1
|
Rehman AS, Kumar P, Parvez S. Dopamine-D2-agonist targets mitochondrial dysfunction via diminishing Drp1 mediated fission and normalizing PGC1-α/SIRT3 pathways in a rodent model of Subarachnoid Haemorrhage. Neuroscience 2025; 564:60-78. [PMID: 39542343 DOI: 10.1016/j.neuroscience.2024.11.028] [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: 07/17/2024] [Revised: 09/28/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
The adverse impact of disturbmitochondrialbiogenesis onearly brain injury (EBI) following subarachnoid haemorrhage (SAH) has been broadly recognized and is closely associated with oxidative stress and neuronal apoptosis. Previous studies have indicated the therapeutic potential of Ropinirole, a dopamine D2 agonist, in Ischemic Stroke. However, there is a lack of evidence regarding the ability of Ropinirole to enhance mitochondrial biogenesis and quality control after subarachnoid haemorrhage. The objective of this study is to investigate the effects of Ropinirole specific doses (10 & 20 mg/kg b. wt.) on mitochondria dysfunction in endovascular perforation SAH model in male Wistar rat. An endovascular perforation model was established using male Wistar rats that had sustained SAH injury. After the SAH injury, SAH grading on blood clot, Nissl staining, and neurobehavioral assessment were used to determine the severity. ROS and MMP, which are indicators of oxidative stress, were examined using flow cytometry. The findings demonstrated that the use of Ropinirole improved neurobehavioral outcomes, decreased brain edema, and reduced oxidative stress and mitochondrial based apoptosis. Further research showed that, Ropinirole therapy inhibit Drp1-mediated fission by accelerating the activity of fusion protein Mfn2/OPA1 along with regulating the translocation of PGC1-α and SIRT3 through restricting cytochrome C inside mitochondria to maintain mitochondrial metabolism. Ropinirole exerted neuroprotective effects by improving mitochondrial activity in a PGC1-α/SIRT3-dependent way via regulating Drp1 mediated fission. The effective treatment for SAH-induced EBI may involve increasing biogenesis and inhibiting excessive mitochondrial fission with Ropinirole.
Collapse
Affiliation(s)
- Ahmed Shaney Rehman
- Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi 110042, India
| | - Suhel Parvez
- Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
2
|
Godínez-Chaparro B, Rodríguez-Ramos MC, Martínez-Lorenzana MG, González-Morales E, Pérez-Ruíz KP, Espinosa de Los Monteros-Zuñiga A, Mendoza-Pérez F, Condes-Lara M. Pramipexole decreases allodynia and hyperalgesia via NF-κB in astrocytes in rats with Parkinson's disease. Pharmacol Biochem Behav 2024; 247:173945. [PMID: 39675389 DOI: 10.1016/j.pbb.2024.173945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/30/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
Pain is one of the principal non-motor symptoms of Parkinson's disease (PD), negatively impacting the patient's quality of life. This study aimed to demonstrate whether an effective dose of pramipexole (PPX) can modulate the NF-κB/p-p65 activation in glial cells (astrocytes and microglia) and diminish the hypersensitivity (allodynia and hyperalgesia) in male Wistar rats with PD. For this, 2 μl of 6-hydroxydopamine (6-OHDA, 8 μg/μL/0.2 μl/min) was administered unilaterally in the Substantia Nigra of the Pars Compacta (SNpc) to establish a PD model rat. Motor behavioral tests were used to validate the PD model, and von Frey filaments were used to evaluate allodynia and hyperalgesia. Immunohistochemical and immunofluorescence were used to analyze the level of tyrosine hydroxylase in SNpc and striatum as well as the expression of GFAP, Iba-1, NF-κB/p-65 in the L4-L6 spinal cord dorsal horn. Unilateral 6-OHDA-lesion reduces motor capacity and produces long-term allodynia and hyperalgesia in both hind paws. L4-L6 spinal cord dorsal horn astrocytes and microglia were active in these 6-OHDA-lesioned rats. Moreover, PPX (1 and 3 mg/Kg, i.p./10 days, n = 10 per group) inhibited the bilateral mechanical hypersensitivity, and PPX (3 mg/Kg/i.p./10 days) reduced 6-OHDA-induced astrocyte and microglia activation, as well as reduced NF-κB/p-p65 expression only in astrocytes of dorsal horn spinal cord in the L5-L6. These findings suggest that PPX could alleviate pain by decreasing the activation of microglia and astrocytes through the NF-κB/p-p65 pathway in the dorsal horn spinal cord. Therefore, PPX could be considered an optional tool for improving pain hypersensitivity in PD patients.
