1
|
Chiang PT, Tsai LK, Tsai HH. New targets in spontaneous intracerebral hemorrhage. Curr Opin Neurol 2024:00019052-990000000-00197. [PMID: 39325041 DOI: 10.1097/wco.0000000000001325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
PURPOSE OF REVIEW Intracerebral hemorrhage (ICH) is a devastating stroke with limited medical treatments; thus, timely exploration of emerging therapeutic targets is essential. This review focuses on the latest strategies to mitigate secondary brain injury post-ICH other than targeting surgery or hemostasis, addressing a significant gap in clinical practice and highlighting potential improvements in patient outcomes. RECENT FINDINGS Promising therapeutic targets to reduce secondary brain injury following ICH have recently been identified, including attenuation of iron toxicity and inhibition of ferroptosis, enhancement of endogenous resorption of hematoma, and modulation of perihematomal inflammatory responses and edema. Additionally, novel insights suggest the lymphatic system of the brain may potentially play a role in hematoma clearance and edema management. Various experimental and early-phase clinical trials have demonstrated these approaches may potentially offer clinical benefits, though most research remains in the preliminary stages. SUMMARY Continued research is essential to identify multifaceted treatment strategies for ICH. Clinical translation of these emerging targets could significantly enhance the efficacy of therapeutic interventions and potentially reduce secondary brain damage and improve neurological recovery. Future efforts should focus on large-scale clinical trials to validate these approaches, to pave the way for more effective treatment protocols for spontaneous ICH.
Collapse
Affiliation(s)
- Pu-Tien Chiang
- Department of Neurology, National Taiwan University Hospital
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital
| | - Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital
| |
Collapse
|
2
|
Zhang M, Duan Y, Gan H, Jiang N, Qin L, Luo Y, Palahati A, He Y, Li C, Zhai X. TYROBP serve as potential immune-related signature genes in the acute phase of intracerebral hemorrhage. Sci Rep 2024; 14:20158. [PMID: 39215129 PMCID: PMC11364555 DOI: 10.1038/s41598-024-71132-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The development of intracerebral hemorrhage (ICH) is a dynamic process and intervention during the acute phase of ICH is critical for subsequent recovery. Therefore, it is crucial to screen potential signature genes and therapeutic target genes in the acute phase of ICH. In this study, based on the results of mRNA sequencing in mouse ICH and mRNA sequencing of human ICH from online databases, top five potential signature genes after ICH, Tyrobp, Itgb2, Tlr2, Ptprc and Itgam, were screened. Quantitative PCR results showed higher mRNA expression of Tyrobp, Itgb2, Tlr2, Ptprc, and Itgam in the 1-, 3- and 5-day mouse ICH groups compared to the sham-operated group. Immune infiltration correlation analysis shows that the top-ranked signature gene, Tyrobp, is negatively correlated with M2 macrophages and plasma cells, and Western blot analysis shows higher expression of the Tyrobp protein in the 1-, 3-, and 5-day mouse ICH groups compared to the sham-operated group. Furthermore, immunohistochemistry revealed that TYROBP protein expression was significantly higher in human ICH tissues than in normal brain tissues. Our results suggest that Tyrobp is a signature gene in the acute phase of ICH and may be a potential target for the treatment of the acute phase of ICH.
Collapse
Affiliation(s)
- Mi Zhang
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuhao Duan
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hui Gan
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ning Jiang
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China
| | - Le Qin
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yujia Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ailiyaer Palahati
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yaying He
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Chenyang Li
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xuan Zhai
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China.
| |
Collapse
|
3
|
Shang Y, Zheng L, Du Y, Shang T, Liu X, Zou W. Role of Regulatory T Cells in Intracerebral Hemorrhage. Mol Neurobiol 2024:10.1007/s12035-024-04281-7. [PMID: 38877366 DOI: 10.1007/s12035-024-04281-7] [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: 03/06/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a common cerebrovascular disease that can lead to severe neurological dysfunction in surviving patients, resulting in a heavy burden on patients and their families. When ICH occurs, the blood‒brain barrier is disrupted, thereby promoting immune cell migration into damaged brain tissue. As important immunosuppressive T cells, regulatory T (Treg) cells are involved in the maintenance of immune homeostasis and the suppression of immune responses after ICH. Treg cells mitigate brain tissue damage after ICH in a variety of ways, such as inhibiting the neuroinflammatory response, protecting against blood‒brain barrier damage, reducing oxidative stress damage and promoting nerve repair. In this review, we discuss the changes in Treg cells in ICH clinical patients and experimental animals, the mechanisms by which Treg cells regulate ICH and treatments targeting Treg cells in ICH, aiming to support new therapeutic strategies for clinical treatment.
Collapse
Affiliation(s)
- Yaxin Shang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Lei Zheng
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Yunpeng Du
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Tong Shang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Xueting Liu
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Wei Zou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China.
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China.
| |
Collapse
|
4
|
Yao S, Gao Z, Fang W, Fu Y, Xue Q, Lai T, Shangguan H, Sun W, Lin Y, Lin F, Kang D. DPA714 PET Imaging Shows That Inflammation of the Choroid Plexus Is Active in Chronic-Phase Intracerebral Hemorrhage. Clin Nucl Med 2024; 49:56-65. [PMID: 38054504 DOI: 10.1097/rlu.0000000000004948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
PURPOSE Our aims were to investigate the presence of choroid plexus (CP) inflammation in chronic-phase intracerebral hemorrhage (ICH) patients and to characterize any inflammatory cells in the CP. PATIENTS AND METHODS An in vivo 18 F-DPA714 PET study was undertaken in 22 chronic-phase ICH patients who were admitted to the First Affiliated Hospital of Fujian Medical University or Tianjin Medical University General Hospital from April 2017 to June 2020. Ten control participants with nonhemorrhagic central nervous system diseases were included. Choroid plexus 18 F-DPA714 uptake was calculated as the average SUVR. To aid the interpretation of the 18 F-DPA714 uptake results at the CP level, Cy5-DPA714 in vivo imaging and immunofluorescence staining were used to show the presence of CP inflammation in an ICH mouse model during the chronic phase (14 weeks after ICH). Then immunofluorescence staining against translocator protein and other specific biomarkers was used to characterize the cells present in the inflamed CP of ICH mice in the chronic phase. RESULTS PET imaging showed that CP DPA714 SUVRs in chronic-phase ICH patients were higher than in controls (mean CP SUVR ± SD; ICH group: 1.05 ± 0.35; control group: 0.81 ± 0.21; P = 0.006). Immunofluorescence staining of the CP in ICH model mice identified a population of CD45 + immune cells, peripheral monocyte-derived CD14 + cells, CD68 + phagocytes, and CD11b + resident microglia/macrophages expressing translocator protein, possibly contributing to the increased 18 F-DPA714 uptake. CONCLUSIONS Our study shows that CP DPA714 uptake in chronic-phase ICH patients was higher than that of participants with nonhemorrhagic central nervous system diseases, which means that CP inflammation is still active in chronic-phase ICH patients.
Collapse
Affiliation(s)
- Shaobo Yao
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | - Zhuyu Gao
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | | | - Ying Fu
- Department of Neurology, Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian
| | - Qianqian Xue
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | - Tianmin Lai
- Department of Neurology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Huangcheng Shangguan
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | - Weiwei Sun
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | | | - Fuxin Lin
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | | |
Collapse
|
5
|
Xia X, Yang Z, Zhang J, Fu X, Han B, Xiong Q, Yu A. E3 ligase Nedd4L promotes macrophage M1 polarization and exacerbates brain damage by TRAF3/TBK1 signaling pathway after ICH in mice. Immunol Lett 2023; 264:36-45. [PMID: 37940007 DOI: 10.1016/j.imlet.2023.11.002] [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: 08/08/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a serious medical problem, and promising strategy is limited. Macrophage initiated brain inflammatory injury following ICH, but the molecular mechanism had not been well identified. E3 ligase Nedd4L is implicated in the pathogenesis of the inflammatory immune response. METHODS In the present study, we detected the levels of Nedd4L in macrophages following ICH. Furthermore, Macrophage M1 polarization, pro-inflammatory cytokine production, BBB disruption, brain water content and neurological function were examined in ICH mice. RESULTS Here, we demonstrated that E3 ligase Nedd4L levels of macrophage increased following ICH, promoted M1 polarization inflammation by TRAF3. Nedd4L promoted BBB disruption, as well as neurological deficits. Inhibition of Nedd4L significantly attenuated M1 polarization in vivo. Inhibition of Nedd4L decreased TRAF3 and TBK1 levels, and subsequent phosphorylation of p38 and NF-κB p65 subunit following ICH. CONCLUSIONS Our data demonstrated that Nedd4L was involved in the pathogenesis of ICH, which promoted inflammatory responses and exacerbated brain damage by TRAF3 following ICH.
Collapse
Affiliation(s)
- Xiaohui Xia
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Zhao Yang
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Jiangwei Zhang
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Xiongjie Fu
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Bin Han
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Qijiang Xiong
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China.
| | - Anyong Yu
- Department of Emergency, Afffliated Hospital of Zunyi Medical University, Guizhou 563003, China.
| |
Collapse
|
6
|
Pokrajac NT, Tokarew NJA, Gurdita A, Ortin-Martinez A, Wallace VA. Meningeal macrophages inhibit chemokine signaling in pre-tumor cells to suppress mouse medulloblastoma initiation. Dev Cell 2023; 58:2015-2031.e8. [PMID: 37774709 DOI: 10.1016/j.devcel.2023.08.033] [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: 03/10/2023] [Revised: 07/10/2023] [Accepted: 08/30/2023] [Indexed: 10/01/2023]
Abstract
The microenvironment profoundly influences tumor initiation across numerous tissues but remains understudied in brain tumors. In the cerebellum, canonical Wnt signaling controlled by Norrin/Frizzled4 (Fzd4) activation in meningeal endothelial cells is a potent inhibitor of preneoplasia and tumor progression in mouse models of Sonic hedgehog medulloblastoma (Shh-MB). Single-cell transcriptome profiling and phenotyping of the meninges indicate that Norrin/Frizzled4 sustains the activation of meningeal macrophages (mMΦs), characterized by Lyve1 and CXCL4 expression, during the critical preneoplastic period. Depleting mMΦs during this period enhances preneoplasia and tumorigenesis, phenocopying the effects of Norrin loss. The anti-tumorigenic function of mMΦs is derived from the expression of CXCL4, which counters CXCL12/CXCR4 signaling in pre-tumor cells, thereby inhibiting cell-cycle progression and promoting migration away from the pre-tumor niche. These findings identify a pivotal role for mMΦs as key mediators in chemokine-regulated anti-cancer crosstalk between the stroma and pre-tumor cells in the control of MB initiation.
