1
|
Saha E, Shimochi S, Keller T, Eskola O, López-Picón F, Rajander J, Löyttyniemi E, Forsback S, Solin O, Grönroos TJ, Parikka V. Evaluation of PET imaging as a tool for detecting neonatal hypoxic-ischemic encephalopathy in a preclinical animal model. Exp Neurol 2024; 373:114673. [PMID: 38163475 DOI: 10.1016/j.expneurol.2023.114673] [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: 09/13/2023] [Revised: 12/09/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
Hypoxic-ischemic encephalopathy due to insufficient oxygen delivery to brain tissue is a leading cause of death or severe morbidity in neonates. The early recognition of the most severely affected individuals remains a clinical challenge. We hypothesized that hypoxic-ischemic injury can be detected using PET radiotracers for hypoxia ([18F]EF5), glucose metabolism ([18F]FDG), and inflammation ([18F]F-DPA). METHODS A preclinical model of neonatal hypoxic-ischemic brain injury was made in 9-d-old rat pups by permanent ligation of the left common carotid artery followed by hypoxia (8% oxygen and 92% nitrogen) for 120 min. In vivo PET imaging was performed immediately after injury induction or at different timepoints up to 21 d later. After imaging, ex vivo brain autoradiography was performed. Brain sections were stained with cresyl violet to evaluate the extent of the brain injury and to correlate it with [18F]FDG uptake. RESULTS PET imaging revealed that all three of the radiotracers tested had significant uptake in the injured brain hemisphere. Ex vivo autoradiography revealed high [18F]EF5 uptake in the hypoxic hemisphere immediately after the injury (P < 0.0001), decreasing to baseline even 1 d postinjury. [18F]FDG uptake was highest in the injured hemisphere on the day of injury (P < 0.0001), whereas [18F]F-DPA uptake was evident after 4 d (P = 0.029), peaking 7 d postinjury (P < 0.0001), and remained significant 21 d after the injury. Targeted evaluation demonstrated that [18F]FDG uptake measured by in vivo imaging 1 d postinjury correlated positively with the brain volume loss detected 21 d later (r = 0.72, P = 0.028). CONCLUSION Neonatal hypoxic-ischemic brain injury can be detected using PET imaging. Different types of radiotracers illustrate distinct phases of hypoxic brain damage. PET may be a new useful technique, worthy of being explored for clinical use, to predict and evaluate the course of the injury.
Collapse
Affiliation(s)
- Emma Saha
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland; MediCity Research Laboratories, University of Turku, Turku, Finland; Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland.
| | - Saeka Shimochi
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland; MediCity Research Laboratories, University of Turku, Turku, Finland
| | - Thomas Keller
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Olli Eskola
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Francisco López-Picón
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland; MediCity Research Laboratories, University of Turku, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | | | - Sarita Forsback
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Olof Solin
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland; Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland; Department of Chemistry, University of Turku, Finland
| | - Tove J Grönroos
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland; MediCity Research Laboratories, University of Turku, Turku, Finland
| | - Vilhelmiina Parikka
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland; MediCity Research Laboratories, University of Turku, Turku, Finland; Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland; InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
2
|
Ge Y, Yang J, Chen J, Dai M, Dou X, Yao S, Yao C, Lin Y. Absence in CX3CR1 receptor signaling promotes post-ischemic stroke cognitive function recovery through suppressed microglial pyroptosis in mice. CNS Neurosci Ther 2024; 30:e14551. [PMID: 38421089 PMCID: PMC10850801 DOI: 10.1111/cns.14551] [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: 08/19/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Post-stroke cognitive impairment (PSCI) is a major source of morbidity and mortality after stroke, but the pathological mechanisms remain unclear. Previous studies have demonstrated that the CX3CR1 receptor plays a crucial role in maintaining an early protective microenvironment after stroke, but whether it persistently influences cognitive dysfunction in the chronic phase requires further investigation. METHODS Mouse was used to establish a middle cerebral artery occlusion (MCAO)/reperfusion model to study PSCI. Cognitive function was assessed by the Morris water maze (MWM) and the novel object recognition test. Neurogenesis was assessed by immunofluorescence staining with Nestin+ /Ki67+ and DCX+ /BrdU+ double-positive cells. The cerebral damage was monitored by [18 F]-DPA-714 positron emission tomography, Nissel, and TTC staining. The pyroptosis was histologically, biochemically, and electron microscopically examined. RESULTS Upon MCAO, at 28 to 35 days, CX3CR1 knockout (CX3CR1-/- ) mice had better cognitive behavioral performance both in MWM and novel object recognition test than their CX3CR1+/- counterparts. Upon MCAO, at 7 days, CX3CR1-/- mice increased the numbers of Nestin+ /Ki67+ and DCX+ /BrdU+ cells, and meanwhile it decreased the protein expression of GSDMD, NLRP3 inflammasome subunit, caspase-1, mature IL-1β/IL-18, and p-P65 in the hippocampus as compared with CX3CR1+/- mice. In addition, CX3CR1-/- mice could reverse infarct volume in the hippocampus region post-stroke. CONCLUSION Our study demonstrated that CX3CR1 gene deletion was beneficial to PSCI recovery. The mechanism might lie in inhibited pyroptosis and enhanced neurogenesis. CX3CR1 receptor may serve as a therapeutic target for improving the PSCI.
