters the affinity with which TSPO binds drug ligands. As cholesterol also binds TSPO in the same transmembrane domain, it is suggested that this substitution may impair the ability of TSPO to bind or import cholesterol, and hence may affect steroid synthesis and hypothalamic-pituitary-adrenal function. The analysis was carried out on older birth cohort (n = 655) of the longitudinal Estonian Children Personality, Behavior and Health Study sample. Anxiety, aggressive behavior, impulsiveness, and history of stressful life events were self-reported in various data collection waves. Psychiatric assessment of lifetime prevalence of anxiety disorders was carried out at 25 years of age by experienced clinical psychologists. TSPO rs6971 was genotyped in all participants. TSPO rs6971 was not associated with self-reported levels of anxiety or lifetime prevalence of anxiety disorders. However, participants homozygous for the minor A allele displayed the highest aggressiveness and dysfunctional impulsivity scores. The positive, adaptive aspect of impulsivity was sensitive to stressful life events, as the AA genotype was associated with functional impulsivity only when the participants had experienced a low number of stressful life events during childhood. TSPO rs6971 polymorphism may be related to development of aggressiveness and impulsivity by adulthood, regardless of the participants' gender.
Collapse
Affiliation(s)
- Mariliis Vaht
- Institute of Psychology, University of Tartu, Tartu, Estonia.,Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
38
|
The Interplay of Cholesterol and Ligand Binding in hTSPO from Classical Molecular Dynamics Simulations. Molecules 2021; 26:molecules26051250. [PMID: 33652554 PMCID: PMC7956637 DOI: 10.3390/molecules26051250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/28/2021] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
The translocator protein (TSPO) is a 18kDa transmembrane protein, ubiquitously present in human mitochondria. It is overexpressed in tumor cells and at the sites of neuroinflammation, thus representing an important biomarker, as well as a promising drug target. In mammalian TSPO, there are cholesterol–binding motifs, as well as a binding cavity able to accommodate different chemical compounds. Given the lack of structural information for the human protein, we built a model of human (h) TSPO in the apo state and in complex with PK11195, a molecule routinely used in positron emission tomography (PET) for imaging of neuroinflammatory sites. To better understand the interactions of PK11195 and cholesterol with this pharmacologically relevant protein, we ran molecular dynamics simulations of the apo and holo proteins embedded in a model membrane. We found that: (i) PK11195 stabilizes hTSPO structural fold; (ii) PK11195 might enter in the binding site through transmembrane helices I and II of hTSPO; (iii) PK11195 reduces the frequency of cholesterol binding to the lower, N–terminal part of hTSPO in the inner membrane leaflet, while this impact is less pronounced for the upper, C–terminal part in the outer membrane leaflet, where the ligand binding site is located; (iv) very interestingly, cholesterol most frequently binds simultaneously to the so-called CRAC and CARC regions in TM V in the free form (residues L150–X–Y152–X(3)–R156 and R135–X(2)–Y138–X(2)–L141, respectively). However, when the protein is in complex with PK11195, cholesterol binds equally frequently to the CRAC–resembling motif that we observed in TM I (residues L17–X(2)–F20–X(3)–R24) and to CRAC in TM V. We expect that the CRAC–like motif in TM I will be of interest in future experimental investigations. Thus, our MD simulations provide insight into the structural features of hTSPO and the previously unknown interplay between PK11195 and cholesterol interactions with this pharmacologically relevant protein.