Collapse
Affiliation(s)
- Nenad T Pokrajac
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nicholas J A Tokarew
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Akshay Gurdita
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Arturo Ortin-Martinez
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Valerie A Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada.
| |
Collapse
|
7
|
Xu SY, Jia JQ, Sun M, Bao XY, Xia SN, Shu S, Liu PY, Ji SL, Ye L, Cao X, Xu Y. QHRD106 ameliorates ischemic stroke injury as a long-acting tissue kallikrein preparation. iScience 2023; 26:107268. [PMID: 37496671 PMCID: PMC10366503 DOI: 10.1016/j.isci.2023.107268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/07/2023] [Accepted: 06/28/2023] [Indexed: 07/28/2023] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide, and there are limited effective treatment strategies. QHRD106, a polyethyleneglycol (PEG)-modified long-acting tissue kallikrein preparation, has not been reported previously. In this study, we aimed to investigate the therapeutic effect of QHRD106 in ischemic stroke and its possible mechanism. We found that QHRD106 treatment alleviated brain injury after stroke via bradykinin (BK) receptor B2 (B2R) instead of BK receptor B1 (B1R). Mechanistically, QHRD106 reduced high-mobility group box 1 (HMGB1)-induced apoptosis and inflammation after ischemic stroke in vivo and in vitro. Moreover, we confirmed that QHRD106 reduced the level of acetylated HMGB1 and reduced the binding between heat shock protein 90 alpha family class A member 1 (HSP90AA1) and HMGB1, thus inhibiting the translocation and release of HMGB1. In summary, these findings indicate that QHRD106 treatment has therapeutic potential for cerebral ischemic stroke.
Collapse
Affiliation(s)
- Si-Yi Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu 210008, P.R. China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Jun-Qiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xin-Yu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, P.R. China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, P.R. China
| | - Sheng-Nan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, P.R. China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, P.R. China
| | - Shu Shu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, P.R. China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, P.R. China
| | - Pin-yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, P.R. China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, P.R. China
| | - Sen-lin Ji
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, P.R. China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, P.R. China
| | - Lei Ye
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu 210008, P.R. China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, P.R. China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, P.R. China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu 210008, P.R. China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, P.R. China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
8
|
Dysregulation of Serum MicroRNA after Intracerebral Hemorrhage in Aged Mice. Biomedicines 2023; 11:biomedicines11030822. [PMID: 36979801 PMCID: PMC10044892 DOI: 10.3390/biomedicines11030822] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 03/12/2023] Open
Abstract
Stroke is one of the most common diseases that leads to brain injury and mortality in patients, and intracerebral hemorrhage (ICH) is the most devastating subtype of stroke. Though the prevalence of ICH increases with aging, the effect of aging on the pathophysiology of ICH remains largely understudied. Moreover, there is no effective treatment for ICH. Recent studies have demonstrated the potential of circulating microRNAs as non-invasive diagnostic and prognostic biomarkers in various pathological conditions. While many studies have identified microRNAs that play roles in the pathophysiology of brain injury, few demonstrated their functions and roles after ICH. Given this significant knowledge gap, the present study aims to identify microRNAs that could serve as potential biomarkers of ICH in the elderly. To this end, sham or ICH was induced in aged C57BL/6 mice (18–24 months), and 24 h post-ICH, serum microRNAs were isolated, and expressions were analyzed. We identified 28 significantly dysregulated microRNAs between ICH and sham groups, suggesting their potential to serve as blood biomarkers of acute ICH. Among those microRNAs, based on the current literature, miR-124-3p, miR-137-5p, miR-138-5p, miR-219a-2-3p, miR-135a-5p, miR-541-5p, and miR-770-3p may serve as the most promising blood biomarker candidates of ICH, warranting further investigation.
Collapse
|
9
|
Wang J, Du Y, Wang A, Zhang X, Bian L, Lu J, Zhao X, Wang W. Systemic inflammation and immune index predicting outcomes in patients with intracerebral hemorrhage. Neurol Sci 2023:10.1007/s10072-023-06632-z. [PMID: 36813976 DOI: 10.1007/s10072-023-06632-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/18/2023] [Indexed: 02/24/2023]
Abstract
OBJECT Recent evidence has suggested that systemic inflammatory and immune index (SIRI) and systematic inflammation index (SII) could predict prognosis in stroke patients. This study aimed to determine the effects of SIRI and SII on predicting in-hospital infections and unfavorable outcomes in patients with acute intracerebral hemorrhage (ICH). METHODS We used the data from a prospective and registry-based study recruiting ICH patients between January 2014 and September 2016 in a single comprehensive stroke center. All patients were stratified by quartiles of SIRI or SII. Logistic regression analysis was used to estimate the associations with follow-up prognosis. The receiver operating characteristics (ROC) curves were performed to examine the predictive utility of these indexes for infections and prognosis. RESULTS Six hundred and forty spontaneous ICH patients were enrolled in this study. Compared with the lowest quartile (Q1), SIRI or SII values both showed positive correlations with increased risks for poor 1-month outcomes (adjusted ORs in Q4 was 2.162 [95% CI: 1.240-3.772] for SIRI, 1.797 [95% CI: 1.052-3.070] for SII). Additionally, a higher level of SIRI, but not SII, was independently associated with a higher risk of infections and an unfavorable 3-month prognosis. The C-statistic for the combined SIRI and ICH score was higher than SIRI or ICH score alone for predicting in-hospital infections and poor outcomes. CONCLUSION Elevated SIRI values were associated with in-hospital infections and poor functional outcomes. It may provide a new biomarker for ICH prognosis prediction, especially in the acute stage.
Collapse
Affiliation(s)
- Jinjin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yang Du
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiaoli Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Liheng Bian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jingjing Lu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xingquan Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China. .,China National Clinical Research Center for Neurological Diseases, Beijing, China. .,Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China. .,Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Wenjuan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4Th Ring West Road, Fengtai District, Beijing, 100070, China. .,China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
10
|
Wang J, Bian L, Du Y, Wang D, Jiang R, Lu J, Zhao X. The roles of chemokines following intracerebral hemorrhage in animal models and humans. Front Mol Neurosci 2023; 15:1091498. [PMID: 36704330 PMCID: PMC9871786 DOI: 10.3389/fnmol.2022.1091498] [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: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 01/12/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is one common yet devastating stroke subtype, imposing considerable burdens on families and society. Current guidelines are limited to symptomatic treatments after ICH, and the death rate remains significant in the acute stage. Thus, it is crucial to promote research to develop new targets on brain injury after ICH. In response to hematoma formation, amounts of chemokines are released in the brain, triggering the infiltration of resident immune cells in the brain and the chemotaxis of peripheral immune cells via the broken blood-brain barrier. During the past decades, mounting studies have focused on the roles of chemokines and their receptors in ICH injury. This review summarizes the latest advances in the study of chemokine functions in the ICH. First, we provide an overview of ICH epidemiology and underlying injury mechanisms in the pathogenesis of ICH. Second, we introduce the biology of chemokines and their receptors in brief. Third, we outline the roles of chemokines in ICH according to subgroups, including CCL2, CCL3, CCL5, CCL12, CCL17, CXCL8, CXCL12, and CX3CL1. Finally, we summarize current drug usage targeting chemokines in ICH and other cardio-cerebrovascular diseases. This review discusses the expressions of these chemokines and receptors under normal or hemorrhagic conditions and cell-specific sources. Above all, we highlight the related data of these chemokines in the progression and outcomes of the ICH disease in preclinical and clinical studies and point to therapeutic opportunities targeting chemokines productions and interactions in treating ICH, such as accelerating hematoma absorption and alleviating brain edema.
Collapse
Affiliation(s)
- Jinjin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Liheng Bian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yang Du
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dandan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ruixuan Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jingjing Lu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,*Correspondence: Jingjing Lu, ✉
| | - Xingquan Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China,Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China,Xingquan Zhao, ✉
| |
Collapse
|
11
|
TIPE2 attenuates neuroinflammation and brain injury through Bcl-2/Bax/cleaved caspase-3 apoptotic pathways after intracerebral hemorrhage in mice. Brain Res Bull 2022; 191:1-8. [PMID: 36179971 DOI: 10.1016/j.brainresbull.2022.09.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/17/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a serious disease with high mortality and morbidity, and effective treatment is limited. A large amount of evidence suggests that the inflammatory response contributes to secondary brain damage following ICH. TIPE2 is an essential negative regulator of both innate and adaptive immunity, and depletion of TIPE2 causes inflammatory disease. However, the possible role of TIPE2 following ICH has not been reported. METHODS In this study, we investigated TIPE2 levels and inflammation in microglia treated with erythrocyte lysate in vitro. In addition, we analyzed the role of Bcl-2/Bax/cleaved caspase-3 apoptotic pathways in ICH mice. Furthermore, we observed proinflammatory cytokine production, BBB disruption, cerebral water content and neurological damage in ICH mice. RESULTS We found that TIPE2 levels were significantly decreased in erythrocyte lysate-treated microglia compared to control microglia.Upregulation of TIPE2 decreased microglia activation and cytokine production and accelerated brain damage in ICH mice. Furthermore, upregulation of TIPE2 decreased the higher ratio of Blc-2/Bax and increased cleaved caspase-3 levels in ICH mice. In addition, upregulation of TIPE2 attenuated proinflammatory cytokine production, BBB disruption, and severe brain inflammation after ICH. CONCLUSION These results demonstrated that TIPE2 was negatively correlated with the pathogenesis of ICH, which prevented brain injury and attenuated deleterious inflammatory responses following ICH. TIPE2 might serve as a novel target for ICH therapy.