Collapse
Affiliation(s)
- Yangyang Ge
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Juexi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jiayi Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Maosha Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoke Dou
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chenye Yao
- Department of Neurology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
3
|
Poblete RA, Yaceczko S, Aliakbar R, Saini P, Hazany S, Breit H, Louie SG, Lyden PD, Partikian A. Optimization of Nutrition after Brain Injury: Mechanistic and Therapeutic Considerations. Biomedicines 2023; 11:2551. [PMID: 37760993 PMCID: PMC10526443 DOI: 10.3390/biomedicines11092551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Emerging science continues to establish the detrimental effects of malnutrition in acute neurological diseases such as traumatic brain injury, stroke, status epilepticus and anoxic brain injury. The primary pathological pathways responsible for secondary brain injury include neuroinflammation, catabolism, immune suppression and metabolic failure, and these are exacerbated by malnutrition. Given this, there is growing interest in novel nutritional interventions to promote neurological recovery after acute brain injury. In this review, we will describe how malnutrition impacts the biomolecular mechanisms of secondary brain injury in acute neurological disorders, and how nutritional status can be optimized in both pediatric and adult populations. We will further highlight emerging therapeutic approaches, including specialized diets that aim to resolve neuroinflammation, immunodeficiency and metabolic crisis, by providing pre-clinical and clinical evidence that their use promotes neurologic recovery. Using nutrition as a targeted treatment is appealing for several reasons that will be discussed. Given the high mortality and both short- and long-term morbidity associated with acute brain injuries, novel translational and clinical approaches are needed.
Collapse
Affiliation(s)
- Roy A. Poblete
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Shelby Yaceczko
- UCLA Health, University of California, 100 Medical Plaza, Suite 345, Los Angeles, CA 90024, USA;
| | - Raya Aliakbar
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Pravesh Saini
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Saman Hazany
- Department of Radiology, Keck School of Medicine, The University of Southern California, 1500 San Pablo Street, Los Angeles, CA 90033, USA;
| | - Hannah Breit
- Department of Neurology, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA; (R.A.); (P.S.); (H.B.)
| | - Stan G. Louie
- Department of Clinical Pharmacy, School of Pharmacy, The University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA;
| | - Patrick D. Lyden
- Department of Neurology, Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, The University of Southern California, 1540 Alcazar Street, Suite 215, Los Angeles, CA 90033, USA;
| | - Arthur Partikian
- Department of Neurology, Department of Pediatrics, Keck School of Medicine, The University of Southern California, 2010 Zonal Avenue, Building B, 3P61, Los Angeles, CA 90033, USA;
| |
Collapse
|
4
|
Wang Z, Song Y, Bai S, Xiang W, Zhou X, Han L, Zhu D, Guan Y. Imaging of microglia in post-stroke inflammation. Nucl Med Biol 2023; 118-119:108336. [PMID: 37028196 DOI: 10.1016/j.nucmedbio.2023.108336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Microglia constantly survey the central nervous system microenvironment and maintain brain homeostasis. Microglia activation, polarization and inflammatory response are of great importance in the pathophysiology of ischemic stroke. For exploring biochemical processes in vivo, positron emission tomography (PET) is a superior imaging tool. Translocator protein 18 kDa (TSPO), is a validated neuroinflammatory biomarker which is widely used to evaluate various central nervous system (CNS) pathologies in both preclinical and clinical studies. TSPO level can be elevated due to peripheral inflammatory cells infiltration and glial cells activation. Therefore, a clear understanding of the dynamic changes between microglia and TSPO is critical for interpreting PET studies and understanding the pathophysiology after ischemic stroke. Our review discusses alternative biological targets that have attracted considerable interest for the imaging of microglia activation in recent years, and the potential value of imaging of microglia in the assessment of stroke therapies.
Collapse
|
5
|
Liu Y, Wang L, Pan D, Li M, Li Y, Wang Y, Xu Y, Wang X, Yan J, Wu Q, Lu L, Yuan K, Yang M. PET evaluation of light-induced modulation of microglial activation and GLP-1R expression in depressive rats. Transl Psychiatry 2021; 11:26. [PMID: 33414373 PMCID: PMC7791059 DOI: 10.1038/s41398-020-01155-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/27/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
Light therapy has been accepted as a promising therapeutic choice for depression. Positron emission tomography (PET) combined with specific radiotracers has great benefits for revealing pathogenesis and developing therapeutics. This study aimed to investigate the influences of light therapy on microglial activation and glucagon-like peptide-1 receptor (GLP-1R) expression in the brain of depressive rats using [18F]DPA-714 and [18F]exendin-4 PET. The results showed that chronic unpredictable mild stress (CUMS)-induced depressive rats had poorer performance in behavioral tests compared to normal rats (p < 0.05) and the depressive-like behavior could be ameliorated by light therapy. Besides, depressive rats had significantly higher [18F]DPA-714 uptake and lower [18F]FDG uptake compare to normal rats in 11 and 9 regions of interest (ROIs) of the brain, respectively (p < 0.05). After 5 weeks of light therapy, higher [18F]FDG and [18F]exendin-4 uptake was observed in most ROIs of light therapy-treated depressive rats compared to untreated depressive rats (p < 0.05) and no significant differences existed in [18F]DPA-714 uptake between the two groups. This study demonstrated that light therapy can ameliorate depressive-like behavior, improve glucose metabolism, and halt the decline of brain GLP-1R expression of depressive rats, but have no effects on microglial activation caused by CUMS. Besides, this study validated that [18F]DPA-714 and [18F]exendin-4 PET have the potential for noninvasive evaluation of microglial activation and GLP-1R expression in the brain of depression.
