1
|
Xu Y, Tang L, Zhou C, Sun L, Hu Y, Zhang Z, Xia S, Bao X, Yang H, Xu Y. Inhibition of ADORA3 promotes microglial phagocytosis and alleviates chronic ischemic white matter injury. CNS Neurosci Ther 2024; 30:e14742. [PMID: 38715283 PMCID: PMC11076989 DOI: 10.1111/cns.14742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/31/2024] [Accepted: 04/13/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Adenosine A3 receptor (ADORA3) belongs to the adenosine receptor families and the role of ADORA3 in vascular dementia (VaD) is largely unexplored. The present study sought to determine the therapeutic role of ADORA3 antagonist in a mouse model of VaD. METHODS The GSE122063 dataset was selected to screen the differential expression genes and pathways between VaD patients and controls. A mouse model of bilateral carotid artery stenosis (BCAS) was established. The cognitive functions were examined by the novel object recognition test, Y maze test, and fear of conditioning test. The white matter injury (WMI) was examined by 9.4 T MRI, western blot, and immunofluorescence staining. The mechanisms of ADORA3-regulated phagocytosis by microglia were examined using qPCR, western blot, dual immunofluorescence staining, and flow cytometry. RESULTS The expression of ADORA3 was elevated in brain tissues of VaD patients and ADORA3 was indicated as a key gene for VaD in the GSE122063. In BCAS mice, the expression of ADORA3 was predominantly elevated in microglia in the corpus callosum. ADORA3 antagonist promotes microglial phagocytosis to myelin debris by facilitating cAMP/PKA/p-CREB pathway and thereby ameliorates WMI and cognitive impairment in BCAS mice. The therapeutic effect of ADORA3 antagonist was partially reversed by the inhibition of the cAMP/PKA pathway. CONCLUSIONS ADORA3 antagonist alleviates chronic ischemic WMI by modulating myelin clearance of microglia, which may be a potential therapeutic target for the treatment of VaD.
Collapse
Affiliation(s)
- Yuhao Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Limoran Tang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Chao Zhou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Liang Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Yujie Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Nanjing Neurology Clinical Medical CenterNanjingChina
| |
Collapse
|
2
|
Jeong SP, Sharma N, An SSA. Role of Calcitriol and Vitamin D Receptor ( VDR) Gene Polymorphisms in Alzheimer's Disease. Int J Mol Sci 2024; 25:4806. [PMID: 38732025 PMCID: PMC11084202 DOI: 10.3390/ijms25094806] [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: 02/15/2024] [Revised: 02/21/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid beta (Aβ) buildup and neuronal degeneration. An association between low serum vitamin D levels and an increased risk of AD has been reported in several epidemiological studies. Calcitriol (1,25-dihydroxycholecalciferol) is the active form of vitamin D, and is generated in the kidney and many other tissues/organs, including the brain. It is a steroid hormone that regulates important functions like calcium/phosphorous levels, bone mineralization, and immunomodulation, indicating its broader systemic significance. In addition, calcitriol confers neuroprotection by mitigating oxidative stress and neuroinflammation, promoting the clearance of Aβ, myelin formation, neurogenesis, neurotransmission, and autophagy. The receptors to which calcitriol binds (vitamin D receptors; VDRs) to exert its effects are distributed over many organs and tissues, representing other significant roles of calcitriol beyond sustaining bone health. The biological effects of calcitriol are manifested through genomic (classical) and non-genomic actions through different pathways. The first is a slow genomic effect involving nuclear VDR directly affecting gene transcription. The association of AD with VDR gene polymorphisms relies on the changes in vitamin D consumption, which lowers VDR expression, protein stability, and binding affinity. It leads to the altered expression of genes involved in the neuroprotective effects of calcitriol. This review summarizes the neuroprotective mechanism of calcitriol and the role of VDR polymorphisms in AD, and might help develop potential therapeutic strategies and markers for AD in the future.