Collapse
|
12
|
Mo Y, Xu W, Fu K, Chen H, Wen J, Huang Q, Guo F, Mo L, Yan J. The dual function of microglial polarization and its treatment targets in ischemic stroke. Front Neurol 2022; 13:921705. [PMID: 36212660 PMCID: PMC9538667 DOI: 10.3389/fneur.2022.921705] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Stroke is the leading cause of disability and death worldwide, with ischemic stroke occurring in ~5% of the global population every year. Recently, many studies have been conducted on the inflammatory response after stroke. Microglial/macrophage polarization has a dual function and is critical to the pathology of ischemic stroke. Microglial/macrophage activation is important in reducing neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after ischemic stroke. In this review, we investigate the physiological characteristics and functions of microglia in the brain, the activation and phenotypic polarization of microglia and macrophages after stroke, the signaling mechanisms of polarization states, and the contribution of microglia to brain pathology and repair. We summarize recent advances in stroke-related microglia research, highlighting breakthroughs in therapeutic strategies for microglial responses after stroke, thereby providing new ideas for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yong Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaijing Fu
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hainan Chen
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jing Wen
- Department of Rheumatism, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qianrong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fangzhou Guo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ligen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Ligen Mo
| | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Jun Yan
| |
Collapse
|
13
|
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
|
14
|
Cakmak JD, Liu L, Poirier SE, Schaefer B, Poolacherla R, Burhan AM, Sabesan P, St. Lawrence K, Théberge J, Hicks JW, Finger E, Palaniyappan L, Anazodo UC. The functional and structural associations of aberrant microglial activity in major depressive disorder. J Psychiatry Neurosci 2022; 47:E197-E208. [PMID: 35654450 PMCID: PMC9343118 DOI: 10.1503/jpn.210124] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Accepted: 03/13/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a debilitating mental illness that has been linked to increases in markers of inflammation, as well as to changes in brain functional and structural connectivity, particularly between the insula and the subgenual anterior cingulate cortex (sgACC). In this study, we directly related inflammation and dysconnectivity in treatment-resistant MDD by concurrently measuring the following: microglial activity with [18F]N-2-(fluoroethoxyl)benzyl-N-(4phenoxypyridin-3-yl)acetamide ([18F]FEPPA) positron emission tomography (PET); the severity of MDD; and functional or structural connectivity among insula or sgACC nodes. METHODS Twelve patients with treatment-resistant MDD (8 female, 4 male; mean age ± standard deviation 54.9 ± 4.5 years and 23 healthy controls (11 female, 12 male; 60.3 ± 8.5 years) completed a hybrid [18F]FEPPA PET and MRI acquisition. From these, we extracted relative standardized uptake values for [18F]FEPPA activity and Pearson r-to-z scores representing functional connectivity from our regions of interest. We extracted diffusion tensor imaging metrics from the cingulum bundle, a key white matter bundle in MDD. We performed regressions to relate microglial activity with functional connectivity, structural connectivity and scores on the 17-item Hamilton Depression Rating Scale. RESULTS We found significantly increased [18F]FEPPA uptake in the left sgACC in patients with treatment-resistant MDD compared to healthy controls. Patients with MDD also had a reduction in connectivity between the sgACC and the insula. The [18F]FEPPA uptake in the left sgACC was significantly related to functional connectivity with the insula, and to the structural connectivity of the cingulum bundle. [18F]FEPPA uptake also predicted scores on the Hamilton Depression Rating Scale.Limitations: A relatively small sample size, lack of functional task data and concomitant medication use may have affected our findings. CONCLUSION We present preliminary evidence linking a network-level dysfunction relevant to the pathophysiology of depression and related to increased microglial activity in MDD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Lena Palaniyappan
- From the Department of Neuroscience, Western University, London, Ont. (Cakmak, Schaefer, Sabesan, Palaniyappan); the Robarts Research Institute, Western University, London, Ont. (Cakmak, Palaniyappan); the Lawson Health Research Institute, London, Ont. (Liu, Poirier, Burhan, St. Lawrence, Théberge, Hicks, Finger, Anazodo); the Department of Medical Biophysics, Western University, London, Ont. (Poirier, Sabesan, St. Lawrence, Théberge, Hicks, Anazodo); the London Health Sciences Centre, London, Ont. (Schaefer, Poolacherla, Palaniyappan); the Department of Psychiatry, Western University, London, Ont. (Burhan, Théberge, Palaniyappan); the Department of Psychiatry, University of Toronto, Toronto, Ont. (Burhan); the Ontario Shores Centre for Mental Health Sciences, Whitby, Ont. (Burhan); the Department of Clinical Neurological Sciences, Western University, London, Ont. (Finger); the Department of Anesthesia and Perioperative Medicine, Western University, London, Ont. (Poolacherla)
| | | |
Collapse
|
15
|
Watson N, Bonsack F, Sukumari-Ramesh S. Intracerebral Hemorrhage: The Effects of Aging on Brain Injury. Front Aging Neurosci 2022; 14:859067. [PMID: 35547620 PMCID: PMC9082316 DOI: 10.3389/fnagi.2022.859067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating subtype of stroke with high rates of mortality and morbidity. ICH patients often suffer devastating and debilitating neurological impairments, from which the majority of victims are unable to fully recover to functional independence. Unfortunately, there is no established medical therapy for ICH, which is partly attributed to the lack of understanding of the complex pathology of the disorder. Despite advanced age being a major risk factor of ICH, most preclinical studies on ICH employed young animal subjects. Due to this discrepancy, the molecular level changes in the aging brain after ICH are largely unknown, limiting the translation of preclinical studies into potential human treatments. The purpose of this review is to highlight the effects of advanced age on ICH- induced brain injury and recovery and to draw attention to current knowledge gaps, which warrant further investigation.
Collapse
|
16
|
An P, Zhao XC, Liu MJ, You YQ, Li JY, Gong HS. Dexmedetomidine Alleviates Intracerebral Hemorrhage-Induced Anxiety-Like Behaviors in Mice Through the Inhibition of TRPV4 Opening. Front Pharmacol 2022; 13:852401. [PMID: 35431940 PMCID: PMC9012538 DOI: 10.3389/fphar.2022.852401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
Post-stroke anxiety severely affects recovery in patients with intracerebral hemorrhage (ICH). Dexmedetomidine (Dex), a highly selective alpha 2 adrenal receptor (α2-AR) agonist, was recently found to exert an excellent protective effect against mental disorders including anxiety. The transient receptor potential vanilloid 4 (TRPV4) channel is involved in a series of diseases such as asthma, cancer, anxiety, and cardiac hypertrophy. This study examines whether Dex improved ICH-induced anxiety via the inhibition of TRPV4 channel opening. A rodent model of moderate ICH in the basal ganglia was established using autologous blood injection (20 μl). Mice were treated with Dex (25 μg/kg, intraperitoneal injection) every day for 3 days post-ICH. GSK1016790A (1 μmol/2 μl), an agonist of TRPV4, was administered via the left lateral ventricle. Thirty days post-ICH, post-stroke anxiety was evaluated by elevated plus-maze and open-field tests. Following behavioral tests, superoxide dismutase (SOD), malondialdehyde (MDA), astrocytic activation, and A1-and A2-type astrocytes were determined. Primary astrocytes were exposed to hemin to simulate ICH in vitro. Compared with sham-treated mice, Dex administration ameliorates ICH-induced decreases of distance and time in the open-arm, reduces distance and time in the central zone, increases astrocytic activation and A1-type astrocytes, elevates MDA content, downregulates total SOD contents, and decreases A2-type astrocytes. However, GSK1016790A partially reversed the neuroprotective effects of Dex. In addition, Dex significantly inhibited hemin-induced astrocytic activation in vitro. Dex improves ICH-induced anxiety-like behaviors in mice, and the mechanism might be associated with the inhibition of TRPV4-channel opening.
Collapse
Affiliation(s)
- Ping An
- Department of Neurobiology, School of Life Science, China Medical University, Shenyang, China
| | - Xiao-Chun Zhao
- Department of Anesthesiology, School and Hospital of Stomatology, China Medical University, Shenyang, China
- *Correspondence: Xiao-Chun Zhao,
| | - Man-Jia Liu
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Yu-Qing You
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Jing-Ya Li
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - He-Song Gong
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
Neuropeptidergic control of neurosteroids biosynthesis. Front Neuroendocrinol 2022; 65:100976. [PMID: 34999057 DOI: 10.1016/j.yfrne.2021.100976] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 01/14/2023]
Abstract
Neurosteroids are steroids synthesized within the central nervous system either from cholesterol or by metabolic reactions of circulating steroid hormone precursors. It has been suggested that neurosteroids exert pleiotropic activities within the central nervous system, such as organization and activation of the central nervous system and behavioral regulation. It is also increasingly becoming clear that neuropeptides exert pleiotropic activities within the central nervous system, such as modulation of neuronal functions and regulation of behavior, besides traditional neuroendocrinological functions. It was hypothesized that some of the physiological functions of neuropeptides acting within the central nervous system may be through the regulation of neurosteroids biosynthesis. Various neuropeptides reviewed in this study possibly regulate neurosteroids biosynthesis by controlling the activities of enzymes that catalyze the production of neurosteroids. It is now required to thoroughly investigate the neuropeptidergic control mechanisms of neurosteroids biosynthesis to characterize the physiological significance of this new neuroendocrinological phenomenon.
Collapse
|
18
|
Guilarte TR, Rodichkin AN, McGlothan JL, Acanda De La Rocha AM, Azzam DJ. Imaging neuroinflammation with TSPO: A new perspective on the cellular sources and subcellular localization. Pharmacol Ther 2021; 234:108048. [PMID: 34848203 DOI: 10.1016/j.pharmthera.2021.108048] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/04/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Translocator Protein 18 kDa (TSPO), previously named Peripheral Benzodiazepine Receptor, is a well-validated and widely used biomarker of neuroinflammation to assess diverse central nervous system (CNS) pathologies in preclinical and clinical studies. Many studies have shown that in animal models of human neurological and neurodegenerative disease and in the human condition, TSPO levels increase in the brain neuropil, and this increase is driven by infiltration of peripheral inflammatory cells and activation of glial cells. Therefore, a clear understanding of the dynamics of the cellular sources of the TSPO response is critically important in the interpretation of Positron Emission Tomography (PET) studies and for understanding the pathophysiology of CNS diseases. Within the normal brain compartment, there are tissues and cells such as the choroid plexus, ependymal cells of the lining of the ventricles, and vascular endothelial cells that also express TSPO at even higher levels than in glial cells. However, there is a paucity of knowledge if these cell types respond and increase TSPO in the diseased brain. These cells do provide a background signal that needs to be accounted for in TSPO-PET imaging studies. More recently, there are reports that TSPO may be expressed in neurons of the adult brain and TSPO expression may be increased by neuronal activity. Therefore, it is essential to study this topic with a great deal of detail, methodological rigor, and rule out alternative interpretations and imaging artifacts. High levels of TSPO are present in the outer mitochondrial membrane. Recent studies have provided evidence of its localization in other cellular compartments including the plasma membrane and perinuclear regions which may define functions that are different from that in mitochondria. A greater understanding of the TSPO subcellular localization in glial cells and infiltrating peripheral immune cells and associated function(s) may provide an additional layer of information to the understanding of TSPO neurobiology. This review is an effort to outline recent advances in understanding the cellular sources and subcellular localization of TSPO in brain cells and to examine remaining questions that require rigorous investigation.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America.
| | - Alexander N Rodichkin
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| | - Jennifer L McGlothan
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| | - Arlet Maria Acanda De La Rocha
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| | - Diana J Azzam
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| |
Collapse
|
19
|
Liu J, He J, Huang Y, Ge L, Xiao H, Zeng L, Jiang Z, Lu M, Hu Z. Hypoxia-preconditioned mesenchymal stem cells attenuate microglial pyroptosis after intracerebral hemorrhage. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1362. [PMID: 34733914 PMCID: PMC8506532 DOI: 10.21037/atm-21-2590] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/14/2021] [Indexed: 01/01/2023]
Abstract
Background Microglia plays a vital role in neuroinflammation, contributing to the pathogenesis of intracerebral hemorrhage (ICH)-induced brain injury. Mesenchymal stem cells (MSCs) hold great potential for treating ICH. We previously revealed that MSCs ameliorate the microglial pyroptosis caused by an ischemic stroke. However, whether MSCs can modulate microglial pyroptosis after ICH remains unknown. This study aimed to investigate the neuroprotective effects of hypoxia-preconditioned olfactory mucosa MSCs (OM-MSCs) on ICH and the possible mechanisms. Methods ICH was induced in mice via administration of collagenase IV. At 6 h post-ICH, 2-4×105 normoxic/hypoxic OM-MSCs or saline were intracerebrally administered. To evaluate the neuroprotective effects, the behavioral outcome, apoptosis, and neuronal injury were measured. Microglia activation and pro-inflammatory cytokines were applied to detect neuroinflammation. Microglial pyroptosis was determined by western blotting, immunofluorescence staining, and transmission electron microscopy (TEM). Results The two OM-MSC-transplanted groups exhibited significantly improved functional recovery and reduced neuronal injury, especially the hypoxic OM-MSCs group. Hypoxic OM-MSCs attenuated microglial activation as well as the levels of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). Moreover, we found that hypoxia-preconditioned OM-MSCs ameliorated pyroptosis by diminishing the levels of pyroptosis-associated proteins in peri-hematoma brain tissues, decreasing the expression of the microglial nod-like receptor family protein 3 (NLRP3) and caspase-1, and reducing the membrane pores on microglia post-ICH. Conclusions Our study showed that hypoxic preconditioning augments the therapeutic efficacy of OM-MSCs, and hypoxia-preconditioned OM-MSCs alleviate microglial pyroptosis in the ICH model.