Collapse
Affiliation(s)
- Yu Liu
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China
| | - Lizhen Wang
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Donghui Pan
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Mingzhu Li
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Yaoqi Li
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Yan Wang
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Yuping Xu
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Xinyu Wang
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Junjie Yan
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Qiong Wu
- grid.412676.00000 0004 1799 0784NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063 Jiangsu China
| | - Lin Lu
- grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China ,grid.11135.370000 0001 2256 9319Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191 Beijing, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China.
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu, China.
| |
Collapse
|
6
|
|
7
|
Al-Khishman NU, Qi Q, Roseborough AD, Levit A, Allman BL, Anazodo UC, Fox MS, Whitehead SN, Thiessen JD. TSPO PET detects acute neuroinflammation but not diffuse chronically activated MHCII microglia in the rat. EJNMMI Res 2020; 10:113. [PMID: 32990808 PMCID: PMC7524910 DOI: 10.1186/s13550-020-00699-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
Background Accurate and sensitive imaging biomarkers are required to study the progression of white matter (WM) inflammation in neurodegenerative diseases. Radioligands targeting the translocator protein (TSPO) are considered sensitive indicators of neuroinflammation, but it is not clear how well the expression of TSPO coincides with major histocompatibility complex class II (MHCII) molecules in WM. This study aimed to test the ability of TSPO to detect activated WM microglia that are immunohistochemically positive for MHCII in rat models of prodromal Alzheimer’s disease and acute subcortical stroke. Methods Fischer 344 wild-type (n = 12) and TgAPP21 (n = 11) rats were imaged with [18F]FEPPA PET and MRI to investigate TSPO tracer uptake in the corpus callosum, a WM region known to have high levels of MHCII activated microglia in TgAPP21 rats. Wild-type rats subsequently received an endothelin-1 (ET1) subcortical stroke and were imaged at days 7 and 28 post-stroke before immunohistochemistry of TSPO, GFAP, iNOS, and the MHCII rat antigen, OX6. Results [18F]FEPPA PET was not significantly affected by genotype in WM and only detected increases near the ET1 infarct (P = 0.033, infarct/cerebellum uptake ratio: baseline = 0.94 ± 0.16; day 7 = 2.10 ± 0.78; day 28 = 1.77 ± 0.35). Immunohistochemistry confirmed that only the infarct (TSPO cells/mm2: day 7 = 555 ± 181; day 28 = 307 ± 153) and WM that is proximal to the infarct had TSPO expression (TSPO cells/mm2: day 7 = 113 ± 93; day 28 = 5 ± 7). TSPO and iNOS were not able to detect the chronic WM microglial activation that was detected with MHCII in the contralateral corpus callosum (day 28 OX6% area: saline = 0.62 ± 0.38; stroke = 4.30 ± 2.83; P = .029). Conclusion TSPO was only expressed in the stroke-induced insult and proximal tissue and therefore was unable to detect remote and non-insult-related chronically activated microglia overexpressing MHCII in WM. This suggests that research in neuroinflammation, particularly in the WM, would benefit from MHCII-sensitive radiotracers.
Collapse
Affiliation(s)
- Nassir U Al-Khishman
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada
| | - Qi Qi
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada
| | - Austyn D Roseborough
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Alexander Levit
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Brian L Allman
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Udunna C Anazodo
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Research Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Matthew S Fox
- Department of Physics and Astronomy, Western University, London, ON, Canada.,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jonathan D Thiessen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada.