Collapse
Affiliation(s)
| | - Niti Sharma
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| | - Seong Soo A. An
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
3
|
Mirarchi A, Albi E, Beccari T, Arcuri C. Microglia and Brain Disorders: The Role of Vitamin D and Its Receptor. Int J Mol Sci 2023; 24:11892. [PMID: 37569267 PMCID: PMC10419106 DOI: 10.3390/ijms241511892] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Accounting for 5-20% of the total glial cells present in the adult brain, microglia are involved in several functions: maintenance of the neural environment, response to injury and repair, immunesurveillance, cytokine secretion, regulation of phagocytosis, synaptic pruning, and sculpting postnatal neural circuits. Microglia contribute to some neurodevelopmental disorders, such as Nasu-Hakola disease (NHD), Tourette syndrome (TS), autism spectrum disorder (ASD), and schizophrenia. Moreover, microglial involvement in neurodegenerative diseases, such as Alzheimer's (AD) and Parkinson's (PD) diseases, has also been well established. During the last two decades, epidemiological and research studies have demonstrated the involvement of vitamin D3 (VD3) in the brain's pathophysiology. VD3 is a fat-soluble metabolite that is required for the proper regulation of many of the body's systems, as well as for normal human growth and development, and shows neurotrophic and neuroprotective actions and influences on neurotransmission and synaptic plasticity, playing a role in various neurological diseases. In order to better understand the exact mechanisms behind the diverse actions of VD3 in the brain, a large number of studies have been performed on isolated cells or tissues of the central nervous system (CNS). Here, we discuss the involvement of VD3 and microglia on neurodegeneration- and aging-related diseases.
Collapse
Affiliation(s)
- Alessandra Mirarchi
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy;
| | - Elisabetta Albi
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy; (E.A.); (T.B.)
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy; (E.A.); (T.B.)
| | - Cataldo Arcuri
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy;
| |
Collapse
|
4
|
Drug-induced microglial phagocytosis in multiple sclerosis and experimental autoimmune encephalomyelitis and the underlying mechanisms. Mol Biol Rep 2023; 50:749-759. [PMID: 36309614 DOI: 10.1007/s11033-022-07968-z] [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: 04/11/2022] [Accepted: 09/21/2022] [Indexed: 02/01/2023]
Abstract
Microglia are resident macrophages of the central nervous system (CNS). It plays a significant role in immune surveillance under physiological conditions. On stimulation by pathogens, microglia change their phenotypes, phagocytize toxic molecules, secrete pro-inflammatory/anti-inflammatory factors, promotes tissue repair, and maintain the homeostasis in CNS. Accumulation of myelin debris in multiple sclerosis (MS)/experimental autoimmune encephalomyelitis (EAE) inhibits remyelination by decreasing the phagocytosis by microglia and prevent the recovery of MS/EAE. Drug induced microglia phagocytosis could be a novel therapeutic intervention for the treatment of MS/EAE. But the abnormal phagocytosis of neurons and synapses by activated microglia will lead to neuronal damage and degeneration. It indicates that the phagocytosis of microglia has many beneficial and harmful effects in central neurodegenerative diseases. Therefore, simply promoting or inhibiting the phagocytic activity of microglia may not achieve ideal therapeutic results. However, limited reports are available to elucidate the microglia mediated phagocytosis and its underlying molecular mechanisms. On this basis, the present review describes microglia-mediated phagocytosis, drug-induced microglia phagocytosis, molecular mechanism, and novel approach for MS/EAE treatment.
Collapse
|
5
|
New Insights into Risk Genes and Their Candidates in Multiple Sclerosis. Neurol Int 2022; 15:24-39. [PMID: 36648967 PMCID: PMC9844300 DOI: 10.3390/neurolint15010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Oligodendrocytes are central nervous system glial cells that wrap neuronal axons with their differentiated myelin membranes as biological insulators. There has recently been an emerging concept that multiple sclerosis could be triggered and promoted by various risk genes that appear likely to contribute to the degeneration of oligodendrocytes. Despite the known involvement of vitamin D, immunity, and inflammatory cytokines in disease progression, the common causes and key genetic mechanisms remain unknown. Herein, we focus on recently identified risk factors and risk genes in the background of multiple sclerosis and discuss their relationships.