Collapse
Affiliation(s)
- Beatriz Godínez-Chaparro
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico.
| | - Maria Cristina Rodríguez-Ramos
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - María Guadalupe Martínez-Lorenzana
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Estefanía González-Morales
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - Karen Pamela Pérez-Ruíz
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - Antonio Espinosa de Los Monteros-Zuñiga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Felipe Mendoza-Pérez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - Miguel Condes-Lara
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| |
Collapse
|
3
|
Gurram PC, Satarker S, Nassar A, Begum F, Mudgal J, Arora D, Nampoothiri M. Dopamine agonist Rotigotine mitigates lipopolysaccharide-induced neuroinflammation and memory impairment in mice. Metab Brain Dis 2024; 40:23. [PMID: 39565417 DOI: 10.1007/s11011-024-01463-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/17/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Dopaminergic signaling in the Central Nervous System (CNS) has been observed in the pathophysiology of memory deficits. Rotigotine belongs to a non-ergot-based dopamine receptor agonist possessing anti-inflammatory properties. However, it is uncertain if it has a role in ameliorating cognitive decline. Here, we evaluated the actions of rotigotine on neuroinflammation and memory impairment. METHODOLOGY Rotigotine 1, 3, and 5 mg/kg were administered to mice subcutaneously once a day for fifteen days. Lipopolysaccharide (LPS) 750 µg/kg was administered intraperitoneally for seven days to produce cognitive impairment in mice. Morris water maze and Passive avoidance step-down tests were performed to evaluate memory function. Further, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and amyloid-beta (Aβ) were estimated by ELISA. The mouse brain was analyzed for acetylcholinesterase (AChE) activity, lipid peroxidation, catalase, and reduced glutathione levels. RESULTS LPS elevated IL-6, Aβ, TNF-α, and AChE activity, promoted oxidative stress, and caused memory decline in mice. Lower doses of rotigotine 1 and 3 mg/kg significantly reduced neuroinflammation, oxidative stress, and AChE activity, followed by improved cognitive impairment. CONCLUSION Our data suggest that rotigotine 1 and 3 mg/kg could reverse the neuroinflammation-associated memory impairment.
Collapse
Affiliation(s)
- Prasada Chowdari Gurram
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Green fields, Vaddeswaram, A.P-522302, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Ajmal Nassar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Farmiza Begum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Devinder Arora
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
4
|
Shen W, Zhang X, Tang M, Chen W, Wang Y, Zhou H. Targeting of ubiquitination and degradation of KLF15 by E3 ubiquitin ligase KBTBD7 regulates LPS-induced septic brain injury in microglia. Exp Cell Res 2024; 443:114317. [PMID: 39489209 DOI: 10.1016/j.yexcr.2024.114317] [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: 06/20/2024] [Revised: 09/29/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Septic brain injury is a serious disease of the central nervous system that involves inflammation. Kelch repeat and BTB domain containing 7 (KBTBD7), an E3 ubiquitin ligase, is demonstrated to facilitate the pathological changes of various diseases, but its impact on septic brain injury is unclear. In this study, a rat model of septic brain injury was induced by cecal ligation and puncture (CLP). The neurobehavioral score and survival rate of CLP group were worse than those of sham group. In addition, CLP was found to evoke microglia activation, increase inflammation, induce the activation of NLRP3 inflammasome and NF-κB signaling pathway, and upregulate KBTBD7 expression. Immunofluorescence revealed strong positive KBTBD7 staining in CLP rat microglia. Furthermore, primary microglia were exposed to lipopolysaccharide (LPS) to explore the role and mechanism of KBTBD7. The results showed that KBTBD7 expression was increased in LPS-treated microglia. Knockdown of KBTBD7 markedly inhibited LPS-induced proinflammatory cytokine release, as well as the activation of NLRP3 inflammasome and NF-κB signaling pathway. The downstream molecular mechanism of KBTBD7 was then mined. Notably, co-immunoprecipitation (co-IP) results confirmed that KBTBD7 was a novel interacting protein of KLF transcription factor 15 (KLF15) and acted as an E3 ubiquitin ligase that catalyzed the ubiquitination degradation of KLF15 through the ubiquitin-proteasome system. Moreover, recovery experiment data suggested that KLF15 knockdown abolished the anti-inflammatory role of KBTBD7 knockdown in microglia, implying that KLF15 influenced the function of KBTBD7. Taken together, our results reveal a novel KBTBD7-KLF15 signal transduction pathway involved in septic brain injury and provide a potential therapeutic strategy for its treatment.