Collapse
|
39
|
Cui Y, Liang Y, Ip MSM, Mak JCW. Cigarette smoke induces apoptosis via 18 kDa translocator protein in human bronchial epithelial cells. Life Sci 2021; 265:118862. [PMID: 33301812 DOI: 10.1016/j.lfs.2020.118862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/20/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
AIMS The 18 kDa translocator protein (TSPO) - also known as peripheral benzodiazepine receptor, is found to be expressed in lung epithelium and pneumocytes, which is closely associated with the mitochondrial permeability transition pore (mPTP) and apoptosis. Cigarette smoking, a key risk factor for the development of chronic obstructive pulmonary disease (COPD), is known to induce apoptosis. We aimed to investigate TSPO subcellular localization and to examine whether cigarette smoke medium (CSM) induce apoptosis via TSPO in airway epithelial cells. MAIN METHODS TSPO subcellular localization and expression were evaluated using immunofluorescent staining and Western blot analysis respectively. TSPO ligands either PK 11195 (a specific antagonist) or AC-5216 (a specific agonist) were pre-incubated in human bronchial epithelial cells before treating with 2% CSM for measurements of apoptotic cells, mitochondrial membrane potential (ΔΨm), cytoplasmic/mitochondrial reactive oxygen species (ROS) and inflammatory marker interleukin (IL)-8 respectively. KEY FINDINGS TSPO was localized around the nucleus and overlapped with mitochondria in BEAS-2B cells. CSM caused an increase in apoptotic cells along with elevation of TSPO protein expression. Pretreatment of PK 11195 suppressed while AC-5216 potentiated CSM-induced apoptosis, collapse of ΔΨm, elevation of cytoplasmic/mitochondrial ROS levels and IL-8 release. In support, knockdown of TSPO caused a significant suppression of CSM-induced IL-8 release in BEAS-2B cells. SIGNIFICANCE The findings suggest that TSPO may play a crucial role in the regulation of cigarette smoke-induced mitochondrial dysfunction via mPTP. Therefore, the development of specific TSPO antagonists like PK11195 may be beneficial to combat smoking-related diseases, such as COPD.
Collapse
Affiliation(s)
- Yuting Cui
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingmin Liang
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Mary S M Ip
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Judith C W Mak
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology & Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
40
|
Dixon T, Uyar A, Ferguson-Miller S, Dickson A. Membrane-Mediated Ligand Unbinding of the PK-11195 Ligand from TSPO. Biophys J 2021; 120:158-167. [PMID: 33221248 DOI: 10.1101/2020.01.21.914127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/30/2020] [Accepted: 11/10/2020] [Indexed: 05/19/2023] Open
Abstract
The translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is of longstanding medical interest as both a biomarker for neuroinjury and a potential drug target for neuroinflammation and other disorders. Recently, it was shown that ligand residence time is a key factor determining steroidogenic efficacy of TSPO-binding compounds. This spurs interest in simulations of (un)binding pathways of TSPO ligands, which could reveal the molecular interactions governing ligand residence time. In this study, we use a weighted ensemble algorithm to determine the unbinding pathway for different poses of PK-11195, a TSPO ligand used in neuroimaging. In contrast with previous studies, our results show that PK-11195 does not dissociate directly into the solvent but instead dissociates via the lipid membrane by going between the transmembrane helices. We analyze this path ensemble in detail, constructing descriptors that can facilitate a general understanding of membrane-mediated ligand binding. We construct a set of Markov state models augmented with additional straightforward simulations to determine pose-specific ligand residence times. Together, we combine over 40 μs of trajectory data to form a coherent picture of the ligand binding landscape. We find that multiple starting poses yield residence times that roughly agree with the experimental quantity. The ligand binding transition states predicted by these Markov state models occur when PK-11195 is already in the membrane and involves only minimal ligand-protein interactions. This has implications for the design of new long-residence-time TSPO ligands.