Collapse
|
6
|
Galoppin M, Kari S, Soldati S, Pal A, Rival M, Engelhardt B, Astier A, Thouvenot E. Full spectrum of vitamin D immunomodulation in multiple sclerosis: mechanisms and therapeutic implications. Brain Commun 2022; 4:fcac171. [PMID: 35813882 PMCID: PMC9260308 DOI: 10.1093/braincomms/fcac171] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/03/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Vitamin D deficiency has been associated with the risk of multiple sclerosis, disease activity and progression. Results from in vitro experiments, animal models and analysis of human samples from randomized controlled trials provide comprehensive data illustrating the pleiotropic actions of Vitamin D on the immune system. They globally result in immunomodulation by decreasing differentiation of effector T and B cells while promoting regulatory subsets. Vitamin D also modulates innate immune cells such as macrophages, monocytes and dendritic cells, and acts at the level of the blood–brain barrier reducing immune cell trafficking. Vitamin D exerts additional activity within the central nervous system reducing microglial and astrocytic activation. The immunomodulatory role of Vitamin D detected in animal models of multiple sclerosis has suggested its potential therapeutic use for treating multiple sclerosis. In this review, we focus on recent published data describing the biological effects of Vitamin D in animal models of multiple sclerosis on immune cells, blood–brain barrier function, activation of glial cells and its potential neuroprotective effects. Based on the current knowledge, we also discuss optimization of therapeutic interventions with Vitamin D in patients with multiple sclerosis, as well as new technologies allowing in-depth analysis of immune cell regulations by vitamin D.
Collapse
Affiliation(s)
- Manon Galoppin
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
| | - Saniya Kari
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Arindam Pal
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Manon Rival
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| | | | - Anne Astier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Eric Thouvenot
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| |
Collapse
|
7
|
Zheng L, Jia J, Chen Y, Liu R, Cao R, Duan M, Zhang M, Xu Y. Pentoxifylline alleviates ischemic white matter injury through up-regulating Mertk-mediated myelin clearance. J Neuroinflammation 2022; 19:128. [PMID: 35642056 PMCID: PMC9153105 DOI: 10.1186/s12974-022-02480-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/15/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Vascular dementia (VAD) is the second most common type of dementia lacking effective treatments. Pentoxifylline (PTX), a nonselective phosphodiesterase inhibitor, displays protective effects in multiple cerebral diseases. In this study, we aimed to investigate the therapeutic effects and potential mechanisms of PTX in VAD. METHODS Bilateral common carotid artery stenosis (BCAS) mouse model was established to mimic VAD. Mouse behavior was tested by open field test, novel object recognition test, Y-maze and Morris water maze (MWM) tests. Histological staining, magnetic resonance imaging (MRI) and electron microscopy were used to define white matter integrity. The impact of PTX on microglia phagocytosis, peroxisome proliferator-activated receptors-γ (PPAR-γ) activation and Mer receptor tyrosine kinase (Mertk) expression was assessed by immunofluorescence, western blotting and flow cytometry with the application of microglia-specific Mertk knockout mice, Mertk inhibitor and PPAR-γ inhibitor. RESULTS Here, we found that PTX treatment alleviated cognitive impairment in novel object recognition test, Y-maze and Morris water maze tests. Furthermore, PTX alleviated white matter injury in corpus callosum (CC) and internal capsule (IC) areas as shown by histological staining and MRI analysis. PTX-treatment group presented thicker myelin sheath than vehicle group by electron microscopy. Mechanistically, PTX facilitated microglial phagocytosis of myelin debris by up-regulating the expression of Mertk in BCAS model and primary cultured microglia. Importantly, microglia-specific Mertk knockout blocked the therapeutic effects of PTX in BCAS model. Moreover, Mertk expression was regulated by the nuclear translocation of PPAR-γ. Through modulating PPAR-γ, PTX enhanced Mertk expression. CONCLUSIONS Collectively, our results demonstrated that PTX showed therapeutic potentials in VAD and alleviated ischemic white matter injury via modulating Mertk-mediated myelin clearance in microglia.