Collapse
Affiliation(s)
- Wei Shen
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xuzhong Zhang
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Min Tang
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wei Chen
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ying Wang
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Haoquan Zhou
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
5
|
Liu J, Pang SY, Zhou SY, He QY, Zhao RY, Qu Y, Yang Y, Guo ZN. Lipocalin-2 aggravates blood-brain barrier dysfunction after intravenous thrombolysis by promoting endothelial cell ferroptosis via regulating the HMGB1/Nrf2/HO-1 pathway. Redox Biol 2024; 76:103342. [PMID: 39265498 PMCID: PMC11415874 DOI: 10.1016/j.redox.2024.103342] [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: 07/29/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Disruption of the blood-brain barrier (BBB) is a major contributor to hemorrhagic transformation (HT) in patients with acute ischemic stroke (AIS) following intravenous thrombolysis (IVT). However, the clinical therapies aimed at BBB protection after IVT remain limited. METHODS One hundred patients with AIS who underwent IVT were enrolled (42 with HT and 58 without HT 24 h after IVT). Based on the cytokine chip, the serum levels of several AIS-related proteins, including LCN2, ferritin, matrix metalloproteinase-3, vascular endothelial-derived growth factor, and X-linked inhibitor of apoptosis, were detected upon admission, and their associations with HT were analyzed. After finding that LCN2 was related to HT in patients with IVT, we clarified whether the modulation of LCN2 influenced BBB dysfunction and HT after thrombolysis and investigated the potential mechanism. RESULTS In patients with AIS following IVT, logistic regression analysis showed that baseline serum LCN2 (p = 0.023) and ferritin (p = 0.046) levels were independently associated with HT. A positive correlation between serum LCN2 and ferritin levels was identified in patients with HT. In experimental studies, recombinant LCN2 (rLCN2) significantly aggravated BBB dysfunction and HT in the thromboembolic stroke rats after thrombolysis, whereas LCN2 inhibition by ZINC006440089 exerted opposite effects. Further mechanistic studies showed that, LCN2 promoted endothelial cell ferroptosis, accompanied by the induction of high mobility group box 1 (HMGB1) and the inhibition of nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins. Ferroptosis inhibitor ferrostatin-1 (fer-1) significantly restricted the LCN2-mediated BBB disruption. Transfection of LCN2 and HMGB1 siRNA inhibited the endothelial cell ferroptosis, and this effects was reversed by Nrf2 siRNA. CONCLUSION LCN2 aggravated BBB disruption after thrombolysis by promoting endothelial cell ferroptosis via regulating the HMGB1/Nrf2/HO-1 pathway, this may provide a promising therapeutic target for the prevention of HT after IVT.
Collapse
Affiliation(s)
- Jie Liu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Shu-Yan Pang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Ruo-Yu Zhao
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, Department of Neurology, First Hospital of Jilin University, Chang Chun, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, Department of Neurology, First Hospital of Jilin University, Chang Chun, China.
| |
Collapse
|
6
|
Ma Y, Liu C, Ren L, Li J, Xu Y, Liang J, Wang P. β-1,4-Galactosyltransferase 1 protects against cerebral ischemia injury in mice by suppressing ferroptosis via the TAZ/Nrf2/HO-1 signaling pathway. CNS Neurosci Ther 2024; 30:e70030. [PMID: 39233353 PMCID: PMC11374693 DOI: 10.1111/cns.70030] [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: 07/16/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Ischemic stroke leads a primary cause of mortality in human diseases, with a high disability rate worldwide. This study aims to investigate the function of β-1,4-galactosyltransferase 1 (B4galt1) in mouse brain ischemia/reperfusion (I/R) injury. METHODS Recombinant human B4galt1 (rh-B4galt1) was intranasally administered to the mice model of middle cerebral artery occlusion (MCAO)/reperfusion. In this study, the impact of rh-B4galt1 on cerebral injury assessed using multiple methods, including the neurological disability status scale, 2,3,5-triphenyltetrazolium chloride (TTC), Nissl and TUNEL staining. This study utilized laser speckle Doppler flowmeter to monitor the cerebral blood flow. Western blotting was performed to assess the protein expression levels, and fluorescence-labeled dihydroethidium method was performed to determine the superoxide anion generation. Assay kits were used for the measurement of iron, malondialdehyde (MDA) and glutathione (GSH) levels. RESULTS We demonstrated that rh-B4galt1 markedly improved neurological function, reduced cerebral infarct volume and preserved the completeness of blood-brain barrier (BBB) for preventing damage. These findings further illustrated that rh-B4galt1 alleviated oxidative stress, lipid peroxidation, as well as iron deposition induced by I/R. The vital role of ferroptosis was proved in brain injury. Furthermore, the rh-B4galt1 could increase the levels of TAZ, Nrf2 and HO-1 after I/R. And TAZ-siRNA and ML385 reversed the neuroprotective effects of rh-B4galt1. CONCLUSIONS The results indicated that rh-B4galt1 implements neuroprotective effects by modulating ferroptosis, primarily via upregulating TAZ/Nrf2/HO-1 pathway. Thus, B4galt1 could be seen as a promising novel objective for ischemic stroke therapy.