Collapse
Affiliation(s)
- Tom Dixon
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, Michigan; Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Arzu Uyar
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Shelagh Ferguson-Miller
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Alex Dickson
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, Michigan; Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
41
|
Fan J, Papadopoulos V. Mitochondrial TSPO Deficiency Triggers Retrograde Signaling in MA-10 Mouse Tumor Leydig Cells. Int J Mol Sci 2020; 22:ijms22010252. [PMID: 33383772 PMCID: PMC7795497 DOI: 10.3390/ijms22010252] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
The mitochondrial translocator protein (TSPO) has been shown to bind cholesterol with high affinity and is involved in mediating its availability for steroidogenesis. We recently reported that targeted Tspo gene deletion in MA-10 mouse tumor Leydig cells resulted in reduced cAMP-stimulated steroid formation and significant reduction in the mitochondrial membrane potential (ΔΨm) compared to control cells. We hypothesized that ΔΨm reduction in the absence of TSPO probably reflects the dysregulation and/or maintenance failure of some basic mitochondrial function(s). To explore the consequences of TSPO depletion via CRISPR-Cas9-mediated deletion (indel) mutation in MA-10 cells, we assessed the transcriptome changes in TSPO-mutant versus wild-type (Wt) cells using RNA-seq. Gene expression profiles were validated using real-time PCR. We report herein that there are significant changes in nuclear gene expression in Tspo mutant versus Wt cells. The identified transcriptome changes were mapped to several signaling pathways including the regulation of membrane potential, calcium signaling, extracellular matrix, and phagocytosis. This is a retrograde signaling pathway from the mitochondria to the nucleus and is probably the result of changes in expression of several transcription factors, including key members of the NF-κB pathway. In conclusion, TSPO regulates nuclear gene expression through intracellular signaling. This is the first evidence of a compensatory response to the loss of TSPO with transcriptome changes at the cellular level.
Collapse
Affiliation(s)
- Jinjiang Fan
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: ; Fax: +1-323-442-1681
| |
Collapse
|
42
|
Tuominen S, Keller T, Petruk N, López-Picón F, Eichin D, Löyttyniemi E, Verhassel A, Rajander J, Sandholm J, Tuomela J, Grönroos TJ. Evaluation of [ 18F]F-DPA as a target for TSPO in head and neck cancer under normal conditions and after radiotherapy. Eur J Nucl Med Mol Imaging 2020; 48:1312-1326. [PMID: 33340054 PMCID: PMC8113193 DOI: 10.1007/s00259-020-05115-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/09/2020] [Indexed: 11/05/2022]
Abstract
Background Many malignant tumours have increased TSPO expression, which has been related to a poor prognosis. TSPO-PET tracers have not comprehensively been evaluated in peripherally located tumours. This study aimed to evaluate whether N,N-diethyl-2-(2-(4-([18F]fluoro)phenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide ([18F]F-DPA) can reflect radiotherapy (RT)-induced changes in TSPO activity in head and neck squamous cell carcinoma (HNSCC). Methods RT was used to induce inflammatory responses in HNSCC xenografts and cells. [18F]F-DPA uptake was measured in vivo in non-irradiated and irradiated tumours, followed by ex vivo biodistribution, autoradiography, and radiometabolite analysis. In vitro studies were performed in parental and TSPO-silenced (TSPO siRNA) cells. TSPO protein and mRNA expression, as well as tumour-associated macrophages (TAMs), were also assessed. Results In vivo imaging and ex vivo measurement revealed significantly higher [18F]F-DPA uptake in irradiated, compared to non-irradiated tumours. In vitro labelling studies with cells confirmed this finding, whereas no effect of RT on [18F]F-DPA uptake was detected in TSPO siRNA cells. Radiometabolite analysis showed that the amount of unchanged [18F]F-DPA in tumours was 95%, also after irradiation. PK11195 pre-treatment reduced the tumour-to-blood ratio of [18F]F-DPA by 73% in xenografts and by 88% in cells. TSPO protein and mRNA levels increased after RT, but were highly variable. The proportion of M1/M2 TAMs decreased after RT, whereas the proportion of monocytes and migratory monocytes/macrophages increased. Conclusions [18F]F-DPA can detect changes in TSPO expression levels after RT in HNSCC, which does not seem to reflect inflammation. Further studies are however needed to clarify the physiological mechanisms regulated by TSPO after RT. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-020-05115-z.