Collapse
Affiliation(s)
- Lili Zheng
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
| | - Junqiu Jia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
| | - Yan Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Renyuan Liu
- Department of Radiology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Runjing Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
| | - Manlin Duan
- Department of Anesthesiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Meijuan Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.
| |
Collapse
|
8
|
Sokolova TV, Rychkova MP, Basova NE, Yefimova MG. Vitamin D3 Inhibits Phagocytic Activity of Rat Brain Astrocytes in Primary Culture. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022030048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Wong SW, Vivash L, Mudududdla R, Nguyen N, Hermans SJ, Shackleford DM, Field J, Xue L, Aprico A, Hancock NC, Haskali M, Stashko MA, Frye SV, Wang X, Binder MD, Ackermann U, Parker MW, Kilpatrick TJ, Baell JB. Development of [ 18F]MIPS15692, a radiotracer with in vitro proof-of-concept for the imaging of MER tyrosine kinase (MERTK) in neuroinflammatory disease. Eur J Med Chem 2021; 226:113822. [PMID: 34563964 DOI: 10.1016/j.ejmech.2021.113822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
MER tyrosine kinase (MERTK) upregulation is associated with M2 polarization of microglia, which plays a vital role in neuroregeneration following damage induced by neuroinflammatory diseases such as multiple sclerosis (MS). Therefore, a radiotracer specific for MERTK could be of great utility in the clinical management of MS, for the detection and differentiation of neuroregenerative and neurodegenerative processes. This study aimed to develop an [18F] ligand with high affinity and selectivity for MERTK as a potential positron emission tomography (PET) radiotracer. MIPS15691 and MIPS15692 were synthesized and kinase assays were utilized to determine potency and selectivity for MERTK. Both compounds were shown to be potent against MERTK, with respective IC50 values of 4.6 nM and 4.0 nM, and were also MERTK-selective. Plasma and brain pharmacokinetics were measured in mice and led to selection of MIPS15692 over MIPS15691. X-ray crystallography was used to visualize how MIPS15692 is recognized by the enzyme. [18F]MIPS15692 was synthesized using an automated iPHASE FlexLab module, with a molar activity (Am) of 49 ± 26 GBq/μmol. The radiochemical purity of [18F]MIPS15692 was >99% and the decay-corrected radiochemical yields (RCYs) were determined as 2.45 ± 0.85%. Brain MERTK protein density was measured by a saturation binding assay in the brain slices of a cuprizone mouse model of MS. High levels of specific binding of [18F]MIPS15692 to MERTK were found, especially in the corpus callosum/hippocampus (CC/HC). The in vivo PET imaging study of [18F]MIPS15692 suggested that its neuroPK is sub-optimal for clinical use. Current efforts are underway to optimize the neuroPK of our next generation PET radiotracers for maximal in vivo utility.
Collapse
Affiliation(s)
- Siu Wai Wong
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Lucy Vivash
- The Central Clinical School and The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Ramesh Mudududdla
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Nghi Nguyen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Stefan J Hermans
- ACRF Rational Drug Discovery Centre, St Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
| | - David M Shackleford
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Judith Field
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3010, Australia
| | - Lian Xue
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Andrea Aprico
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3010, Australia
| | - Nancy C Hancock
- ACRF Rational Drug Discovery Centre, St Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
| | - Mohammad Haskali
- Centre for Molecular Imaging, Peter MacCallum Cancer Centre, Parkville, Victoria, 3052, Australia
| | - Michael A Stashko
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Stephen V Frye
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Michele D Binder
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3010, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Uwe Ackermann
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Michael W Parker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, 3010, Australia; ACRF Rational Drug Discovery Centre, St Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
| | - Trevor J Kilpatrick
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3010, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia.