Collapse
Affiliation(s)
- Jianyang Liu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jialin He
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Huang
- National Health Commission Key Laboratory of Birth Defects Research, Prevention, and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Lite Ge
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Han Xiao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Liuwang Zeng
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Jiang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Lu
- Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China.,Hunan Provincial Key Laboratory of Neurorestoratology, Second Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Positron emission tomography in multiple sclerosis - straight to the target. Nat Rev Neurol 2021; 17:663-675. [PMID: 34545219 DOI: 10.1038/s41582-021-00537-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Following the impressive progress in the treatment of relapsing-remitting multiple sclerosis (MS), the major challenge ahead is the development of treatments to prevent or delay the irreversible accumulation of clinical disability in progressive forms of the disease. The substrate of clinical progression is neuro-axonal degeneration, and a deep understanding of the mechanisms that underlie this process is a precondition for the development of therapies for progressive MS. PET imaging involves the use of radiolabelled compounds that bind to specific cellular and metabolic targets, thereby enabling direct in vivo measurement of several pathological processes. This approach can provide key insights into the clinical relevance of these processes and their chronological sequence during the disease course. In this Review, we focus on the contribution that PET is making to our understanding of extraneuronal and intraneuronal mechanisms that are involved in the pathogenesis of irreversible neuro-axonal damage in MS. We consider the major challenges with the use of PET in MS and the steps necessary to realize clinical benefits of the technique. In addition, we discuss the potential of emerging PET tracers and future applications of existing compounds to facilitate the identification of effective neuroprotective treatments for patients with MS.
Collapse
|
21
|
Translocator Protein Regulate Polarization Phenotype Transformation of Microglia after Cerebral Ischemia-reperfusion Injury. Neuroscience 2021; 480:203-216. [PMID: 34624453 DOI: 10.1016/j.neuroscience.2021.09.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022]
Abstract
Microglia cells are activated after cerebral ischemia-reperfusion injury (CIRI), playing a dual role in aggravating the injury or promoting tissue repair by polarization. Translocator protein (TSPO) is a biomarker of neuroinflammation or microglia activation. Its expression is significantly increased while brain injury and neuroinflammation occur. However, the relationship between TSPO and microglia polarization in CIRI is still not clear. In the present study, the middle cerebral artery occlusion (MCAO) methods in rats were used to simulate CIRI. We found that the expressions of M1 markers (CD86, IL-1β, and TNF-α) and M2 markers (CD206, IL-10, and TGF-β) were significantly increased. Moreover, the injection of TSPO ligand, PK11195, inhibited the increase of M1 polarization markers but promoted the expressions of M2 polarization markers, which significantly ameliorated the neurological damage after MCAO in rats. In vitro studies showed that shRNA-mediated TSPO knock-down promoted M1 polarization but inhibited M2 polarization, accompanied by a significant decrease in cell viability. On the contrary, overexpression of TSPO inhibited M1 polarization, promoted M2 polarization, and significantly improved cell viability. In summary, TSPO plays a neuroprotective role in CIRI by inhibiting M1 polarization and promoting M2 polarization, which suggests that TSPO may have the potential to serve as a therapeutic target for stroke.
Collapse
|
22
|
Feasibility of TSPO-Specific Positron Emission Tomography Radiotracer for Evaluating Paracetamol-Induced Liver Injury. Diagnostics (Basel) 2021; 11:diagnostics11091661. [PMID: 34574002 PMCID: PMC8467059 DOI: 10.3390/diagnostics11091661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 01/16/2023] Open
Abstract
Macrophages are activated during the early phase of paracetamol-induced liver injury (PLI). [18F]GE180 is a radiolabeled ligand that recognizes the macrophage translocator protein (TSPO). In this study, we evaluated the feasibility of a TSPO-specific radiotracer in a rat model of PLI. A rat model of liver injury was induced by intraperitoneal administration of paracetamol. [18F]GE180 positron emission tomography (PET) images were obtained after 24 h. The maximal and mean standardized uptake values (SUVmax and SUVav) of the liver and serum biomarker levels were examined. The TSPO expression level was examined using real-time polymerase chain reaction and Western blot analysis. [18F]GE180 hepatic uptake in the PLI group was significantly higher than that in the control group (SUVmax p = 0.001; SUVav p = 0.005). Both mRNA and protein TSPO expression levels were higher in the PLI group. The mRNA expression level of TSPO was significantly correlated with [18F]GE180 hepatic uptake in both groups (SUVmax p = 0.019; SUVav p = 0.007). [18F]GE180 hepatic uptake in the PLI group showed a significant positive correlation with ALT24 and ALT48 (ALT24 p = 0.016; ALT48p = 0.002). [18F]GE180 enabled visualization of PLI through TSPO overexpression. Our results support the potential utility of hepatic uptake by TSPO-PET as a non-invasive imaging biomarker for the early phase of PLI.
Collapse
|
23
|
Zhang Y, Long H, Wang S, Xiao W, Xiong M, Liu J, Chen L, Chen R, Wei X, Shu Y, Zeng Y, Zhang L. Genome-Wide DNA Methylation Pattern in Whole Blood Associated With Primary Intracerebral Hemorrhage. Front Immunol 2021; 12:702244. [PMID: 34484198 PMCID: PMC8414634 DOI: 10.3389/fimmu.2021.702244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Primary intracerebral hemorrhage (ICH) is a significant cause of morbidity and mortality throughout the world. ICH is a multifactorial disease that emerges from interactions among multiple genetic and environmental factors. DNA methylation plays an important role in the etiology of complex traits and diseases. We used the Illumina Infinium Human Methylation 850k BeadChip to detect changes in DNA methylation in peripheral blood samples from patients with ICH and healthy controls to explore DNA methylation patterns in ICH. Here, we compared genomic DNA methylation patterns in whole blood from ICH patients (n = 30) and controls (n = 34). The ICH and control groups showed significantly different DNA methylation patterns at 1530 sites (p-value < 5.92E-08), with 1377 hypermethylated sites and 153 hypomethylated sites in ICH patients compared to the methylation status in healthy controls. A total of 371 hypermethylated sites and 35 hypomethylated sites were in promoters, while 738 hypermethylated sites and 67 hypomethylated sites were in coding regions. Furthermore, the differentially methylated genes between ICH patients and controls were largely related to inflammatory pathways. Abnormalities in the DNA methylation pattern identified in the peripheral blood of ICH patients may play an important role in the development of ICH and warranted further investigation.
Collapse
Affiliation(s)
- Yupeng Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongyu Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Sai Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenbiao Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Meishan Xiong
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianyi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Ruijuan Chen
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xueli Wei
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Shu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Zeng
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Le Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Kashif H, Shah D, Sukumari-Ramesh S. Dysregulation of microRNA and Intracerebral Hemorrhage: Roles in Neuroinflammation. Int J Mol Sci 2021; 22:8115. [PMID: 34360881 PMCID: PMC8347974 DOI: 10.3390/ijms22158115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/23/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a major public health problem and devastating subtype of stroke with high morbidity and mortality. Notably, there is no effective treatment for ICH. Neuroinflammation, a pathological hallmark of ICH, contributes to both brain injury and repair and hence, it is regarded as a potential target for therapeutic intervention. Recent studies document that microRNAs, small non-coding RNA molecules, can regulate inflammatory brain response after ICH and are viable molecular targets to alter brain function. Therefore, there is an escalating interest in studying the role of microRNAs in the pathophysiology of ICH. Herein, we provide, for the first time, an overview of the microRNAs that play roles in ICH-induced neuroinflammation and identify the critical knowledge gap in the field, as it would help design future studies.
Collapse
Affiliation(s)
| | | | - Sangeetha Sukumari-Ramesh
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (H.K.); (D.S.)
| |
Collapse
|
25
|
Chang CW, Chiu CH, Lin MH, Wu HM, Yu TH, Wang PY, Kuo YY, Huang YY, Shiue CY, Huang WS, Yeh SHH. GMP-compliant fully automated radiosynthesis of [ 18F]FEPPA for PET/MRI imaging of regional brain TSPO expression. EJNMMI Res 2021; 11:26. [PMID: 33725191 PMCID: PMC7966678 DOI: 10.1186/s13550-021-00768-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/03/2021] [Indexed: 12/02/2022] Open
Abstract
Background Expression of translocator protein (TSPO) on the outer mitochondrial membrane of activated microglia is strongly associated with neuroinflammation. The second-generation PET ligand [18F]FEPPA specifically binds TSPO to enable in vivo visualization and quantification of neuroinflammation. We optimized a fully automated radiosynthesis method and evaluated the utility of [18F]FEPPA, the second-generation PET ligand specifically binds TSPO, in a mouse model of systemic LPS challenge to detect TSPO-associated signals of central and peripheral inflammation. In vivo dynamic PET/MR imaging was performed in LPS-induced and control mice after [18F]FEPPA administration. The relationship between the [18F]FEPPA signal and the dose of LPS was assessed. The cytokine levels (i.e., TNF-α, Il-1β, Il-6) in LPS-induced mice were measured by RT-PCR. Standard uptake value (SUV), total volume of distribution (VT) and area under the curve (AUC) were determined based on the metabolite-uncorrected plasma input function. Western blotting and immunostaining were used to measure TSPO expression in the brain. Results The fully automated [18F]FEPPA radiosynthesis produced an uncorrected radiochemical yield of 30 ± 2% within 80 min, with a radiochemical purity greater than 99% and specific activity of 148.9‒216.8 GBq/µmol. Significant differences were observed in the brain after [18F]FEPPA administration: SUV, VT and AUC were 1.61 ± 0.1, 1.25 ± 0.12 and 1.58 ± 0.09-fold higher in LPS-injected mice than controls. TNF-α, Il-1β and Il-6 mRNA levels were also elevated in the brains of LPS-injected mice. Western blotting revealed TSPO (p < 0.05) and Iba-1 (p < 0.01) were upregulated in the brain after LPS administration. In LPS-injected mice, TSPO immunoactivity colocalized with Iba-1 in the cerebrum and TSPO was significantly overexpressed in the hippocampus and cerebellum. The peripheral organs (heart, lung) of LPS-injected mice had higher [18F]FEPPA signal-to-noise ratios than control mice. Conclusions Based on the current data on ligand specificity and selectivity in central tissues using 7 T PET/MR imaging, we demonstrate that [18F]FEPPA accumulations significant increased in the specific brain regions of systemic LPS-induced neuroinflammation (5 mg/kg). Future investigations are needed to determine the sensitivity of [18F]FEPPA as a biomarker of neuroinflammation as well as the correlation between the PET signal intensity and the expression levels of TSPO. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00768-9.