Collapse
Affiliation(s)
- Yao Ma
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chang Liu
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lili Ren
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jiachen Li
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yunhao Xu
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jia Liang
- Institute of Life Science, Jinzhou Medical University, Jinzhou, Liaoning, China
- Liaoning Provincial Key Laboratory of Neurodegenerative Diseases, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Peng Wang
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
- Liaoning Provincial Key Laboratory of Neurodegenerative Diseases, Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
7
|
Amato S, Averna M, Farsetti E, Guidolin D, Pedrazzi M, Gatta E, Candiani S, Maura G, Agnati LF, Cervetto C, Marcoli M. Control of Dopamine Signal in High-Order Receptor Complex on Striatal Astrocytes. Int J Mol Sci 2024; 25:8610. [PMID: 39201299 PMCID: PMC11354247 DOI: 10.3390/ijms25168610] [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: 06/28/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
The receptor-receptor interaction (RRI) of G protein-coupled receptors (GPCRs) leads to new functional entities that are conceptually distinct from the simple addition of signals mediated by the activation of the receptors that form the heteromers. Focusing on astrocytes, there is evidence for the existence of inhibitory and facilitatory RRIs, including the heteromers formed by the adenosine A2A and the dopamine D2 receptors, by A2A and the oxytocin receptor (OTR), and the D2-OTR heteromers. The possible involvement of these receptors in mosaicism has never been investigated in striatal astrocytes. By biophysical and functional approaches, we focused our attention on the existence of an A2A-D2-OTR high-order receptor complex and its role in modulating cytosolic calcium levels and endogenous glutamate release, when striatal astrocyte processes were stimulated with 4-aminopyridine. Functional data indicate a permissive role of OTR on dopamine signaling in the regulation of the glutamatergic transmission, and an inhibitory control mediated by A2A on both the D2-mediated signaling and on the OTR-facilitating effect on D2. Imaging biochemical and bioinformatic evidence confirmed the existence of the A2A-D2-OTR complex and its ternary structure in the membrane. In conclusion, the D2 receptor appears to be a hotspot in the control of the glutamate release from the astrocytic processes and may contribute to the regulation and integration of different neurotransmitter-mediated signaling in the striatum by the A2A-D2-OTR heterotrimers. Considering the possible selectivity of allosteric interventions on GPCRs organized as receptor mosaics, A2A-D2-OTR heterotrimers may offer selective pharmacological targets in neuropsychiatric disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Monica Averna
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Elisa Farsetti
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Via Gabelli 63, 35122 Padova, Italy
| | - Marco Pedrazzi
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Elena Gatta
- DIFILAB, Department of Physics, University of Genova, Via Dodecaneso 33, 16146 Genova, Italy
| | - Simona Candiani
- Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Via Largo Benzi 10, 16132 Genova, Italy
| | - Guido Maura
- Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy
| | - Luigi Francesco Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Via Largo Benzi 10, 16132 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | - Manuela Marcoli
- Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| |
Collapse
|
8
|
Kato C, Ueda K, Morimoto S, Takahashi S, Nakamura S, Ozawa F, Ito D, Daté Y, Okada K, Kobayashi N, Nakahara J, Okano H. Proteomic insights into extracellular vesicles in ALS for therapeutic potential of Ropinirole and biomarker discovery. Inflamm Regen 2024; 44:32. [PMID: 38997748 PMCID: PMC11241965 DOI: 10.1186/s41232-024-00346-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) hold the potential for elucidating the pathogenesis of amyotrophic lateral sclerosis (ALS) and serve as biomarkers. Notably, the comparative and longitudinal alterations in the protein profiles of EVs in serum (sEVs) and cerebrospinal fluid (CSF; cEVs) of sporadic ALS (SALS) patients remain uncharted. Ropinirole hydrochloride (ROPI; dopamine D2 receptor [D2R] agonist), a new anti-ALS drug candidate identified through induced pluripotent stem cell (iPSC)-based drug discovery, has been suggested to inhibit ALS disease progression in the Ropinirole Hydrochloride Remedy for Amyotrophic Lateral Sclerosis (ROPALS) trial, but its mechanism of action is not well understood. Therefore, we tried to reveal longitudinal changes with disease progression and the effects of ROPI on protein profiles of EVs. METHODS We collected serum and CSF at fixed intervals from ten controls and from 20 SALS patients participating in the ROPALS trial. Comprehensive proteomic analysis of EVs, extracted from these samples, was conducted using liquid chromatography/mass spectrometer (LC/MS). Furthermore, we generated iPSC-derived astrocytes (iPasts) and performed RNA sequencing on astrocytes with or without ROPI treatment. RESULTS The findings revealed notable disparities yet high congruity in sEVs and cEVs protein profiles concerning disease status, time and ROPI administration. In SALS, both sEVs and cEVs presented elevated levels of inflammation-related proteins but reduced levels associated with unfolded protein response (UPR). These results mirrored the longitudinal changes after disease onset and correlated with the revised ALS Functional Rating Scale (ALSFRS-R) at sampling time, suggesting a link to the onset and progression of SALS. ROPI appeared to counteract these changes, attenuating inflammation-related protein levels and boosting those tied to UPR in SALS, proposing an anti-ALS impact on EV protein profiles. Reverse translational research using iPasts indicated that these changes may partly reflect the DRD2-dependent neuroinflammatory inhibitory effects of ROPI. We have also identified biomarkers that predict diagnosis and disease progression by machine learning-driven biomarker search. CONCLUSIONS Despite the limited sample size, this study pioneers in reporting time-series proteomic alterations in serum and CSF EVs from SALS patients, offering comprehensive insights into SALS pathogenesis, ROPI-induced changes, and potential prognostic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Chris Kato
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Satoru Morimoto
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan.
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan.
| | - Shinichi Takahashi
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Saitama, 350-1298, Japan
| | - Shiho Nakamura
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Fumiko Ozawa
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Daisuke Ito
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yugaku Daté
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kensuke Okada
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Naoki Kobayashi
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan.