Collapse
Affiliation(s)
- Sanni Tuominen
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland.,Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Thomas Keller
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Nataliia Petruk
- Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Francisco López-Picón
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Dominik Eichin
- MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Alejandra Verhassel
- Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Johanna Tuomela
- Institute of Biomedicine and FICAN West Cancer Research Laboratory, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Tove J Grönroos
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland. .,MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland. .,Department of Oncology and Radiotherapy, Turku University Hospital, Hämeenkatu 11, FI-20520, Turku, Finland.
| |
Collapse
|
43
|
Marchetto A, Si Chaib Z, Rossi CA, Ribeiro R, Pantano S, Rossetti G, Giorgetti A. CGMD Platform: Integrated Web Servers for the Preparation, Running, and Analysis of Coarse-Grained Molecular Dynamics Simulations. Molecules 2020; 25:E5934. [PMID: 33333836 PMCID: PMC7765266 DOI: 10.3390/molecules25245934] [Citation(s) in RCA: 5] [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: 10/30/2020] [Revised: 11/21/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Advances in coarse-grained molecular dynamics (CGMD) simulations have extended the use of computational studies on biological macromolecules and their complexes, as well as the interactions of membrane protein and lipid complexes at a reduced level of representation, allowing longer and larger molecular dynamics simulations. Here, we present a computational platform dedicated to the preparation, running, and analysis of CGMD simulations. The platform is built on a completely revisited version of our Martini coarsE gRained MembrAne proteIn Dynamics (MERMAID) web server, and it integrates this with other three dedicated services. In its current version, the platform expands the existing implementation of the Martini force field for membrane proteins to also allow the simulation of soluble proteins using the Martini and the SIRAH force fields. Moreover, it offers an automated protocol for carrying out the backmapping of the coarse-grained description of the system into an atomistic one.
Collapse
Affiliation(s)
- Alessandro Marchetto
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (A.M.); (C.A.R.); (R.R.)
- Computational Biomedicine, Institute for Neuroscience and Medicine (INM-9) and Institute for Advanced Simulations (IAS-5), Forschungszentrum Jülich, 52425 Jülich, Germany;
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen University, 52062 Aachen, Germany
| | - Zeineb Si Chaib
- Computational Biomedicine, Institute for Neuroscience and Medicine (INM-9) and Institute for Advanced Simulations (IAS-5), Forschungszentrum Jülich, 52425 Jülich, Germany;
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen University, 52062 Aachen, Germany
| | - Carlo Alberto Rossi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (A.M.); (C.A.R.); (R.R.)
| | - Rui Ribeiro
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (A.M.); (C.A.R.); (R.R.)
| | - Sergio Pantano
- Institut Pasteur de Montevideo, Mataojo 2020, 11400 Montevideo, Uruguay;
| | - Giulia Rossetti
- Computational Biomedicine, Institute for Neuroscience and Medicine (INM-9) and Institute for Advanced Simulations (IAS-5), Forschungszentrum Jülich, 52425 Jülich, Germany;
- Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, 52428 Jülich, Germany
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, RWTH Aachen University, 52062 Aachen, Germany
| | - Alejandro Giorgetti
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (A.M.); (C.A.R.); (R.R.)