Collapse
Affiliation(s)
- Chi-Wei Chang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medical Imaging and Radiological Technology, The Institute of Radiological Sciences, Tzu Chi University of Science and Technology, Hualien City, Taiwan.,Department of Biomedical Engineering and Environmental Sciences, National Tsinghua University, Hsinchu, Taiwan
| | - Chuang-Hsin Chiu
- Department of Nuclear Medicine, Tri-Service General Hospital, Taipei, Taiwan
| | - Ming-Hsien Lin
- Department of Nuclear Medicine, Cheng Hsin General Hospital, Taipei, Taiwan.,Department of Nuclear Medicine, Camillian Saint Mary's Hospital Luodong, Yilan, Taiwan
| | - Hung-Ming Wu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Tsung-Hsun Yu
- Brain Research Center, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei, 112, Taiwan
| | - Pao-Yeh Wang
- Brain Research Center, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei, 112, Taiwan
| | - Yu-Yeh Kuo
- Brain Research Center, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei, 112, Taiwan
| | - Ya-Yao Huang
- PET Center, Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan.,Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Chyng-Yann Shiue
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan.,PET Center, Department of Nuclear Medicine, Tri-Service General Hospital, Taipei, Taiwan
| | - Wen-Sheng Huang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Nuclear Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Skye Hsin-Hsien Yeh
- Brain Research Center, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei, 112, Taiwan.
| |
Collapse
|
26
|
Biosynthesis and signalling functions of central and peripheral nervous system neurosteroids in health and disease. Essays Biochem 2021; 64:591-606. [PMID: 32756865 PMCID: PMC7517341 DOI: 10.1042/ebc20200043] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Neurosteroids are steroid hormones synthesised de novo in the brain and peripheral nervous tissues. In contrast to adrenal steroid hormones that act on intracellular nuclear receptors, neurosteroids directly modulate plasma membrane ion channels and regulate intracellular signalling. This review provides an overview of the work that led to the discovery of neurosteroids, our current understanding of their intracellular biosynthetic machinery, and their roles in regulating the development and function of nervous tissue. Neurosteroids mediate signalling in the brain via multiple mechanisms. Here, we describe in detail their effects on GABA (inhibitory) and NMDA (excitatory) receptors, two signalling pathways of opposing function. Furthermore, emerging evidence points to altered neurosteroid function and signalling in neurological disease. This review focuses on neurodegenerative diseases associated with altered neurosteroid metabolism, mainly Niemann-Pick type C, multiple sclerosis and Alzheimer disease. Finally, we summarise the use of natural and synthetic neurosteroids as current and emerging therapeutics alongside their potential use as disease biomarkers.
Collapse
|
27
|
Bonsack F, Sukumari-Ramesh S. Entinostat improves acute neurological outcomes and attenuates hematoma volume after Intracerebral Hemorrhage. Brain Res 2020; 1752:147222. [PMID: 33358731 DOI: 10.1016/j.brainres.2020.147222] [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: 02/14/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
Intracerebral hemorrhage (ICH) or hemorrhagic stroke is a major public health problem with no effective treatment. Given the emerging role of epigenetic mechanisms in the pathophysiology of ICH, we tested the hypothesis that a class 1 histone deacetylase inhibitor (HDACi), Entinostat, attenuates neurodegeneration and improves neurobehavioral outcomes after ICH. To address this, we employed a preclinical mouse model of ICH and Entinostat was administered intraperitoneally one-hour post induction of ICH. Entinostat treatment significantly reduced the number of degenerating neurons and TUNEL-positive cells after ICH in comparison to vehicle-treated controls. Moreover, Entinostat treatment significantly reduced hematoma volume, T2-weighted hemorrhagic lesion volume and improved acute neurological outcomes after ICH. Further, Entinostat significantly reduced the hemin-induced release of proinflammatory cytokines in vitro. Consistently, the expression of proinflammatory microglial/macrophage marker, CD16/32, was remarkably reduced in Entinostat treated group after ICH in comparison to control. Altogether, data implicates the potential of class 1 HDACi, Entinostat, in improving acute neurological function after ICH warranting further investigation.
Collapse
Affiliation(s)
- Frederick Bonsack
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Sangeetha Sukumari-Ramesh
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
28
|
Dasari R, Bonsack F, Sukumari-Ramesh S. Brain injury and repair after intracerebral hemorrhage: The role of microglia and brain-infiltrating macrophages. Neurochem Int 2020; 142:104923. [PMID: 33248206 DOI: 10.1016/j.neuint.2020.104923] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/13/2020] [Accepted: 11/22/2020] [Indexed: 12/12/2022]
Abstract
Intracerebral hemorrhage (ICH) is a major public health problem characterized by cerebral bleeding. Despite recent advances in preclinical studies, there is no effective treatment for ICH making it the deadliest subtype of stroke. The lack of effective treatment options partly attributes to the complexity as well as poorly defined pathophysiology of ICH. The emerging evidence indicates the potential of targeting secondary brain damage and hematoma resolution for improving neurological outcomes after ICH. Herein, we provide an overview of our understanding of the functional roles of activated microglia and brain-infiltrating monocyte-derived macrophages in brain injury and repair after ICH. The clinical and preclinical aspects that we discuss in this manuscript are related to ICH that occurs in adults, but not in infants. Also, we attempt to identify the knowledge gap in the field for future functional studies given the potential of targeting microglia and brain-infiltrating macrophages for therapeutic intervention after ICH.
Collapse
Affiliation(s)
- Rajaneekar Dasari
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Frederick Bonsack
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Sangeetha Sukumari-Ramesh
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
29
|
Ren H, Han R, Chen X, Liu X, Wan J, Wang L, Yang X, Wang J. Potential therapeutic targets for intracerebral hemorrhage-associated inflammation: An update. J Cereb Blood Flow Metab 2020; 40:1752-1768. [PMID: 32423330 PMCID: PMC7446569 DOI: 10.1177/0271678x20923551] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke with high mortality and disability but no specific or effective treatment. In the last two decades, much has been learned about the pathologic mechanisms of ICH. It is now known that after ICH onset, immune and inflammatory responses contribute to blood-brain barrier disruption, edema development, and cell death processes, jointly resulting in secondary brain injury. However, the translation of potential therapies from preclinical to clinical success has been disappointing. With the development of new laboratory technology, recent progress has been made in the understanding of ICH pathomechanisms, and promising therapeutic targets have been identified. This review provides an update of recent progress on ICH and describes the prospects for further preclinical studies in this field. Our goal is to discuss new therapeutic targets and directions for the treatment of ICH and promote the effective transformation from preclinical to clinical trials.
Collapse
Affiliation(s)
- Honglei Ren
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ranran Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xuemei Chen
- Department of Human Anatomy, Basic Medical College of Zhengzhou University, Zhengzhou, China
| | - Xi Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jieru Wan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Limin Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiuli Yang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jian Wang
- Department of Human Anatomy, Basic Medical College of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Zammit M, Tao Y, Olsen ME, Metzger J, Vermilyea SC, Bjornson K, Slesarev M, Block WF, Fuchs K, Phillips S, Bondarenko V, Zhang SC, Emborg ME, Christian BT. [ 18F]FEPPA PET imaging for monitoring CD68-positive microglia/macrophage neuroinflammation in nonhuman primates. EJNMMI Res 2020; 10:93. [PMID: 32761399 PMCID: PMC7410886 DOI: 10.1186/s13550-020-00683-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The aim of this study was to examine whether the translocator protein 18-kDa (TSPO) PET ligand [18F]FEPPA has the sensitivity for detecting changes in CD68-positive microglial/macrophage activation in hemiparkinsonian rhesus macaques treated with allogeneic grafts of induced pluripotent stem cell-derived midbrain dopaminergic neurons (iPSC-mDA). METHODS In vivo positron emission tomography (PET) imaging with [18F]FEPPA was used in conjunction with postmortem CD68 immunostaining to evaluate neuroinflammation in the brains of hemiparkinsonian rhesus macaques (n = 6) that received allogeneic iPSC-mDA grafts in the putamen ipsilateral to MPTP administration. RESULTS Based on assessment of radiotracer uptake and confirmed by visual inspection of the imaging data, nonhuman primates with allogeneic grafts showed increased [18F]FEPPA binding at the graft sites relative to the contralateral putamen. From PET asymmetry analysis of the images, the mean asymmetry index of the monkeys was AI = - 0.085 ± 0.018. Evaluation and scoring of CD68 immunoreactivity by an investigator blind to the treatment identified significantly more neuroinflammation in the grafted areas of the putamen compared to the contralateral putamen (p = 0.0004). [18F]FEPPA PET AI showed a positive correlation with CD68 immunoreactivity AI ratings in the monkeys (Spearman's ρ = 0.94; p = 0.005). CONCLUSION These findings reveal that [18F]FEPPA PET is an effective marker for detecting increased CD68-positive microglial/macrophage activation and demonstrates sufficient sensitivity to detect changes in neuroinflammation in vivo following allogeneic cell engraftment.
Collapse
Affiliation(s)
- Matthew Zammit
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Yunlong Tao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Miles E Olsen
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeanette Metzger
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA
- Cellular and Molecular Pathology Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Scott C Vermilyea
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Kathryn Bjornson
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Maxim Slesarev
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Walter F Block
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Kerri Fuchs
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA
| | - Sean Phillips
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA
| | - Viktorya Bondarenko
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Marina E Emborg
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA.
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA.
- Cellular and Molecular Pathology Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA.
| | - Bradley T Christian
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
31
|
Rao R, Diharce J, Dugué B, Ostuni MA, Cadet F, Etchebest C. Versatile Dimerisation Process of Translocator Protein (TSPO) Revealed by an Extensive Sampling Based on a Coarse-Grained Dynamics Study. J Chem Inf Model 2020; 60:3944-3957. [DOI: 10.1021/acs.jcim.0c00246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Rajas Rao
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, F-75015, Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
- Université de la Réunion, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, Faculté des Sciences & Technologies Saint-Denis, F-97715 St. Denis, France
| | - Julien Diharce
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, F-75015, Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
- Université de la Réunion, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, Faculté des Sciences & Technologies Saint-Denis, F-97715 St. Denis, France
| | - Bérénice Dugué
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, F-75015, Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
- Université de la Réunion, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, Faculté des Sciences & Technologies Saint-Denis, F-97715 St. Denis, France
| | - Mariano A. Ostuni
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, F-75015, Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
| | - Frédéric Cadet
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, F-75015, Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
- Université de la Réunion, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, Faculté des Sciences & Technologies Saint-Denis, F-97715 St. Denis, France
- PEACCEL, Artificial Intelligence Department, 6 Square Albin Cachot, Box 42, 75013 Paris, France
| | - Catherine Etchebest
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, F-75015, Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
- Université de la Réunion, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, Faculté des Sciences & Technologies Saint-Denis, F-97715 St. Denis, France
| |
Collapse
|
32
|
Su Z, Chang Q, Drelich A, Shelite T, Judy B, Liu Y, Xiao J, Zhou C, He X, Jin Y, Saito T, Tang S, Soong L, Wakamiya M, Fang X, Bukreyev A, Ksiazek T, Russell WK, Gong B. Annexin A2 depletion exacerbates the intracerebral microhemorrhage induced by acute rickettsia and Ebola virus infections. PLoS Negl Trop Dis 2020; 14:e0007960. [PMID: 32687500 PMCID: PMC7392349 DOI: 10.1371/journal.pntd.0007960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 07/30/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022] Open
Abstract
Intracerebral microhemorrhages (CMHs) are small foci of hemorrhages in the cerebrum. Acute infections induced by some intracellular pathogens, including rickettsia, can result in CMHs. Annexin a2 (ANXA2) has been documented to play a functional role during intracellular bacterial adhesion. Here we report that ANXA2-knockout (KO) mice are more susceptible to CMHs in response to rickettsia and Ebola virus infections, suggesting an essential role of ANXA2 in protecting vascular integrity during these intracellular pathogen infections. Proteomic analysis via mass spectrometry of whole brain lysates and brain-derived endosomes from ANXA2-KO and wild-type (WT) mice post-infection with R. australis revealed that a variety of significant proteins were differentially expressed, and the follow-up function enrichment analysis had identified several relevant cell-cell junction functions. Immunohistology study confirmed that both infected WT and infected ANXA2-KO mice were subjected to adherens junctional protein (VE-cadherin) damages. However, key blood-brain barrier (BBB) components, tight junctional proteins ZO-1 and occludin, were disorganized in the brains from R. australis-infected ANXA2-KO mice, but not those of infected WT mice. Similar ANXA2-KO dependent CMHs and fragments of ZO-1 and occludin were also observed in Ebola virus-infected ANXA2-KO mice, but not found in infected WT mice. Overall, our study revealed a novel role of ANXA2 in the formation of CMHs during R. australis and Ebola virus infections; and the underlying mechanism is relevant to the role of ANXA2-regulated tight junctions and its role in stabilizing the BBB in these deadly infections.