| |
Collapse
|
8
|
Wang Z, Mascarenhas C, Jia X. Positron Emission Tomography After Ischemic Brain Injury: Current Challenges and Future Developments. Transl Stroke Res 2020; 11:628-642. [PMID: 31939060 PMCID: PMC7347441 DOI: 10.1007/s12975-019-00765-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/22/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022]
Abstract
Positron emission tomography (PET) is widely used in clinical and animal studies, along with the development of diverse tracers. The biochemical characteristics of PET tracers may help uncover the pathophysiological consequences of cardiac arrest (CA) and ischemic stroke, which include cerebral ischemia and reperfusion, depletion of oxygen and glucose, and neuroinflammation. PubMed was searched for studies of the application of PET for "cardiac arrest," "ischemic stroke," and "targeted temperature management." Available studies were included and classified according to the biochemical properties involved and metabolic processes of PET tracers, and were summarized. The mechanisms of ischemic brain injuries were investigated by PET with various tracers to elucidate the pathological process from the initial decrease of cerebral blood flow (CBF) to the subsequent abnormalities in energy and oxygen metabolism, to the monitoring of inflammation. In general, the trends of cerebral blood flow and oxygen metabolism after ischemic attack are not unidirectional but closely related to the time point of injury and recovery. Glucose metabolism after injury showed significant differences in different brain regions whereas global cerebral metabolic rate of glucose (CMRglc) declined. PET monitoring of neuroinflammation shows comparable efficacy to immunostaining. The technology of PET targeting in brain metabolism and the development of tracers provide new tools to track and evaluate the brain's pathological changes after ischemic brain injury. Despite no existing evidence for an available PET-based prediction method, discoveries of new tracers are expected to provide more possibilities for the whole field.
Collapse
Affiliation(s)
- Zhuoran Wang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 43007, China
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA
| | - Conrad Mascarenhas
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA.
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
9
|
Assessing the Effects of Cytoprotectants on Selective Neuronal Loss, Sensorimotor Deficit and Microglial Activation after Temporary Middle Cerebral Occlusion. Brain Sci 2019; 9:brainsci9100287. [PMID: 31652564 PMCID: PMC6827002 DOI: 10.3390/brainsci9100287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/18/2019] [Accepted: 10/20/2019] [Indexed: 01/21/2023] Open
Abstract
Although early reperfusion after stroke salvages the still-viable ischemic tissue, peri-infarct selective neuronal loss (SNL) can cause sensorimotor deficits (SMD). We designed a longitudinal protocol to assess the effects of cytoprotectants on SMD, microglial activation (MA) and SNL, and specifically tested whether the KCa3.1-blocker TRAM-34 would prevent SNL. Spontaneously hypertensive rats underwent 15 min middle-cerebral artery occlusion and were randomized into control or treatment group, which received TRAM-34 intraperitoneally for 4 weeks starting 12 h after reperfusion. SMD was assessed longitudinally using the sticky-label test. MA was quantified at day 14 using in vivo [11C]-PK111195 positron emission tomography (PET), and again across the same regions-of-interest template by immunofluorescence together with SNL at day 28. SMD recovered significantly faster in the treated group (p = 0.004). On PET, MA was present in 5/6 rats in each group, with no significant between-group difference. On immunofluorescence, both SNL and MA were present in 5/6 control rats and 4/6 TRAM-34 rats, with a non-significantly lower degree of MA but a significantly (p = 0.009) lower degree of SNL in the treated group. These findings document the utility of our longitudinal protocol and suggest that TRAM-34 reduces SNL and hastens behavioural recovery without marked MA blocking at the assessed time-points.
Collapse
|
10
|
Wang J, Li J, Wang Q, Kong Y, Zhou F, Li Q, Li W, Sun Y, Wang Y, Guan Y, Wu M, Wen T. Dcf1 Deficiency Attenuates the Role of Activated Microglia During Neuroinflammation. Front Mol Neurosci 2018; 11:256. [PMID: 30104955 PMCID: PMC6077288 DOI: 10.3389/fnmol.2018.00256] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 07/06/2018] [Indexed: 12/14/2022] Open
Abstract
Microglia serve as the principal immune cells and play crucial roles in the central nervous system, responding to neuroinflammation via migration and the execution of phagocytosis. Dendritic cell-derived factor 1 (Dcf1) is known to play an important role in neural stem cell differentiation, glioma apoptosis, dendritic spine formation, and Alzheimer’s disease (AD), nevertheless, the involvement of the Dcf1 gene in the brain immune response has not yet been reported. In the present paper, the RNA-sequencing and function enrichment analysis suggested that the majority of the down-regulated genes in Dcf1-/- (Dcf1-KO) mice are immune-related. In vivo experiments showed that Dcf1 deletion produced profound effects on microglial function, increased the expression of microglial activation markers, such as ionized calcium binding adaptor molecule 1 (Iba1), Cluster of Differentiation 68 (CD68) and translocator protein (TSPO), as well as certain proinflammatory cytokines (Cxcl1, Ccl7, and IL17D), but decreased the migratory and phagocytic abilities of microglial cells, and reduced the expression levels of some other proinflammatory cytokines (Cox-2, IL-1β, IL-6, TNF-α, and Csf1) in the mouse hippocampus. Furthermore, in vitro experiments revealed that in the absence of lipopolysaccharide (LPS), the majority of microglia were ramified and existed in a resting state, with only approximately 10% of cells exhibiting an amoeboid-like morphology, indicative of an activated state. LPS treatment dramatically increased the ratio of activated to resting cells, and Dcf1 downregulation further increased this ratio. These data indicated that Dcf1 deletion mediates neuroinflammation and induces dysfunction of activated microglia, preventing migration and the execution of phagocytosis. These findings support further investigation into the biological mechanisms underlying microglia-related neuroinflammatory diseases, and the role of Dcf1 in the immune response.