- Computational Biomedicine, Institute for Neuroscience and Medicine (INM-9) and Institute for Advanced Simulations (IAS-5), Forschungszentrum Jülich, 52425 Jülich, Germany;
| |
Collapse
|
44
|
Dixon T, Uyar A, Ferguson-Miller S, Dickson A. Membrane-Mediated Ligand Unbinding of the PK-11195 Ligand from TSPO. Biophys J 2020; 120:158-167. [PMID: 33221248 DOI: 10.1016/j.bpj.2020.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/30/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
The translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is of longstanding medical interest as both a biomarker for neuroinjury and a potential drug target for neuroinflammation and other disorders. Recently, it was shown that ligand residence time is a key factor determining steroidogenic efficacy of TSPO-binding compounds. This spurs interest in simulations of (un)binding pathways of TSPO ligands, which could reveal the molecular interactions governing ligand residence time. In this study, we use a weighted ensemble algorithm to determine the unbinding pathway for different poses of PK-11195, a TSPO ligand used in neuroimaging. In contrast with previous studies, our results show that PK-11195 does not dissociate directly into the solvent but instead dissociates via the lipid membrane by going between the transmembrane helices. We analyze this path ensemble in detail, constructing descriptors that can facilitate a general understanding of membrane-mediated ligand binding. We construct a set of Markov state models augmented with additional straightforward simulations to determine pose-specific ligand residence times. Together, we combine over 40 μs of trajectory data to form a coherent picture of the ligand binding landscape. We find that multiple starting poses yield residence times that roughly agree with the experimental quantity. The ligand binding transition states predicted by these Markov state models occur when PK-11195 is already in the membrane and involves only minimal ligand-protein interactions. This has implications for the design of new long-residence-time TSPO ligands.
Collapse
Affiliation(s)
- Tom Dixon
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, Michigan; Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Arzu Uyar
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Shelagh Ferguson-Miller
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Alex Dickson
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, Michigan; Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
45
|
Blevins LK, Crawford RB, Azzam DJ, Guilarte TR, Kaminski NE. Surface translocator protein 18 kDa (TSPO) localization on immune cells upon stimulation with LPS and in ART-treated HIV + subjects. J Leukoc Biol 2020; 110:123-140. [PMID: 33205494 DOI: 10.1002/jlb.3a1219-729rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 11/09/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a well-known outer mitochondrial membrane protein and it is widely used as a biomarker of neuroinflammation and brain injury. Although it is thought that TSPO plays key roles in a multitude of host cell functions, including steroid biosynthesis, apoptosis, generation of reactive oxygen species, and proliferation, some of these functions have recently been questioned. Here, we report the unexpected finding that circulating immune cells differentially express basal levels of TSPO on their cell surface, with a high percentage of monocytes and neutrophils expressing cell surface TSPO. In vitro stimulation of monocytes with LPS significantly increases the frequency of cells with surface TSPO expression in the absence of altered gene expression. Importantly, the LPS increase in TSPO cell surface expression in monocytes appears to be selective for LPS because two other distinct monocyte activators failed to increase the frequency of cells with surface TSPO. Finally, when we quantified immune cell TSPO surface expression in antiretroviral therapy-treated HIV+ donors, a chronic inflammatory disease, we found significant increases in the frequency of TSPO surface localization, which could be pharmacologically suppressed with ∆9 -tetrahydrocannabinol. These findings suggest that cell surface TSPO in circulating leukocytes could serve as a peripheral blood-based biomarker of inflammation.
Collapse
Affiliation(s)
- Lance K Blevins
- Department of Pharmacology and Toxicology, Center for Research on Ingredient Safety, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Robert B Crawford
- Department of Pharmacology and Toxicology, Center for Research on Ingredient Safety, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, Florida, USA
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, Florida, USA
| | - Norbert E Kaminski
- Department of Pharmacology and Toxicology, Center for Research on Ingredient Safety, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
46
|
Zhang H, Ma L, Guo WZ, Jiao LB, Zhao HY, Ma YQ, Hao XM. TSPO ligand etifoxine attenuates LPS-induced cognitive dysfunction in mice. Brain Res Bull 2020; 165:178-184. [PMID: 33075418 DOI: 10.1016/j.brainresbull.2020.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/14/2020] [Accepted: 10/09/2020] [Indexed: 12/19/2022]
Abstract
The translocator protein (TSPO), once known as peripheral-type benzodiazepine receptor, was reported to be related with several physiological functions. Etifoxine is a clinically available anxiolytic drug, and has recently shown neuroprotective effects as a TSPO ligand. The aim of the present study was to investigate the influence of etifoxine on LPS-induced neuroinflammation and cognitive dysfunction. C57/BL6 male mice were injected with etifoxine (50 mg/kg, i.p.) three days before lipopolysaccharide (LPS, 500 μg/kg, i.p.) administration. Etifoxine pretreatment alleviated hippocampal inflammation, increased brain levels of progesterone, allopregnanolone and attenuated cognitive dysfunction in LPS-injected mice. While LPS increased expression of caspase-3 and decreased p-Akt/Akt, etifoxine returned caspase-3 and p-Akt/Akt to control levels. Finasteride, a 5α-reductase inhibitor that blocked allopregnanolone production, partially reversed the effects of etifoxine. We concluded that etifoxine exerted neuroprotective effects in LPS-induced neuroinflammation and the neuroprotection may be related with increase of neurosteroids synthesis and decrease of apoptosis.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Neurosurgery, Air Force Medical Center of the Chinese PLA, Beijing, 100142, China.