Collapse
Affiliation(s)
- Zhengchen Su
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Aleksandra Drelich
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas Shelite
- Department of Internal Medicine, Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Barbara Judy
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yakun Liu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jie Xiao
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Changchen Zhou
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Xi He
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, Massachusetts, United States of America
| | - Tais Saito
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - Shaojun Tang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Galveston National Laboratory, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Maki Wakamiya
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Xiang Fang
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
33
|
Dimitrova-Shumkovska J, Krstanoski L, Veenman L. Diagnostic and Therapeutic Potential of TSPO Studies Regarding Neurodegenerative Diseases, Psychiatric Disorders, Alcohol Use Disorders, Traumatic Brain Injury, and Stroke: An Update. Cells 2020; 9:cells9040870. [PMID: 32252470 PMCID: PMC7226777 DOI: 10.3390/cells9040870] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation and cell death are among the common symptoms of many central nervous system diseases and injuries. Neuroinflammation and programmed cell death of the various cell types in the brain appear to be part of these disorders, and characteristic for each cell type, including neurons and glia cells. Concerning the effects of 18-kDa translocator protein (TSPO) on glial activation, as well as being associated with neuronal cell death, as a response mechanism to oxidative stress, the changes of its expression assayed with the aid of TSPO-specific positron emission tomography (PET) tracers' uptake could also offer evidence for following the pathogenesis of these disorders. This could potentially increase the number of diagnostic tests to accurately establish the stadium and development of the disease in question. Nonetheless, the differences in results regarding TSPO PET signals of first and second generations of tracers measured in patients with neurological disorders versus healthy controls indicate that we still have to understand more regarding TSPO characteristics. Expanding on investigations regarding the neuroprotective and healing effects of TSPO ligands could also contribute to a better understanding of the therapeutic potential of TSPO activity for brain damage due to brain injury and disease. Studies so far have directed attention to the effects on neurons and glia, and processes, such as death, inflammation, and regeneration. It is definitely worthwhile to drive such studies forward. From recent research it also appears that TSPO ligands, such as PK11195, Etifoxine, Emapunil, and 2-Cl-MGV-1, demonstrate the potential of targeting TSPO for treatments of brain diseases and disorders.
Collapse
Affiliation(s)
- Jasmina Dimitrova-Shumkovska
- Department of Experimental Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University Ss Cyril and Methodius, Arhimedova 3, P.O. Box 162, 1000 Skopje, Republic of North Macedonia;
- Correspondence: (J.D.-S.); (L.V.)
| | - Ljupcho Krstanoski
- Department of Experimental Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University Ss Cyril and Methodius, Arhimedova 3, P.O. Box 162, 1000 Skopje, Republic of North Macedonia;
| | - Leo Veenman
- Technion-Israel Institute of Technology, Faculty of Medicine, Rappaport Institute of Medical Research, 1 Efron Street, P.O. Box 9697, Haifa 31096, Israel
- Correspondence: (J.D.-S.); (L.V.)
| |
Collapse
|
34
|
Tanimoto Y, Yamasaki T, Nagoshi N, Nishiyama Y, Nori S, Nishimura S, Iida T, Ozaki M, Tsuji O, Ji B, Aoki I, Jinzaki M, Matsumoto M, Fujibayashi Y, Zhang MR, Nakamura M, Okano H. In vivo monitoring of remnant undifferentiated neural cells following human induced pluripotent stem cell-derived neural stem/progenitor cells transplantation. Stem Cells Transl Med 2020; 9:465-477. [PMID: 31904914 PMCID: PMC7103627 DOI: 10.1002/sctm.19-0150] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 11/30/2019] [Indexed: 12/16/2022] Open
Abstract
Transplantation of human-induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs) is a promising treatment for a variety of neuropathological conditions. Although previous reports have indicated the effectiveness of hiPSC-NS/PCs transplantation into the injured spinal cord of rodents and nonhuman primates, long-term observation of hiPSC-NS/PCs post-transplantation suggested some "unsafe" differentiation-resistant properties, resulting in disordered overgrowth. These findings suggest that, even if "safe" NS/PCs are transplanted into the human central nervous system (CNS), the dynamics of cellular differentiation of stem cells should be noninvasively tracked to ensure safety. Positron emission tomography (PET) provides molecular-functional information and helps to detect specific disease conditions. The current study was conducted to visualize Nestin (an NS/PC marker)-positive undifferentiated neural cells in the CNS of immune-deficient (nonobese diabetic-severe combined immune-deficient) mice after hiPSC-NS/PCs transplantation with PET, using 18 kDa translocator protein (TSPO) ligands as labels. TSPO was recently found to be expressed in rodent NS/PCs, and its expression decreased with the progression of neuronal differentiation. We hypothesized that TSPO would also be present in hiPSC-NS/PCs and expressed strongly in residual immature neural cells after transplantation. The results showed high levels of TSPO expression in immature hiPSC-NS/PCs-derived cells, and decreased TSPO expression as neural differentiation progressed in vitro. Furthermore, PET with [18 F] FEDAC (a TSPO radioligand) was able to visualize the remnant undifferentiated hiPSC-NS/PCs-derived cells consisting of TSPO and Nestin+ cells in vivo. These findings suggest that PET with [18 F] FEDAC could play a key role in the safe clinical application of CNS repair in regenerative medicine.
Collapse
Affiliation(s)
- Yuji Tanimoto
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuichiro Nishiyama
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Nori
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Soraya Nishimura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Iida
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Ozaki
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Osahiko Tsuji
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Bin Ji
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Ichio Aoki
- Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasuhisa Fujibayashi
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
A Combined Proteomics and Bioinformatics Approach Reveals Novel Signaling Pathways and Molecular Targets After Intracerebral Hemorrhage. J Mol Neurosci 2020; 70:1186-1197. [PMID: 32170712 PMCID: PMC7359136 DOI: 10.1007/s12031-020-01526-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/28/2020] [Indexed: 12/18/2022]
Abstract
Intracerebral hemorrhage (ICH) is a non-traumatic cerebrovascular disorder with very high morbidity and mortality and regarded as one of the deadliest stroke subtypes. Notably, there is no effective treatment for ICH. Despite an overall increase in preclinical studies, the pathophysiology of ICH is complex and remains enigmatic. To this end, ICH was induced in male CD-1 mice and the ipsilateral brain tissue was characterized in an unbiased manner using a combination of proteomics and bioinformatics approaches. A total of 4833 proteins were revealed by quantitative proteomic analysis. Of those, 207 proteins exhibited significantly altered expression after ICH in comparison to sham. It was found that 46 proteins were significantly upregulated and 161 proteins were significantly downregulated after ICH compared to sham. The quantitative proteomics approach combined with bioinformatics revealed several novel molecular targets (cyclin-dependent-like kinase 5, E3 ubiquitin-protein ligase, protein phosphatase 2A-alpha, protein phosphatase 2A-beta, serine/threonine-protein kinase PAK1, alpha-actinin-4, calpain-8, axin-1, NCK1, and septin-4), and related signaling pathways, which could play roles in secondary brain injury and long-term neurobehavioral outcomes after ICH warranting further investigation.
Collapse
|
36
|
Ullah F, Liang H, Niedermayer G, Münch G, Gyengesi E. Evaluation of Phytosomal Curcumin as an Anti-inflammatory Agent for Chronic Glial Activation in the GFAP-IL6 Mouse Model. Front Neurosci 2020; 14:170. [PMID: 32226360 PMCID: PMC7081170 DOI: 10.3389/fnins.2020.00170] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/14/2020] [Indexed: 01/13/2023] Open
Abstract
Chronic glial activation is characterized by an increased number of activated microglia and astroglia; these secrete free radicals and cytotoxic cytokines, subsequently causing neuronal damage. This study investigated the hypothesis that a soy-lecithin based phytosomal curcumin formulation can decrease glial activation in the brains of GFAP-IL6 mice, a model of chronic glial activation, which exhibits gliosis in various regions of the brain. Three doses of Meriva curcumin (MC) (874, 436, and 218 PPM) were fed to 3-month-old GFAP-IL6 and wild-type (WT) mice for 4 weeks. As markers of glial activation, the total numbers of Iba-1+ and TSPO+ microglia and macrophages, and GFAP+ astrocytes, were determined in the cerebellum and hippocampus by immunohistochemistry and unbiased stereology. Furthermore, the morphology of the glial cells was assessed by confocal microscopy and Sholl analysis. Administration of phytosomal curcumin led to a dose-dependent reduction in neuroinflammatory markers. Phytosomal curcumin (874 PPM) decreased the number of microglia by 26.2% in the hippocampus and by 48% in the cerebellum of the GFAP-IL6 mice compared with the GFAP-IL6 mice on normal food. Additionally, GFAP+ astrocyte numbers in the hippocampus of the GFAP-IL6 mice were decreased by 42%. The GFAP-IL6 mice exhibited a different microglial morphology to the WT mice, showing an increased soma size and perimeter. This difference was significantly reduced by the 874 PPM phytosomal curcumin dose. Our findings demonstrate that phytosomal curcumin is able to attenuate the inflammatory pathology, and potentially reverse the detrimental effects of chronic glial activation.
Collapse
Affiliation(s)
- Faheem Ullah
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Huazheng Liang
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,Department of Neurology, Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Garry Niedermayer
- School of Science, Western Sydney University, Campbelltown, NSW, Australia
| | - Gerald Münch
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,NICM Health Research Institute, Western Sydney University, Campbelltown, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
37
|
Betlazar C, Middleton RJ, Banati R, Liu GJ. The Translocator Protein (TSPO) in Mitochondrial Bioenergetics and Immune Processes. Cells 2020; 9:cells9020512. [PMID: 32102369 PMCID: PMC7072813 DOI: 10.3390/cells9020512] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
The translocator protein (TSPO) is an outer mitochondrial membrane protein that is widely used as a biomarker of neuroinflammation, being markedly upregulated in activated microglia in a range of brain pathologies. Despite its extensive use as a target in molecular imaging studies, the exact cellular functions of this protein remain in question. The long-held view that TSPO plays a fundamental role in the translocation of cholesterol through the mitochondrial membranes, and thus, steroidogenesis, has been disputed by several groups with the advent of TSPO knockout mouse models. Instead, much evidence is emerging that TSPO plays a fundamental role in cellular bioenergetics and associated mitochondrial functions, also part of a greater role in the innate immune processes of microglia. In this review, we examine the more direct experimental literature surrounding the immunomodulatory effects of TSPO. We also review studies which highlight a more central role for TSPO in mitochondrial processes, from energy metabolism, to the propagation of inflammatory responses through reactive oxygen species (ROS) modulation. In this way, we highlight a paradigm shift in approaches to TSPO functioning.