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan.
| |
Collapse
|
9
|
Xu L, Mi Y, Meng Q, Liu Y, Wang F, Zhang G, Liu Y, Chen G, Hou Y. Anti-inflammatory effects of quinolinyl analog of resveratrol targeting TLR4 in MCAO/R ischemic stroke rat model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155344. [PMID: 38493721 DOI: 10.1016/j.phymed.2024.155344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/25/2023] [Accepted: 01/07/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Among adults, stroke is the main causes of mortality and permanent disability. Neuroinflammation is one of the main causes of stoke-mediated neuronal death. Our previous study revealed that (E)-5-(2-(Quinolin-4-yl) vinyl) benzene-1, 3-diol (RV01), a quinolinyl analog of resveratrol, inhibits microglia-induced neuroinflammation and safeguards neurons from inflammatory harm. The preventive role of RV01 in ischemic stroke and its underlying cellular mechanisms and molecular targets remain poorly understood. PURPOSE To investigate whether RV01 alleviates ischemia-reperfusion (I/R) injury by inhibiting microglia-mediated neuroinflammation and determine the potential molecular mechanisms and targets by which RV01 inhibits the I/R-mediated microglia activation. METHODS Rat middle cerebral artery occlusion and reperfusion (MCAO/R) and BV-2 or primary microglial cells oxygen-glucose deprivation and reperfusion (OGD/R) models were established. The neurological behavior scores, 2, 3, 5-triphenyl tetrazolium chloride staining and immunofluorescence were used to detect the neuroprotective effect of RV01 in the MCAO/R rats. In addition, the mRNA expression levels of IL-6, TNF-α, and IL-1β were detected to reveal the antineuroinflammatory effect of RV01. Moreover, a western blot assay was performed to explore the protein expression changes in NF-κB-mediated neuroinflammation. Finally, we identified TLR4 as an RV01 target through molecular docking, drug sensitivity target stability analysis, cellular thermal shift analysis, and surface plasmon resonance techniques. RESULTS RV01 reduced the infarct volume and neurological deficits, increased the rotarod duration, and decreased the number of rightward deflections in the MCAO/R rats. RV01 inhibited the NF-κB signaling pathway in vitro and in vivo, as demonstrated by the reduction in the transcription factor p65-mediated expression of several inflammatory factors including IL-6, TNF-α, and IL-1β. Further studies showed that its protective effect was associated with targeting the TLR4 protein. Notably, the anti-inflammatory effect of RV01 was markedly reinforced by the TLR4 knockdown, but inhibited by the overexpression of TLR4. Results revealed that the conditioned medium derived from the RV01-treated BV-2 cells significantly decreased the OGD/R-mediated neuronal damage. CONCLUSION Our results are the first to reveal the protective effects of RV01 on cerebral ischemia, depending on its inhibitory effect on the NF-κB pathway by targeting TLR4. RV01 could be a potential protective agent in ischemic stroke treatment.
Collapse
Affiliation(s)
- Libin Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yan Mi
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Qingqi Meng
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yeshu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Feng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Guijie Zhang
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Key Laboratory of Vascular Biology, Science and Research Center, Shenyang Medical College, Shenyang, China.
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China.
| |
Collapse
|
10
|
Henedak NT, El-Abhar HS, Abdallah DM, Ahmed KA, Soubh AA. Demotion of canonical/non-canonical inflammasome and pyroptosis alleviates ischemia/reperfusion-induced acute kidney injury: Novel role of the D2/D3 receptor agonist ropinirole. Eur J Pharmacol 2024; 969:176460. [PMID: 38402931 DOI: 10.1016/j.ejphar.2024.176460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Ropinirole used to treat Parkinson's disease highly targets the dopaminergic receptor D3 over the D2 receptor but although both are expressed in the kidneys the ropinirole potential to treat kidney injury provoked by ischemia/reperfusion (I/R) is undraped. We investigated whether ropinirole can alleviate renal I/R by studying its anti-inflammatory, antioxidant, and anti-pyroptotic effects targeting its aptitude to inhibit the High-mobility group box 1/Toll-like receptor 4/Nuclear factor-kappa B (HMGB1/TLR4/NF-κB) cue and the canonical/non-canonical NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome trajectories. Herein, bilateral I/R surgery was induced in animals to be either untreated or treated with ropinirole for three days after the insult. Ropinirole successfully improved the histopathological picture and renal function which was confirmed by reducing cystatin C and the standard parameters creatinine and blood urea nitrogen (BUN). Ropinirole achieved this through its anti-inflammatory capacity mediated by reducing the HMGB1/TLR4 axis and inactivating NF-κB, which are upstream regulators of the NLRP3 pathway. As a result, the injurious inflammasome markers (NLRP3, apoptosis-associated speck-like protein (ASC), active caspase-1) and their target cytokines interleukin-1 beta (IL-1β) and IL-18 were decreased. Ropinirole also reduced the pyroptotic cell death markers caspase-11 and gasdermin-D. Furthermore, ropinirole by replenishing antioxidants and decreasing malondialdehyde helped to reduce oxidative stress in the kidneys. The docking findings confirmed that ropinirole highly binds to the dopaminergic D3 receptor more than to the D2 receptor. In conclusion, ropinirole has the potential to be a reno-therapeutic treatment against I/R insult by abating the inflammatory NLRP3 inflammasome signal, pyroptosis, and oxidative stress.