| | - Li Ma
- Department of Anesthesiology, 7th Medical Center of the Chinese PLA General Hospital, Beijing, 100700, China
| | - Wen-Zhi Guo
- Department of Anesthesiology, 7th Medical Center of the Chinese PLA General Hospital, Beijing, 100700, China
| | - Lin-Bo Jiao
- Department of Anesthesiology, Beijing Shouda E.E.N.T Hospital, Beijing, 100070, China
| | - Hong-Yu Zhao
- Department of Anesthesiology, 7th Medical Center of the Chinese PLA General Hospital, Beijing, 100700, China
| | - Ya-Qun Ma
- Department of Anesthesiology, 7th Medical Center of the Chinese PLA General Hospital, Beijing, 100700, China
| | - Xue-Mei Hao
- Operating Room, 7th Medical Center of the Chinese PLA General Hospital, Beijing, 100700, China.
| |
Collapse
|
47
|
Russi M, Martin E, D'Autréaux B, Tixier L, Tricoire H, Monnier V. A Drosophila model of Friedreich ataxia with CRISPR/Cas9 insertion of GAA repeats in the frataxin gene reveals in vivo protection by N-acetyl cysteine. Hum Mol Genet 2020; 29:2831-2844. [PMID: 32744307 DOI: 10.1093/hmg/ddaa170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/10/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Friedreich ataxia (FA) is caused by GAA repeat expansions in the first intron of FXN, the gene encoding frataxin, which results in decreased gene expression. Thanks to the high degree of frataxin conservation, the Drosophila melanogaster fruitfly appears as an adequate animal model to study this disease and to evaluate therapeutic interventions. Here, we generated a Drosophila model of FA with CRISPR/Cas9 insertion of approximately 200 GAA in the intron of the fly frataxin gene fh. These flies exhibit a developmental delay and lethality associated with decreased frataxin expression. We were able to bypass preadult lethality using genetic tools to overexpress frataxin only during the developmental period. These frataxin-deficient adults are short-lived and present strong locomotor defects. RNA-Seq analysis identified deregulation of genes involved in amino-acid metabolism and transcriptomic signatures of oxidative stress. In particular, we observed a progressive increase of Tspo expression, fully rescued by adult frataxin expression. Thus, Tspo expression constitutes a molecular marker of the disease progression in our fly model and might be of interest in other animal models or in patients. Finally, in a candidate drug screening, we observed that N-acetyl cysteine improved the survival, locomotor function, resistance to oxidative stress and aconitase activity of frataxin-deficient flies. Therefore, our model provides the opportunity to elucidate in vivo, the protective mechanisms of this molecule of therapeutic potential. This study also highlights the strength of the CRISPR/Cas9 technology to introduce human mutations in endogenous orthologous genes, leading to Drosophila models of human diseases with improved physiological relevance.