Collapse
Affiliation(s)
- Calina Betlazar
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia
- Correspondence: (C.B.); (G-J.L.)
| | - Ryan J. Middleton
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
| | - Richard Banati
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia
| | - Guo-Jun Liu
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia
- Correspondence: (C.B.); (G-J.L.)
| |
Collapse
|
38
|
Intracerebral Hemorrhage: Blood Components and Neurotoxicity. Brain Sci 2019; 9:brainsci9110316. [PMID: 31717522 PMCID: PMC6896063 DOI: 10.3390/brainsci9110316] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke which is associated with the highest mortality and morbidity rates of all strokes. Although it is a major public health problem, there is no effective treatment for ICH. As a consequence of ICH, various blood components accumulate in the brain parenchyma and are responsible for much of the secondary brain damage and ICH-induced neurological deficits. Therefore, the strategies that could attenuate the blood component-induced neurotoxicity and improve hematoma resolution are highly needed. The present article provides an overview of blood-induced brain injury after ICH and emphasizes the need to conduct further studies elucidating the mechanisms of hematoma resolution after ICH.
Collapse
|
39
|
Murtaj V, Belloli S, Di Grigoli G, Pannese M, Ballarini E, Rodriguez-Menendez V, Marmiroli P, Cappelli A, Masiello V, Monterisi C, Bellelli G, Panina-Bordignon P, Moresco RM. Age and Sex Influence the Neuro-inflammatory Response to a Peripheral Acute LPS Challenge. Front Aging Neurosci 2019; 11:299. [PMID: 31749696 PMCID: PMC6848890 DOI: 10.3389/fnagi.2019.00299] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/17/2019] [Indexed: 12/21/2022] Open
Abstract
Aging is associated with an exaggerated response to peripheral inflammatory challenges together with behavioral and cognitive deficits. Studies considering both age and sex remain limited, despite sex dimorphism of astrocytes and microglial cells is largely recognized. To fill this knowledge gap, we investigated the effect of a single intraperitoneal lipopolysaccharide (LPS) administration in adult and aged mice. We assessed the expression of different inflammatory mediators, and the microglial response through binding of [18F]-VC701 tracer to translocator protein (TSPO) receptors in the male and female brain. Aged female brain showed a higher pro-inflammatory response to LPS compared to adult female and to aged male, as revealed by ex vivo binding to TSPO receptors and pro-inflammatory mediator transcript levels. The highest astroglial reaction was observed in the brain of aged females. Differently to the other groups of animals, in aged males LPS challenge did not affect transcription of triggering receptor expressed on myeloid cells 2 (TREM2). In conclusion, our study shows that in the mouse’s brain the neuro-inflammatory response to an acute peripheral insult is sex- and age-dependent. Moreover, our results might set the basis for further studies aimed at identifying sex-related targets involved in the modulation of the aberrant neuro-inflammatory response that characterizes aging. This knowledge could be relevant for the treatment of conditions such as delirium and dementia.
Collapse
Affiliation(s)
- Valentina Murtaj
- PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Sara Belloli
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, Milan, Italy.,Institute of Molecular Bioimaging and Physiology of National Reasearch Council, Segrate, Italy
| | - Giuseppe Di Grigoli
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, Milan, Italy.,Institute of Molecular Bioimaging and Physiology of National Reasearch Council, Segrate, Italy
| | - Maria Pannese
- Neuroimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Ballarini
- Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Department of Medicine and Surgery, Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
| | - Virginia Rodriguez-Menendez
- Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Department of Medicine and Surgery, Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
| | - Paola Marmiroli
- Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Department of Medicine and Surgery, Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Valeria Masiello
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Monterisi
- Department of Medicine and Surgery, Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
| | - Giuseppe Bellelli
- Acute Geriatric Unit, School of Medicine and Surgery, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Paola Panina-Bordignon
- Neuroimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,School of Medicine and Surgery, San Raffaele Vita-Salute University, Milan, Italy
| | - Rosa Maria Moresco
- PET and Nuclear Medicine Unit, San Raffaele Scientific Institute, Milan, Italy.,Department of Medicine and Surgery, Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
40
|
Recent Developments in TSPO PET Imaging as A Biomarker of Neuroinflammation in Neurodegenerative Disorders. Int J Mol Sci 2019; 20:ijms20133161. [PMID: 31261683 PMCID: PMC6650818 DOI: 10.3390/ijms20133161] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is an inflammatory response in the brain and spinal cord, which can involve the activation of microglia and astrocytes. It is a common feature of many central nervous system disorders, including a range of neurodegenerative disorders. An overlap between activated microglia, pro-inflammatory cytokines and translocator protein (TSPO) ligand binding was shown in early animal studies of neurodegeneration. These findings have been translated in clinical studies, where increases in TSPO positron emission tomography (PET) signal occur in disease-relevant areas across a broad spectrum of neurodegenerative diseases. While this supports the use of TSPO PET as a biomarker to monitor response in clinical trials of novel neurodegenerative therapeutics, the clinical utility of current TSPO PET radioligands has been hampered by the lack of high affinity binding to a prevalent form of polymorphic TSPO (A147T) compared to wild type TSPO. This review details recent developments in exploration of ligand-sensitivity to A147T TSPO that have yielded ligands with improved clinical utility. In addition to developing a non-discriminating TSPO ligand, the final frontier of TSPO biomarker research requires developing an understanding of the cellular and functional interpretation of the TSPO PET signal. Recent insights resulting from single cell analysis of microglial phenotypes are reviewed.
Collapse
|
41
|
Ghadery C, Best LA, Pavese N, Tai YF, Strafella AP. PET Evaluation of Microglial Activation in Non-neurodegenerative Brain Diseases. Curr Neurol Neurosci Rep 2019; 19:38. [PMID: 31139952 PMCID: PMC6538572 DOI: 10.1007/s11910-019-0951-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF THE REVIEW Microglial cell activation is an important component of neuroinflammation, and it is generally well accepted that chronic microglial activation is indicative of accumulating tissue damage in neurodegenerative conditions, particularly in the earlier stages of disease. Until recently, there has been less focus on the role of neuroinflammation in other forms of neurological and neuropsychiatric conditions. Through this review, we hope to demonstrate the important role TSPO PET imaging has played in illuminating the pivotal role of neuroinflammation and microglial activation underpinning these conditions. RECENT FINDINGS TSPO is an 18 kDa protein found on the outer membrane of mitochondria and can act as a marker of microglial activation using nuclear imaging. Through the development of radiopharmaceuticals targeting TSPO, researchers have been able to better characterise the spatial-temporal evolution of chronic neurological conditions, ranging from the focal autoimmune reactions seen in multiple sclerosis to the Wallerian degeneration at remote parts of the brain months following acute cerebral infarction. Development of novel techniques to investigate neuroinflammation within the central nervous system, for the purposes of diagnosis and therapeutics, has flourished over the past few decades. TSPO has proven itself a robust and sensitive biomarker of microglial activation and neuroimaging affords a minimally invasive technique to characterise neuroinflammatory processes in vivo.
Collapse
Affiliation(s)
- Christine Ghadery
- The Edmond J. Safra Program in Parkinson's Disease & Movement Disorder Unit, Toronto Western Hospital & Krembil Research Institute, University Health Network; Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Laura A Best
- Clinical Ageing Research Unit, Newcastle University, Campus for Ageing and Vitality, Westgate Road, Newcastle Upon Tyne, UK.
| | - Nicola Pavese
- Clinical Ageing Research Unit, Newcastle University, Campus for Ageing and Vitality, Westgate Road, Newcastle Upon Tyne, UK
- PET centre, University of Aarhus Denmark, Aarhus, Denmark
| | - Yen Foung Tai
- Imperial College London South Kensington Campus, London, UK
| | - Antonio P Strafella
- The Edmond J. Safra Program in Parkinson's Disease & Movement Disorder Unit, Toronto Western Hospital & Krembil Research Institute, University Health Network; Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Bonsack F, Sukumari-Ramesh S. Differential Cellular Expression of Galectin-1 and Galectin-3 After Intracerebral Hemorrhage. Front Cell Neurosci 2019; 13:157. [PMID: 31156388 PMCID: PMC6530358 DOI: 10.3389/fncel.2019.00157] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating sub-type of stroke with no proven treatment. Given the emerging role of Galectin-1 and Galectin-3 in neuroimmune responses, the objective of the current manuscript is to elucidate hemorrhagic-injury induced modulation and cellular expression of Galectin-1 and Galectin-3 in the brain in a pre-clinical model of ICH. To address this, ICH was induced in male CD1 mice by collagenase injection method. Western blotting as well as Immunofluorescence staining was performed to characterize the temporal expression pattern as well as cellular localization of Galectin-1 and Galectin-3 after ICH. Further, genetic studies were conducted to assess the functional role of Galectin-1 and Galectin-3 in inflammatory response employing a murine macrophage cell line, RAW 264.7. Galectin-1 and Galectin-3 exhibited very profound and increased expression from day 3 to day 7-post-injury, in the perihematomal brain region after ICH in comparison to Sham. Further, Galectin-1 expression was mostly observed in GFAP-positive astrocytes whereas Galectin-3 expression was observed mostly in Iba1-positive microglia/macrophages as well as CD16/32 (M1 microglial/macrophage marker)-positive cells. Moreover, genetic studies revealed a negative regulatory role of both Galectin-1 and Galectin-3 in the release of a proinflammatory cytokine, IL-6 from RAW 264.7 cells depending on the stimulus. Altogether, the present manuscript demonstrates for the first time, increased expression as well as cellular localization of Galectin-1 and Galectin-3 in the perihematomal brain regions after ICH. In addition, the manuscript raises the potential of Galectin-1 and Galectin-3 in modulating glial responses and thereby brain injury after ICH, warranting further investigation.
Collapse
Affiliation(s)
- Frederick Bonsack
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sangeetha Sukumari-Ramesh
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
43
|
Synthesis and in vitro evaluation of new translocator protein ligands designed for positron emission tomography. Future Med Chem 2019; 11:539-550. [PMID: 30888874 DOI: 10.4155/fmc-2018-0444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
AIM Dysregulated levels of the translocator protein TSPO 18 KDa have been reported in several disorders, particularly neurodegenerative diseases. This makes TSPO an interesting target for the development of diagnostic biomarkers. Even though several radioligands have already been developed for in vivo TSPO imaging, the ideal TSPO radiotracer has still not been found. RESULTS Here, we report the chemical synthesis of a set of new TSPO ligands designed for future application in positron emission tomography, together with the determination of their biological activity and applied 11C-labeling strategy. CONCLUSION The lead compound of our series, (R)-[11C]Me@NEBIQUINIDE, showed very promising results and is therefore proposed to be further evaluated under in vivo settings.