Collapse
Affiliation(s)
- Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jie Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yanyan Kong
- Positron Emission Computed Tomography Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangfang Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Weihao Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yangyang Sun
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yanli Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Yihui Guan
- Positron Emission Computed Tomography Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Minghong Wu
- Shanghai Applied Radiation Institute, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China
| | - Tieqiao Wen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
11
|
Chaney AM, Johnson EM, Cropper HC, James ML. PET Imaging of Neuroinflammation Using [11C]DPA-713 in a Mouse Model of Ischemic Stroke. J Vis Exp 2018. [PMID: 29985311 PMCID: PMC6101726 DOI: 10.3791/57243] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neuroinflammation is central to the pathological cascade following ischemic stroke. Non-invasive molecular imaging methods have the potential to provide critical insights into the temporal dynamics and role of certain neuroimmune interactions in stroke. Specifically, Positron Emission Tomography (PET) imaging of translocator protein 18 kDa (TSPO), a marker of activated microglia and peripheral myeloid-lineage cells, provides a means to detect and track neuroinflammation in vivo. Here, we present a method to accurately quantify neuroinflammation using [11C]N,N-Diethyl-2-[2-(4-methoxyphenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl]acetamide ([11C]DPA-713), a promising second generation TSPO-PET radiotracer, in distal middle cerebral artery occlusion (dMCAO) compared to sham-operated mice. MRI was performed 2 days post-dMCAO surgery to confirm stroke and define the infarct location and volume. PET/Computed Tomography (CT) imaging was carried out 6 days post-dMCAO to capture the peak increase in TSPO levels following stroke. Quantitation of PET images was conducted to assess the uptake of [11C]DPA-713 in the brain and spleen of dMCAO and sham mice to assess central and peripheral levels of inflammation. In vivo [11C]DPA-713 brain uptake was confirmed using ex vivo autoradiography.
Collapse
Affiliation(s)
| | | | | | - Michelle L James
- Department of Radiology, Stanford University; Department of Neurology and Neurological Sciences, Stanford University;
| |
Collapse
|
12
|
Mendes BM, Ferreira AV, Nascimento LTC, Ferreira SMZMD, Silveira MB, Silva JB. New Radiation Dosimetry Estimates for [ 18F]FLT based on Voxelized Phantoms. Radiat Res 2018; 190:37-44. [PMID: 29693502 DOI: 10.1667/rr14950.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
3'-Deoxy-3-[18F]fluorothymidine, or [18F]FLT, is a positron emission tomography (PET) tracer used in clinical studies for noninvasive assessment of proliferation activity in several types of cancer. Although the use of this PET tracer is expanding, to date, few studies concerning its dosimetry have been published. In this work, new [18F]FLT dosimetry estimates are determined for human and mice using Monte Carlo simulations. Modern voxelized male and female phantoms and [18F]FLT biokinetic data, both published by the ICRP, were used for simulations of human cases. For most human organs/tissues the absorbed doses were higher than those reported in ICRP Publication 128. An effective dose of 1.70E-02 mSv/MBq to the whole body was determined, which is 13.5% higher than the ICRP reference value. These new human dosimetry estimates obtained using more realistic human phantoms represent an advance in the knowledge of [18F]FLT dosimetry. In addition, mice biokinetic data were obtained experimentally. These data and a previously developed voxelized mouse phantom were used for simulations of animal cases. Concerning animal dosimetry, absorbed doses for organs/tissues ranged from 4.47 ± 0.75 to 155.74 ± 59.36 mGy/MBq. The obtained set of organ/tissue radiation doses for healthy Swiss mice is a useful tool for application in animal experiment design.
Collapse
Affiliation(s)
- B M Mendes
- Centro de Desenvolvimento da Tecnologia Nuclear CDTN/CNEN, Belo Horizonte, Minas Gerais, Brazil
| | - A V Ferreira
- Centro de Desenvolvimento da Tecnologia Nuclear CDTN/CNEN, Belo Horizonte, Minas Gerais, Brazil
| | - L T C Nascimento
- Centro de Desenvolvimento da Tecnologia Nuclear CDTN/CNEN, Belo Horizonte, Minas Gerais, Brazil
| | - S M Z M D Ferreira
- Centro de Desenvolvimento da Tecnologia Nuclear CDTN/CNEN, Belo Horizonte, Minas Gerais, Brazil
| | - M B Silveira
- Centro de Desenvolvimento da Tecnologia Nuclear CDTN/CNEN, Belo Horizonte, Minas Gerais, Brazil
| | - J B Silva
- Centro de Desenvolvimento da Tecnologia Nuclear CDTN/CNEN, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
13
|
Zinnhardt B, Wiesmann M, Honold L, Barca C, Schäfers M, Kiliaan AJ, Jacobs AH. In vivo imaging biomarkers of neuroinflammation in the development and assessment of stroke therapies - towards clinical translation. Theranostics 2018; 8:2603-2620. [PMID: 29774062 PMCID: PMC5956996 DOI: 10.7150/thno.24128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/31/2018] [Indexed: 01/01/2023] Open
Abstract
Modulation of the inflammatory microenvironment after stroke opens a new avenue for the development of novel neurorestorative therapies in stroke. Understanding the spatio-temporal profile of (neuro-)inflammatory imaging biomarkers in detail thereby represents a crucial factor in the development and application of immunomodulatory therapies. The early integration of quantitative molecular imaging biomarkers in stroke drug development may provide key information about (i) early diagnosis and follow-up, (ii) spatio-temporal drug-target engagement (pharmacodynamic biomarker), (iii) differentiation of responders and non-responders in the patient cohort (inclusion/exclusion criteria; predictive biomarkers), and (iv) the mechanism of action. The use of targeted imaging biomarkers for may thus allow clinicians to decipher the profile of patient-specific inflammatory activity and the development of patient-tailored strategies for immunomodulatory and neuro-restorative therapies in stroke. Here, we highlight the recent developments in preclinical and clinical molecular imaging biomarkers of neuroinflammation (endothelial markers, microglia, MMPs, cell labeling, future developments) in stroke and outline how imaging biomarkers can be used in overcoming current translational roadblocks and attrition in order to advance new immunomodulatory compounds within the clinical pipeline.