Collapse
Affiliation(s)
- Maria Russi
- Université de Paris, BFA Unit of Functional and Adaptative Biology, UMR 8251, CNRS, Paris F-75013, France
| | - Elodie Martin
- Université de Paris, BFA Unit of Functional and Adaptative Biology, UMR 8251, CNRS, Paris F-75013, France
| | - Benoit D'Autréaux
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| | - Laura Tixier
- Université de Paris, BFA Unit of Functional and Adaptative Biology, UMR 8251, CNRS, Paris F-75013, France
| | - Hervé Tricoire
- Université de Paris, BFA Unit of Functional and Adaptative Biology, UMR 8251, CNRS, Paris F-75013, France
| | - Véronique Monnier
- Université de Paris, BFA Unit of Functional and Adaptative Biology, UMR 8251, CNRS, Paris F-75013, France
| |
Collapse
|
48
|
Hughes JL, Beech JS, Jones PS, Wang D, Menon DK, Aigbirhio FI, Fryer TD, Baron JC. Early-stage 11C-Flumazenil PET predicts day-14 selective neuronal loss in a rodent model of transient focal cerebral ischemia. J Cereb Blood Flow Metab 2020; 40:1997-2009. [PMID: 31637947 PMCID: PMC7786851 DOI: 10.1177/0271678x19883040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Predicting tissue outcome early after stroke is an important goal. MRI >3 h accurately predicts infarction but is insensitive to selective neuronal loss (SNL). Previous studies suggest that chronic-stage 11C-flumazenil PET (FMZ-PET) is a validated marker of SNL in rats, while early-stage FMZ-PET may predict infarction. Whether early FMZ-PET also predicts SNL is unknown. Following 45-min distal MCA occlusion, adult rats underwent FMZ-PET at 1 h and 48 h post-reperfusion to map distribution volume (VT), which reflects GABA-A receptor binding. NeuN immunohistochemistry was performed at Day 14. In each rat, VT and %NeuN loss were determined in 44 ROIs spanning the hemisphere. NeuN revealed isolated SNL and cortical infarction in five and one rats, respectively. In the SNL subgroup, VT-1 h was mildly reduced and only weakly predicted SNL, while VT-48 h was significantly increased and predicted SNL both individually (p < 0.01, Kendall) and across the group (p < 0.001), i.e. the higher the VT, the stronger the SNL. Similar correlations were found in the rat with infarction. Our findings suggest GABA-A receptors are still present on injured neurons at the 48 h timepoint, and the increased 48 h VT observed here is consistent with earlier rat studies showing early GABA-A receptor upregulation. That FMZ binding at 48 h was predictive of SNL may have clinical implications.
Collapse
Affiliation(s)
- Jessica L Hughes
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - John S Beech
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Dechao Wang
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - David K Menon
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jean-Claude Baron
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Inserm U1266, Paris Descartes University, Sainte-Anne Hospital, Paris, France
| |
Collapse
|
49
|
Hobson BA, Rowland DJ, Sisó S, Guignet MA, Harmany ZT, Bandara SB, Saito N, Harvey DJ, Bruun DA, Garbow JR, Chaudhari AJ, Lein PJ. TSPO PET Using [18F]PBR111 Reveals Persistent Neuroinflammation Following Acute Diisopropylfluorophosphate Intoxication in the Rat. Toxicol Sci 2020; 170:330-344. [PMID: 31087103 DOI: 10.1093/toxsci/kfz096] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Acute intoxication with organophosphates (OPs) can trigger status epilepticus followed by persistent cognitive impairment and/or electroencephalographic abnormalities. Neuroinflammation is widely posited to influence these persistent neurological consequences. However, testing this hypothesis has been challenging, in part because traditional biometrics preclude longitudinal measures of neuroinflammation within the same animal. Therefore, we evaluated the performance of noninvasive positron emission tomography (PET), using the translocator protein (TSPO) radioligand [18F]PBR111 against classic histopathologic measures of neuroinflammation in a preclinical model of acute intoxication with the OP diisopropylfluorophosphate (DFP). Adult male Sprague Dawley rats administered pyridostigmine bromide (0.1 mg/kg, im) 30 min prior to administration of DFP (4 mg/kg, sc), atropine sulfate (2 mg/kg, im) and 