Collapse
Affiliation(s)
- Nada T Henedak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, 11835, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ayman A Soubh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, Egypt
| |
Collapse
|
11
|
Ma Y, Hu X, Shen S, Pan D. Geniposide ameliorates brain injury in mice with intracerebral hemorrhage by inhibiting NF-κB signaling. Neurol Res 2024; 46:346-355. [PMID: 38402902 DOI: 10.1080/01616412.2024.2321014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Neuroinflammation and oxidative stress are critical players in intracerebral hemorrhage (ICH). Geniposide is an active component of Gardenia that has anti-inflammatory effects. This study focused on the roles and mechanisms of geniposide in ICH. METHODS ICH was established by injecting collagenase IV into C57BL/6 mice. To determine the functions of geniposide and NF-κB inhibition in ICH model mice, geniposide (1, 25, or 50 mg/kg) or PDTC (a NF-κB inhibitor) was administered. Neurological functions were assessed with the modified neurological severity score (mNSS) test. Hematoxylin and eosin staining were performed to identify pathological changes. IL-1β and TNF-α levels were estimated with ELISA kits. NF-κB p65 localization was determined by immunofluorescence staining. Oxidative stress was analyzed by measuring ROS levels. RESULTS Geniposide alleviated cerebral edema and neurological deficits. Geniposide inhibited neuroinflammation and oxidative stress after ICH, and the inhibitory effects were enhanced by NF-κB inhibition. Additionally, geniposide inhibited NF-κB signaling. CONCLUSION Geniposide alleviates brain injury by suppressing inflammation and oxidative stress damage in experimental ICH models by inhibiting NF-κB signaling.
Collapse
Affiliation(s)
- Yinghui Ma
- Department of Neurosurgery, Huang Shi Central Hospital, HuangShi, China
| | - Xiao Hu
- Department of Neurosurgery, Huang Shi Central Hospital, HuangShi, China
| | - Songbo Shen
- Department of Neurosurgery, Huang Shi Central Hospital, HuangShi, China
| | - Dongmei Pan
- Department of Geriatrics, Huang Shi Central Hospital, HuangShi, China
| |
Collapse
|
12
|
Wee IC, Arulsamy A, Corrigan F, Collins-Praino L. Long-Term Impact of Diffuse Traumatic Brain Injury on Neuroinflammation and Catecholaminergic Signaling: Potential Relevance for Parkinson's Disease Risk. Molecules 2024; 29:1470. [PMID: 38611750 PMCID: PMC11013319 DOI: 10.3390/molecules29071470] [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: 01/05/2024] [Revised: 02/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Traumatic brain injury (TBI) is associated with an increased risk of developing Parkinson's disease (PD), though the exact mechanisms remain unclear. TBI triggers acute neuroinflammation and catecholamine dysfunction post-injury, both implicated in PD pathophysiology. The long-term impact on these pathways following TBI, however, remains uncertain. In this study, male Sprague-Dawley rats underwent sham surgery or Marmarou's impact acceleration model to induce varying TBI severities: single mild TBI (mTBI), repetitive mild TBI (rmTBI), or moderate-severe TBI (msTBI). At 12 months post-injury, astrocyte reactivity (GFAP) and microglial levels (IBA1) were assessed in the striatum (STR), substantia nigra (SN), and prefrontal cortex (PFC) using immunohistochemistry. Key enzymes and receptors involved in catecholaminergic transmission were measured via Western blot within the same regions. Minimal changes in these markers were observed, regardless of initial injury severity. Following mTBI, elevated protein levels of dopamine D1 receptors (DRD1) were noted in the PFC, while msTBI resulted in increased alpha-2A adrenoceptors (ADRA2A) in the STR and decreased dopamine beta-hydroxylase (DβH) in the SN. Neuroinflammatory changes were subtle, with a reduced number of GFAP+ cells in the SN following msTBI. However, considering the potential for neurodegenerative outcomes to manifest decades after injury, longer post-injury intervals may be necessary to observe PD-relevant alterations within these systems.