Collapse
Affiliation(s)
- Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- EU 7 th FP Programme “Imaging Inflammation in Neurodegenerative Diseases (INMiND)”
- Cells in Motion (CiM) Cluster of Excellence, University of Münster, Münster, Germany
- PET Imaging in Drug Design and Development (PET3D)
- Department of Nuclear Medicine, Universitätsklinikum Münster, Münster, Germany
| | - Maximilian Wiesmann
- Department of Anatomy, Radboud university medical center, Donders Institute for Brain, Cognition & Behaviour, Nijmegen, The Netherlands
| | - Lisa Honold
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
| | - Cristina Barca
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- PET Imaging in Drug Design and Development (PET3D)
| | - Michael Schäfers
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- Cells in Motion (CiM) Cluster of Excellence, University of Münster, Münster, Germany
- Department of Nuclear Medicine, Universitätsklinikum Münster, Münster, Germany
| | - Amanda J Kiliaan
- Department of Anatomy, Radboud university medical center, Donders Institute for Brain, Cognition & Behaviour, Nijmegen, The Netherlands
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms University Münster, Münster, Germany
- EU 7 th FP Programme “Imaging Inflammation in Neurodegenerative Diseases (INMiND)”
- Cells in Motion (CiM) Cluster of Excellence, University of Münster, Münster, Germany
- PET Imaging in Drug Design and Development (PET3D)
- Department of Geriatrics, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| |
Collapse
|
14
|
Evaluation of TSPO PET imaging, a marker of glial activation, to study the neuroimmune footprints of morphine exposure and withdrawal. Drug Alcohol Depend 2017; 170:43-50. [PMID: 27875800 DOI: 10.1016/j.drugalcdep.2016.10.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/12/2016] [Accepted: 10/29/2016] [Indexed: 12/13/2022]
Abstract
INTRODUCTION A growing area of research suggests that neuroimmunity may impact the pharmacology of opioids. Microglia is a key component of the brain immunity. Preclinical and clinical studies have demonstrated that microglial modulators may improve morphine-induced analgesia and prevent the development of tolerance and dependence. Positron emission tomography (PET) using translocator protein 18kDa (TSPO) radioligand is a clinically validated strategy for the non-invasive detection of microglial activation. We hypothesized that TSPO PET imaging may be used to study the neuroimmune component of opioid tolerance and withdrawal. METHODS Healthy rats (n=6 in each group) received either saline or escalating doses of morphine (10-40mg/kg) on five days to achieve tolerance and a withdrawal syndrome after morphine discontinuation. MicroPET imaging with [18F]DPA-714 was performed 60h after morphine withdrawal. Kinetic modeling was performed to estimate [18F]DPA-714 volume of distribution (VT) in several brain regions using dynamic PET images and corresponding metabolite-corrected input functions. Immunohistochemistry (IHC) experiments on striatal brain slices were performed to assess the expression of glial markers (Iba1, GFAP and CD68) during 14days after morphine discontinuation. RESULTS The baseline binding of [18F]DPA-714 to the brain (VT=0.086±0.009mLcm-3) was not increased by morphine exposure and withdrawal (VT=0.079±0.010mLcm-3) indicating the absence of TSPO overexpression, even at the regional level. Accordingly, expression of glial markers did not increase after morphine discontinuation. CONCLUSIONS Morphine tolerance and withdrawal did not detectably activate microglia and had no impact on [18F]DPA-714 brain kinetics in vivo.