2-pralidoxime (25 mg/kg, im) exhibited moderate-to-severe seizure behavior. TSPO PET performed prior to DFP exposure and at 3, 7, 14, 21, and 28 days postexposure revealed distinct lesions, as defined by increased standardized uptake values (SUV). Increased SUV showed high spatial correspondence to immunohistochemical evidence of neuroinflammation, which was corroborated by cytokine gene and protein expression. Regional SUV metrics varied spatiotemporally with days postexposure and correlated with the degree of neuroinflammation detected immunohistochemically. Furthermore, SUV metrics were highly correlated with seizure severity, suggesting that early termination of OP-induced seizures may be critical for attenuating subsequent neuroinflammatory responses. Normalization of SUV values to a cerebellar reference region improved correlations to all outcome measures and seizure severity. Collectively, these results establish TSPO PET using [18F]PBR111 as a robust, noninvasive tool for longitudinal monitoring of neuroinflammation following acute OP intoxication.
Collapse
Affiliation(s)
- Brad A Hobson
- Department of Radiology, University of California Davis School of Medicine, Sacramento, California 95817
| | - Douglas J Rowland
- Center for Molecular and Genomic Imaging, Department of Biomedical Engineering, University of California Davis College of Engineering, Davis, California 95616
| | - Sílvia Sisó
- Department of Pathology, Microbiology and Immunology
| | - Michelle A Guignet
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, California 95616
| | - Zachary T Harmany
- Center for Molecular and Genomic Imaging, Department of Biomedical Engineering, University of California Davis College of Engineering, Davis, California 95616
| | - Suren B Bandara
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, California 95616
| | - Naomi Saito
- Department of Public Health Sciences, University of California Davis School of Medicine, Davis, California 95616
| | - Danielle J Harvey
- Department of Public Health Sciences, University of California Davis School of Medicine, Davis, California 95616
| | - Donald A Bruun
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, California 95616
| | - Joel R Garbow
- Department of Radiology, Washington University in St. Louis, St. Louis, Missouri 63110
| | - Abhijit J Chaudhari
- Department of Radiology, University of California Davis School of Medicine, Sacramento, California 95817.,Center for Molecular and Genomic Imaging, Department of Biomedical Engineering, University of California Davis College of Engineering, Davis, California 95616
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, California 95616
| |
Collapse
|
50
|
Colciago A, Bonalume V, Melfi V, Magnaghi V. Genomic and Non-genomic Action of Neurosteroids in the Peripheral Nervous System. Front Neurosci 2020; 14:796. [PMID: 32848567 PMCID: PMC7403499 DOI: 10.3389/fnins.2020.00796] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/07/2020] [Indexed: 01/12/2023] Open
Abstract
Since the former evidence of biologic actions of neurosteroids in the central nervous system, also the peripheral nervous system (PNS) was reported as a structure affected by these substances. Indeed, neurosteroids are synthesized and active in the PNS, exerting many important actions on the different cell types of this system. PNS is a target for neurosteroids, in their native form or as metabolites. In particular, old and recent evidence indicates that the progesterone metabolite allopregnanolone possesses important functions in the PNS, thus contributing to its physiologic processes. In this review, we will survey the more recent findings on the genomic and non-genomic actions of neurosteroids in nerves, ganglia, and cells forming the PNS, focusing on the mechanisms regulating the peripheral neuron-glial crosstalk. Then, we will refer to the physiopathological significance of the neurosteroid signaling disturbances in the PNS, in to identify new molecular targets for promising pharmacotherapeutic approaches.
Collapse
Affiliation(s)
- Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Veronica Bonalume
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valentina Melfi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|