Collapse
|
44
|
Regional elevations in microglial activation and cerebral glucose utilization in frontal white matter tracts of rhesus monkeys following prolonged cocaine self-administration. Brain Struct Funct 2019; 224:1417-1428. [PMID: 30747315 DOI: 10.1007/s00429-019-01846-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 02/06/2019] [Indexed: 12/18/2022]
Abstract
It has been shown that exposure to cocaine can result in neuroinflammatory responses. Microglia, the resident CNS immune cells, undergo a transition to an activated state when challenged. In rodents, and possibly humans, cocaine exposure activates microglia. The goal of this study was to assess the extent and magnitude of microglial activation in rhesus monkeys with an extensive history of cocaine self-administration. Male rhesus monkeys (N = 4/group) were trained to respond on a fixed-interval 3-min schedule of food or 0.3 mg/kg/injection cocaine presentation (30 reinforcers/session) for 300 sessions. At the end of the final session, monkeys were administered 2-[14C]deoxyglucose intravenously and 45 min later euthanized. Brain sections were used for autoradiographic assessments of glucose utilization and for microglia activation with [3H]PK11195, a marker for the microglial 18-kDa translocator protein. There were no group differences in gray matter [3H]PK11195 binding, while binding was significantly greater in cocaine self-administration animals as compared to food controls in 8 of the 11 white matter tracts measured at the striatal level. Binding did not differ from control at other levels. There were also significant increases in white matter local cerebral glucose utilization at the striatal level, which were positively correlated with [3H]PK11195 binding. The present findings demonstrate an elevation in [3H]PK11195 binding in forebrain white matter tracts of nonhuman primates with a prolonged history of cocaine self-administration. These elevations were also associated with greater cerebral metabolic rates. These data suggest that white matter deficits may contribute to behavioral, motivational, and cognitive impairments observed in cocaine abusers.
Collapse
|
45
|
Azrad M, Zeineh N, Weizman A, Veenman L, Gavish M. The TSPO Ligands 2-Cl-MGV-1, MGV-1, and PK11195 Differentially Suppress the Inflammatory Response of BV-2 Microglial Cell to LPS. Int J Mol Sci 2019; 20:ijms20030594. [PMID: 30704062 PMCID: PMC6387401 DOI: 10.3390/ijms20030594] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 02/07/2023] Open
Abstract
The 18 kDa Translocator Protein (TSPO) is a marker for microglial activation as its expression is enhanced in activated microglia during neuroinflammation. TSPO ligands can attenuate neuroinflammation and neurotoxicity. In the present study, we examined the efficacy of new TSPO ligands designed by our laboratory, MGV-1 and 2-Cl-MGV-1, in mitigating an in vitro neuroinflammatory process compared to the classic TSPO ligand, PK 11195. We exposed BV-2 microglial cells to lipopolysaccharide (LPS) for 24 h to induce inflammatory response and added the three TSPO ligands: (1) one hour before LPS treatment (pretreatment), (2) simultaneously with LPS (cotreatment), and (3) one hour after LPS exposure (post-treatment). We evaluated the capability of TSPO ligands to reduce the levels of three glial inflammatory markers: cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and nitric oxide (NO). We compared the effects of the two novel ligands to PK 11195. Both 2-Cl-MGV-1 and MGV-1 reduced the levels of glial COX-2, iNOS, and NO in LPS-treated BV-2 cells more efficiently than PK 11195. Notably, even when added after exposure to LPS, all ligands were able to suppress the inflammatory response. Due to their pronounced anti-inflammatory activity, 2-Cl-MGV-1 and MGV-1 may serve as potential therapeutics in neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Maya Azrad
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel.
| | - Nidal Zeineh
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel.
| | - Abraham Weizman
- Research Unit at Geha Mental Health Center and the Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva 4910002, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Leo Veenman
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel.
| | - Moshe Gavish
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
46
|
Horiguchi Y, Ohta N, Yamamoto S, Koide M, Fujino Y. Midazolam suppresses the lipopolysaccharide-stimulated immune responses of human macrophages via translocator protein signaling. Int Immunopharmacol 2019; 66:373-382. [DOI: 10.1016/j.intimp.2018.11.050] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022]
|
47
|
Zhao R, Jiang J, Li H, Chen M, Liu R, Sun S, Ma D, Liang X, Wang S. Phosphatidylserine-microbubble targeting-activated microglia/macrophage in inflammation combined with ultrasound for breaking through the blood-brain barrier. J Neuroinflammation 2018; 15:334. [PMID: 30501630 PMCID: PMC6271401 DOI: 10.1186/s12974-018-1368-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/15/2018] [Indexed: 01/21/2023] Open
Abstract
Background and purpose Inflammatory reaction plays a crucial role in cerebral ischemia reperfusion (IR) injury. It has been shown that activated microglia long-term existed in cerebral ischemia and induced second injury. Therefore, we hypothesize that prepared phosphatidylserine (PS)-modified microbubbles (PS-MBs) combined with ultrasound-targeted microbubble destruction (UTMD) can safely open the blood–brain barrier (BBB) and target activated microglia for inflammatory area in the later stage of ischemia reperfusion. Methods To verify our hypothesis, rat model of IR was established, then the change of activated microglia/macrophage (M/M) and permeability of BBB at 1, 7, 14, and 21 days could be clearly observed post IR. And the activated M/M still can be observed during the whole experiment. Results The Evans blue extravasation of BBB gradually declined from day 1 to day 21. Compared to the control group, microbubbles containing PS were taken up more by activated M/M (approximately twofold) both in vitro and in vivo. Conclusions PS-MBs combined with ultrasound (US) exposure could safely open BBB, and the resulting PS nanoparticles (PS-NPs) could further target activated M/M in the neuroinflammation. Electronic supplementary material The online version of this article (10.1186/s12974-018-1368-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ranran Zhao
- Ordos Center Hospital, Ordos, 017000, Inner Mongolia, China.,Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Jie Jiang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Huiwen Li
- Ordos Center Hospital, Ordos, 017000, Inner Mongolia, China
| | - Min Chen
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 10019, China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 10019, China
| | - Sujuan Sun
- Ordos Center Hospital, Ordos, 017000, Inner Mongolia, China
| | - De Ma
- Ordos Center Hospital, Ordos, 017000, Inner Mongolia, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Shumin Wang
- Ordos Center Hospital, Ordos, 017000, Inner Mongolia, China. .,Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
48
|
Cardiac-specific Conditional Knockout of the 18-kDa Mitochondrial Translocator Protein Protects from Pressure Overload Induced Heart Failure. Sci Rep 2018; 8:16213. [PMID: 30385779 PMCID: PMC6212397 DOI: 10.1038/s41598-018-34451-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 10/18/2018] [Indexed: 01/17/2023] Open
Abstract
Heart failure (HF) is characterized by abnormal mitochondrial calcium (Ca2+) handling, energy failure and impaired mitophagy resulting in contractile dysfunction and myocyte death. We have previously shown that the 18-kDa mitochondrial translocator protein of the outer mitochondrial membrane (TSPO) can modulate mitochondrial Ca2+ uptake. Experiments were designed to test the role of the TSPO in a murine pressure-overload model of HF induced by transverse aortic constriction (TAC). Conditional, cardiac-specific TSPO knockout (KO) mice were generated using the Cre-loxP system. TSPO-KO and wild-type (WT) mice underwent TAC for 8 weeks. TAC-induced HF significantly increased TSPO expression in WT mice, associated with a marked reduction in systolic function, mitochondrial Ca2+ uptake, complex I activity and energetics. In contrast, TSPO-KO mice undergoing TAC had preserved ejection fraction, and exhibited fewer clinical signs of HF and fibrosis. Mitochondrial Ca2+ uptake and energetics were restored in TSPO KO mice, associated with decreased ROS, improved complex I activity and preserved mitophagy. Thus, HF increases TSPO expression, while preventing this increase limits the progression of HF, preserves ATP production and decreases oxidative stress, thereby preventing metabolic failure. These findings suggest that pharmacological interventions directed at TSPO may provide novel therapeutics to prevent or treat HF.
Collapse
|
49
|
Yang Z, Liu Q, Shi H, Jiang X, Wang S, Lu Y, Zhang J, Huang X, Yu A. Interleukin 17A exacerbates ER-stress-mediated inflammation of macrophages following ICH. Mol Immunol 2018; 101:38-45. [DOI: 10.1016/j.molimm.2018.05.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/22/2022]
|
50
|
Lillethorup TP, Glud AN, Landeck N, Alstrup AKO, Jakobsen S, Vang K, Doudet DJ, Brooks DJ, Kirik D, Hinz R, Sørensen JC, Landau AM. In vivo quantification of glial activation in minipigs overexpressing human α-synuclein. Synapse 2018; 72:e22060. [PMID: 30009467 DOI: 10.1002/syn.22060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/06/2018] [Accepted: 07/11/2018] [Indexed: 12/25/2022]
Abstract
Parkinson's disease is characterized by a progressive loss of substantia nigra (SN) dopaminergic neurons and the formation of Lewy bodies containing accumulated alpha-synuclein (α-syn). The pathology of Parkinson's disease is associated with neuroinflammatory microglial activation, which may contribute to the ongoing neurodegeneration. This study investigates the in vivo microglial and dopaminergic response to overexpression of α-syn. We used positron emission tomography (PET) and the 18 kDa translocator protein radioligand, [11 C](R)PK11195, to image brain microglial activation and (+)-α-[11 C]dihydrotetrabenazine ([11 C]DTBZ), to measure vesicular monoamine transporter 2 (VMAT2) availability in Göttingen minipigs following injection with recombinant adeno-associated virus (rAAV) vectors expressing either mutant A53T α-syn or green fluorescent protein (GFP) into the SN (4 rAAV-α-syn, 4 rAAV-GFP, 5 non-injected control minipigs). We performed motor symptom assessment and immunohistochemical examination of tyrosine hydroxylase (TH) and transgene expression. Expression of GFP and α-syn was observed at the SN injection site and in the striatum. We observed no motor symptoms or changes in striatal [11 C]DTBZ binding potential in vivo or striatal or SN TH staining in vitro between the groups. The mean [11 C](R)PK11195 total volume of distribution was significantly higher in the basal ganglia and cortical areas of the α-syn group than the control animals. We conclude that mutant α-syn expression in the SN resulted in microglial activation in multiple sub- and cortical regions, while it did not affect TH stains or VMAT2 availability. Our data suggest that microglial activation constitutes an early response to accumulation of α-syn in the absence of dopamine neuron degeneration.
Collapse
Affiliation(s)
- Thea Pinholt Lillethorup
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Andreas Nørgaard Glud
- Center for Experimental Neuroscience (CENSE), Department of Neurosurgery, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Natalie Landeck
- Brain Repair and Imaging in Neural Systems (BRAINS) Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Aage Kristian Olsen Alstrup
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Kim Vang
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Doris J Doudet
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark.,Department of Medicine/Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David J Brooks
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark.,Division of Neuroscience, Department of Medicine, Imperial College London, London, United Kingdom.,Division of Neuroscience, Newcastle University, Newcastle, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS) Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom
| | - Jens Christian Sørensen
- Center for Experimental Neuroscience (CENSE), Department of Neurosurgery, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne M Landau
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark.,Translational Neuropsychiatry Unit, Institute of Clinical Medicine, Aarhus University, Risskov, Denmark
| |
Collapse
|