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This article provides an overview of the recent literature regarding the application of in-vivo brain imaging techniques to animal models of ischemic stroke. RECENT FINDINGS Major breakthroughs concerned the effects of sensory stimulation on neuronal function, local hemodynamics, and tissue outcome in the hyperacute phase of stroke; the novel application to stroke of hybrid scanners allowing simultaneous PET and magnetic resonance; the refinements of magnetic resonance-based oxygen imaging, allowing to map the ischemic penumbra in a completely noninvasive way; the implementation of new PET ligands to selectively map poststroke neuronal death and neuroinflammation; and the use of novel mesoscale imaging techniques to demonstrate the major role of interhemispheric connectivity in poststroke plasticity and functional recovery. SUMMARY The array of techniques to map in vivo the key pathophysiological brain processes involved in stroke is currently enlarging at an amazing pace. This is paralleled by ever-increasing sophistication in postprocessing tools. The combination of techniques allowing simultaneous access to several variables is particularly powerful as it affords unprecedented insights into the intimate processes underlying the tissue and neuronal changes that follow a stroke. These major leaps forward will hopefully lead to therapeutic breakthroughs aiming at improving functional outcome after stroke.
Collapse
|
16
|
Israel I, Ohsiek A, Al-Momani E, Albert-Weissenberger C, Stetter C, Mencl S, Buck AK, Kleinschnitz C, Samnick S, Sirén AL. Combined [(18)F]DPA-714 micro-positron emission tomography and autoradiography imaging of microglia activation after closed head injury in mice. J Neuroinflammation 2016; 13:140. [PMID: 27266706 PMCID: PMC4897946 DOI: 10.1186/s12974-016-0604-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/30/2016] [Indexed: 11/27/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of death and disability. Neuroinflammation contributes to acute damage after TBI and modulates long-term evolution of degenerative and regenerative responses to injury. The aim of the present study was to evaluate the relationship of microglia activation to trauma severity, brain energy metabolism, and cellular reactions to injury in a mouse closed head injury model using combined in vivo PET imaging, ex vivo autoradiography, and immunohistochemistry. Methods A weight-drop closed head injury model was used to produce a mixed diffuse and focal TBI or a purely diffuse mild TBI (mTBI) in C57BL6 mice. Lesion severity was determined by evaluating histological damage and functional outcome using a standardized neuroscore (NSS), gliosis, and axonal injury by immunohistochemistry. Repeated intra-individual in vivo μPET imaging with the specific 18-kDa translocator protein (TSPO) radioligand [18F]DPA-714 was performed on day 1, 7, and 16 and [18F]FDG-μPET imaging for energy metabolism on days 2–5 after trauma using freshly synthesized radiotracers. Immediately after [18F]DPA-714-μPET imaging on days 7 and 16, cellular identity of the [18F]DPA-714 uptake was confirmed by exposing freshly cut cryosections to film autoradiography and successive immunostaining with antibodies against the microglia/macrophage marker IBA-1. Results Functional outcome correlated with focal brain lesions, gliosis, and axonal injury. [18F]DPA-714-μPET showed increased radiotracer uptake in focal brain lesions on days 7 and 16 after TBI and correlated with reduced cerebral [18F]FDG uptake on days 2–5, with functional outcome and number of IBA-1 positive cells on day 7. In autoradiography, [18F]DPA-714 uptake co-localized with areas of IBA1-positive staining and correlated strongly with both NSS and the number of IBA1-positive cells, gliosis, and axonal injury. After mTBI, numbers of IBA-1 positive cells with microglial morphology increased in both brain hemispheres; however, uptake of [18F]DPA-714 was not increased in autoradiography or in μPET imaging. Conclusions [18F]DPA-714 uptake in μPET/autoradiography correlates with trauma severity, brain metabolic deficits, and microglia activation after closed head TBI.
Collapse
Affiliation(s)
- Ina Israel
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Andrea Ohsiek
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Ehab Al-Momani
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christiane Albert-Weissenberger
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany.,Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christian Stetter
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Stine Mencl
- Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany.,Department of Neurology, University Hospital Essen, 45147, Essen, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Anna-Leena Sirén
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
17
|
Kong X, Luo S, Wu JR, Wu S, De Cecco CN, Schoepf UJ, Spandorfer AJ, Wang CY, Tian Y, Chen HJ, Lu GM, Yang GF, Zhang LJ. (18)F-DPA-714 PET Imaging for Detecting Neuroinflammation in Rats with Chronic Hepatic Encephalopathy. Am J Cancer Res 2016; 6:1220-31. [PMID: 27279913 PMCID: PMC4893647 DOI: 10.7150/thno.15362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/19/2016] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is considered to be the pathogenesis of hepatic encephalopathy (HE), and imaging neuroinflammation is implicated in HE management. 11C-PK11195, a typical translocator protein (TSPO) radiotracer, is used for imaging neuroinflammation. However, it has inherent limitations, such as short half-life and limited availability. The purpose of this study was to demonstrate the efficiency of new generation TSPO radiotracer, 18F-DPA-714, in detecting and monitoring neuroinflammation of chronic HE. This study was divided into two parts. The first part compared 18F-DPA-714 and 11C-PK11195 radiotracers in ten HE induced rats [bile duct ligation (BDL) and fed hyperammonemic diet (HD)] and 6 control rats. The animal subjects underwent dynamic positron emission tomography (PET) during 2-day intervals. The 11C-PK11195 PET study showed no differences in whole brain average percent injected dose per gram (%ID/g) values at all time points (all P>0.05), while the 18F-DPA-714 PET study showed higher whole brain average %ID/g values in HE rats compared to control group rats at 900 s to 3300 s after injecting radiotracer (all P<0.05). The second part of the study evaluated the effectiveness of ibuprofen (IBU) treatment to chronic HE. Forty rats were classified into six groups, including Sham+normal saline (NS), Sham+IBU, BDL+NS, BDL+HD+NS, BDL+IBU, and BDL+HD+IBU groups. 18F-DPA-714 PET was used to image neuroinflammation. Whole and regional brain average %ID/g values, neurological features, inflammatory factors and activated microglia showed better in the IBU groups than in the NS groups (all P<0.05) and no difference was seen in the Sham groups compared to IBU groups (all P>0.05). In conclusion, this study demonstrated that 18F-DPA-714 is an ideal TPSO radiotracer for imaging neuroinflammation and monitoring anti-neuroinflammation treatment efficacy of chronic HE.