Collapse
Affiliation(s)
- Ing Chee Wee
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia;
| | - Frances Corrigan
- Head Injury Lab, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lyndsey Collins-Praino
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
13
|
Wang H, Zhao Y, Zhang D, Li J, Yang K, Yang J, Li B. Neuroprotective effects of quinpirole on lithium chloride pilocarpine-induced epilepsy in rats and its underlying mechanisms. Eur J Med Res 2024; 29:121. [PMID: 38355613 PMCID: PMC10865707 DOI: 10.1186/s40001-024-01694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
INTRODUCTION Epilepsy is a common neurological disorder that presents with challenging mechanisms and treatment strategies. This study investigated the neuroprotective effects of quinpirole on lithium chloride pilocarpine-induced epileptic rats and explored its potential mechanisms. METHODS Lithium chloride pilocarpine was used to induce an epileptic model in rats, and the effects of quinpirole on seizure symptoms and cognitive function were evaluated. The Racine scoring method, electroencephalography, and Morris water maze test were used to assess seizure severity and learning and memory functions in rats in the epileptic group. Additionally, immunohistochemistry and Western blot techniques were used to analyze the protein expression levels and morphological changes in glutamate receptor 2 (GluR2; GRIA2), BAX, and BCL2 in the hippocampi of rats in the epileptic group. RESULTS First, it was confirmed that the symptoms in rats in the epileptic group were consistent with features of epilepsy. Furthermore, these rats demonstrated decreased learning and memory function in the Morris water maze test. Additionally, gene and protein levels of GluR2 in the hippocampi of rats in the epileptic group were significantly reduced. Quinpirole treatment significantly delayed seizure onset and decreased the mortality rate after the induction of a seizure. Furthermore, electroencephalography showed a significant decrease in the frequency of the spike waves. In the Morris water maze test, rats from the quinpirole treatment group demonstrated a shorter latency period to reach the platform and an increased number of crossings through the target quadrant. Network pharmacology analysis revealed a close association between quinpirole and GluR2 as well as its involvement in the cAMP signaling pathway, cocaine addiction, and dopaminergic synapses. Furthermore, immunohistochemistry and Western blot analysis showed that quinpirole treatment resulted in a denser arrangement and a more regular morphology of the granule cells in the hippocampi of rats in the epileptic group. Additionally, quinpirole treatment decreased the protein expression of BAX and increased the protein expression of BCL2. CONCLUSION The current study demonstrated that quinpirole exerted neuroprotective effects in the epileptic rat model induced by lithium chloride pilocarpine. Additionally, it was found that the treatment not only alleviated the rats' seizure symptoms, but also improved their learning and memory abilities. This improvement was linked to the modulation of protein expression levels of GLUR2, BAX, and BCL2. These findings provided clues that would be important for further investigation of the therapeutic potential of quinpirole and its underlying mechanisms for epilepsy treatment.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Pediatrics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Yongheng Zhao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Dongqing Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jun Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Kun Yang
- Department of Pediatrics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Junli Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Baomin Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
14
|
Cheng X, Ren Z, Jia H, Wang G. METTL3 Mediates Microglial Activation and Blood-Brain Barrier Permeability in Cerebral Ischemic Stroke by Regulating NLRP3 Inflammasomes Through m6A Methylation Modification. Neurotox Res 2024; 42:15. [PMID: 38349604 DOI: 10.1007/s12640-024-00687-2] [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: 01/18/2023] [Revised: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 02/15/2024]
Abstract
Cerebral ischemic stroke (CIS) is the main cause of disability. METTL3 is implicated in CIS, and we explored its specific mechanism. Middle cerebral artery occlusion (MCAO) rat model and oxygen-glucose deprivation/reperfusion (OGD/R) HAPI cell model were established and treated with LV-METTL3 or DAA, oe-METTL3, miR-335-3p mimics, or DAA, to assess their effects on MCAO rat neurological and motor function, cerebral infarction area, brain water content, microglial activation, blood-brain barrier (BBB) permeability, and NLRP3 inflammasome activation. METTL3, pri-miR-335-3p, mature miR-335-3p, and miR-335-3p mRNA levels were assessed by RT-qPCR; M1/M2 microglial phenotype proportion and M1/M2 microglia ratio, inflammatory factor levels, and m6A modification were assessed. MCAO rats manifested cerebral ischemia injury. METTL3 was under-expressed in CIS. METTL3 overexpression inhibited microglial activation and M1 polarization and BBB permeability in MCAO rats and inhibited OGD/R-induced microglial activation and reduced M1 polarization. METTL3 regulated miR-335-3p expression and inhibited NLRP3 inflammasome activation. m6A methylation inhibition averted METTL3's effects on NLRP3 activation, thus promoting microglial activation in OGD/R-induced cells and METTL3's effects on BBB permeability in MCAO rats. Briefly, METTL3 regulated miR-335-3p expression through RNA m6A methylation and inhibited NLRP3 inflammasome activation, thus repressing microglial activation, BBB permeability, and protecting against CIS.
Collapse
Affiliation(s)
- Xue Cheng
- Department of Clinical Nutrition, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Zhetan Ren
- Clinical Medicine, The First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, China
| | - Huiyang Jia
- Neurology, Jinzhou Medical University, Jinzhou, 121000, China
| | - Gang Wang
- Department of Tumor Intervention, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, 121000, China.