| | - Jonathan B Baell
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing, 211816, People's Republic of China; Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
10
|
Katsuki H. Nuclear receptors of NR1 and NR4 subfamilies in the regulation of microglial functions and pathology. Pharmacol Res Perspect 2021; 9:e00766. [PMID: 34676987 PMCID: PMC8532137 DOI: 10.1002/prp2.766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/01/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
This review provides an overview of researches on the NR1 and NR4 nuclear receptors involved in the regulation of microglial functions. Nuclear receptors are attractive candidates for drug targets in the therapies of the central nervous system disorders, because the activation of these receptors is expected to regulate the functions and the phenotypes of microglia, by controlling the expression of specific gene subsets and also by regulating the cellular signaling mechanisms in a nongenomic manner. Several members of NR1 nuclear receptor subfamily have been examined for their ability to regulate microglial functions. For example, stimulation of vitamin D receptor inhibits the production of pro-inflammatory factors and increases the production of anti-inflammatory cytokines. Similar regulatory actions of nuclear receptor ligands on inflammation-related genes have also been reported for other NR1 members such as retinoic acid receptors, peroxisome proliferator-activated receptors (PPARs), and liver X receptors (LXRs). In addition, stimulation of PPARγ and LXRs may also result in increased phagocytic activities of microglia. Consistent with these actions, the agonists at nuclear receptors of NR1 subfamily are shown to produce therapeutic effects on animal models of various neurological disorders such as experimental allergic encephalomyelitis, Alzheimer's disease, Parkinson's disease, and ischemic/hemorrhagic stroke. On the other hand, increasing lines of evidence suggest that the stimulation of NR4 subfamily members of nuclear receptors such as Nur77 and Nurr1 also regulates microglial functions and alleviates neuropathological events in several disease models. Further advancement of these research fields may prove novel therapeutic opportunities.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico‐Pharmacological SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
11
|
Mike JK, Ferriero DM. Efferocytosis Mediated Modulation of Injury after Neonatal Brain Hypoxia-Ischemia. Cells 2021; 10:1025. [PMID: 33925299 PMCID: PMC8146813 DOI: 10.3390/cells10051025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Neonatal brain hypoxia-ischemia (HI) is a leading cause of morbidity and long-term disabilities in children. While we have made significant progress in describing HI mechanisms, the limited therapies currently offered for HI treatment in the clinical setting stress the importance of discovering new targetable pathways. Efferocytosis is an immunoregulatory and homeostatic process of clearance of apoptotic cells (AC) and cellular debris, best described in the brain during neurodevelopment. The therapeutic potential of stimulating defective efferocytosis has been recognized in neurodegenerative diseases. In this review, we will explore the involvement of efferocytosis after a stroke and HI as a promising target for new HI therapies.
Collapse
Affiliation(s)
- Jana Krystofova Mike
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Donna Marie Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
- Department of Neurology Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Lamorte S, Shinde R, McGaha TL. Nuclear receptors, the aryl hydrocarbon receptor, and macrophage function. Mol Aspects Med 2021; 78:100942. [PMID: 33451803 DOI: 10.1016/j.mam.2021.100942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022]
Abstract
Nuclear receptors (NRs) are key regulators of innate immune responses and tissue homeostasis. Evidence indicates that NRs significantly impact steady-state immune regulation, uptake and processing of apoptotic cells, tolerance induction, and control of inflammatory immunity. In this review, we describe our current understanding of the NR activity for balancing inflammation and tolerance, the signaling cascade inducing the NR activation and functional responses, and different mechanisms of the NR-driven immune effects in the context of autoimmune diseases. We further describe the ligand-activated transcription factor the aryl hydrocarbon receptor (AhR) that exhibits analogous functionality. Moreover, we will discuss the putative role of NRs and AhR in immune regulation and disease pathogenesis providing a rationale for therapeutic targeting as a unique opportunities in the clinical management of autoimmune diseases.
Collapse
Affiliation(s)
- Sara Lamorte
- Tumor Immunotherapy Program, The Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Rahul Shinde
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute Cancer Center, Philadelphia, PA, USA
| | - Tracy L McGaha
- Tumor Immunotherapy Program, The Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; The Department of Immunology, The University of Toronto, Toronto, ON, Canada.
| |
Collapse
|