Collapse
|
18
|
Janssen B, Vugts DJ, Funke U, Molenaar GT, Kruijer PS, van Berckel BNM, Lammertsma AA, Windhorst AD. Imaging of neuroinflammation in Alzheimer's disease, multiple sclerosis and stroke: Recent developments in positron emission tomography. Biochim Biophys Acta Mol Basis Dis 2015; 1862:425-41. [PMID: 26643549 DOI: 10.1016/j.bbadis.2015.11.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/09/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022]
Abstract
Neuroinflammation is thought to play a pivotal role in many diseases affecting the brain, including Alzheimer's disease, multiple sclerosis and stroke. Neuroinflammation is characterised predominantly by microglial activation, which can be visualised using positron emission tomography (PET). Traditionally, translocator protein 18kDa (TSPO) is the target for imaging of neuroinflammation using PET. In this review, recent preclinical and clinical research using PET in Alzheimer's disease, multiple sclerosis and stroke is summarised. In addition, new molecular targets for imaging of neuroinflammation, such as monoamine oxidases, adenosine receptors and cannabinoid receptor type 2, are discussed. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Bieneke Janssen
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| | - Danielle J Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Uta Funke
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; BV Cyclotron VU, Amsterdam, The Netherlands
| | - Ger T Molenaar
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; BV Cyclotron VU, Amsterdam, The Netherlands
| | | | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
|
20
|
Tóth M, Little P, Arnberg F, Häggkvist J, Mulder J, Halldin C, Gulyás B, Holmin S. Acute neuroinflammation in a clinically relevant focal cortical ischemic stroke model in rat: longitudinal positron emission tomography and immunofluorescent tracking. Brain Struct Funct 2015; 221:1279-90. [PMID: 25601153 DOI: 10.1007/s00429-014-0970-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/14/2014] [Indexed: 01/17/2023]
Abstract
Adequate estimation of neuroinflammatory processes following ischemic stroke is essential for better understanding of disease mechanisms, and for the development of treatment strategies. With the TSPO (18 kDa translocator protein) positron emission tomography (PET) radioligand [(11)C]PBR28, we monitored longitudinally the inflammatory response post-transient cerebral ischemia in rats, using a recently developed rat stroke model that produces isolated focal cortical infarcts with clinical relevance in size and pathophysiology. Six Sprague-Dawley rats were subjected to 90 min transient endovascular occlusion of the M2 segment of the middle cerebral artery (M2CAO). Animals were imaged with a nanoScan(®) PET/MRI system at 1, 4, 7 and 14 days after M2CAO with a bolus injection of [(11)C]PBR28. In the infarct region, we found a significantly increased uptake of [(11)C]PBR28 on day 4, 7 and 14 compared to day 1 as well as compared to the contralateral cortex. No significant increase was detected in the contralateral cortex during the 14 days of imaging. The activation in the infarct region gradually decreased between day 4 and day 14. In an additional group of animals (n = 26), immunofluorescence studies were performed with antibodies for activated microglia/monocytes (Cd11b), phagocytes (Cd68), astrocytes (glial fibrillary acidic protein) and TSPO. The TSPO immunofluorescence signal indicated reactive microgliosis post injury, corresponding to PET findings. The present clinically relevant animal model and TSPO PET ligand appear to be well suited for studies on neuroinflammation after ischemic stroke.
Collapse
Affiliation(s)
- Miklós Tóth
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Philip Little
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76, Stockholm, Sweden.,Department of Neuroradiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Fabian Arnberg
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76, Stockholm, Sweden.,Department of Neuroradiology, Karolinska University Hospital, 171 76, Stockholm, Sweden.,Department of Radiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Jenny Häggkvist
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Jan Mulder
- Science for Life Laboratory, Department of Neuroscience, Karolinska Institutet, 17165, Stockholm, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76, Stockholm, Sweden.,Imperial College - NTU, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 639798, Singapore
| | - Balázs Gulyás
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76, Stockholm, Sweden. .,Imperial College - NTU, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 639798, Singapore. .,Imperial College London, Faculty of Medicine, Division of Brain Sciences, London, SW7 2AZ, UK.
| | - Staffan Holmin
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76, Stockholm, Sweden.,Department of Neuroradiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
| |
Collapse
|