| |
Collapse
|
15
|
Xu H, Dong J, Li Y, Zhang L, Yin J, Zhu C, Wang X, Ren K, Zhang H, Zhao D. Neuritin has a neuroprotective role in the rat model of acute ischemia stroke by inhibiting neuronal apoptosis and NLRP3 inflammasome. J Stroke Cerebrovasc Dis 2023; 32:107391. [PMID: 37832268 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107391] [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: 01/28/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVES This study explored the anti-inflammatory, anti-neuronal apoptosis, and neuroprotective effects of Neuritin in rat models of acute ischemia stroke (AIS). METHODS AIS was induced in male Sprague Dawley rats by middle cerebral artery occlusion (MCAO). Rats were divided into sham, MCAO, MCAO+neuritin, MCAO + neuritin + PBS, MCAO + neuritin+MCC950, and MCAO + neuritin + MSU groups. Neurological score assessment, brain water content measurement, HE staining, TTC staining, TUNEL staining, ELISA, and Western blot were performed. RESULTS Neuritin significantly improved the neurobehavioral score, infarct size, brain water content, apoptosis, and neuroinflammatory response compared with the MCAO and MCAO + PBS groups within 24 h after AIS. Moreover, Neuritin inhibited the protein expression of NLRP3 inflammasome, and reduced the expression of IL-18 and IL-1B, thereby reducing the inflammatory response. Meanwhile, the neuroprotection, anti-inflammation, and anti-apoptosis effects of Neuritin were enhanced by MCC950 but partly counteracted by MSU. CONCLUSION Neuritin may reduce brain injury after AIS by inhibiting the expression of NLRP3 inflammasome and then inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Hui Xu
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Jiangtao Dong
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Yang Li
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Lei Zhang
- Department of Neuromedicine, Beitun Hospital, the Tenth Division of Xinjiang Production and Construction Corps, Beitun 836000, China
| | - Jiangwen Yin
- Department of Anesthesiology, First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Chao Zhu
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Xu Wang
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Kunhao Ren
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Hao Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Dong Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China.
| |
Collapse
|
16
|
Gao H, Zhang Y, Luo D, Xu J, Tan S, Li Y, Qi W, Zhai Q, Wang Q. Activation of the Hippocampal DRD2 Alleviates Neuroinflammation, Synaptic Plasticity Damage and Cognitive Impairment After Sleep Deprivation. Mol Neurobiol 2023; 60:7208-7221. [PMID: 37543530 DOI: 10.1007/s12035-023-03514-5] [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: 05/04/2023] [Accepted: 07/15/2023] [Indexed: 08/07/2023]
Abstract
Sleep loss is commonplace nowadays and profoundly impacts cognition. Dopamine receptor D2 (DRD2) makes a specific contribution to cognition, although the precise mechanism underlying how DRD2 affects the cognitive process after sleep deprivation remains unclear. Herein, we observed cognitive impairment and impaired synaptic plasticity, including downregulation of synaptophysin and PSD95, decreased postsynaptic density thickness, neuron complexity, and spine density in chronic sleep restriction (CSR) mice. We also observed downregulated hippocampal DRD2 and Cryab expression in the CSR mice. Meanwhile, NF-κB translocation from the cytoplasm to the nucleus occurred, indicating that neuroinflammation ensued. However, hippocampal delivery of the DRD2 agonist quinpirole effectively rescued these changes. In vitro, quinpirole treatment significantly decreased the release of proinflammatory cytokines in microglial supernatant, indicating a potential anti-neuroinflammatory effect of Drd2/Cryab/NF-κB in CSR mice. Our study provided the evidence that activation of the Drd2 may relieve neuroinflammation and improve sleep deprivation-induced cognitive deficits.
Collapse
Affiliation(s)
- Hui Gao
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Anaesthesiology, Yan'an University Affiliated Hospital, Yan'an, 716000, China
| | - Yuxin Zhang
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200434, China
| | - Danlei Luo
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jing Xu
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shuwen Tan
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ying Li
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Wanling Qi
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Qian Zhai
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Qiang Wang
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
17
|
Akram M, Neha, Pinky, Saqib M, Salman M, Parvez S. Neuroprotective effect of ropinirole against Aβ 1-42 -induced neurochemical perturbations and cognitive impairments in a rodent model. Kaohsiung J Med Sci 2023; 39:1119-1128. [PMID: 37902529 DOI: 10.1002/kjm2.12770] [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: 06/11/2023] [Revised: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 10/31/2023] Open
Abstract
The primary objective of this study was to investigate the protective effects of ropinirole (ROP) medication given for an extended period following the induction of cognitive decline, oxidative stress, and deterioration of mitochondria in a Wistar rat model by Aβ1-42 . This study aimed to examine the neuroprotective efficacy of ROP in a stereotaxis model of AD. The Wistar rats were randomly assigned into four groups. Group I was considered as a sham, group II served as Aβ-infusion alone, Group III was Aβ1-42 + ROP (5 mg/kg/i.p.), and Group IV was Aβ1-42 + ROP (10 mg/kg/i.p.). Our research revealed that ROP (10 mg/kg, b.wt.) attenuates the cognitive deficits caused by Aβ1-42 -infused, which also correlates with the barnes maze, where (10 mg/kg, b.w.t.) shows significant improvement in spatial learning and memory. At the same time, ROP was rescued from oxidative damage, decreased lipid peroxidation rates, and inhibited acetylcholinesterase activity caused, demonstrating antioxidant benefits. In addition, a higher dose of ROP restored mitochondrial membrane potential in Aβ1-42 rats. Furthermore, histopathological examination showed that ROP treatment reduced neuronal loss, especially in the hippocampus. We conclude that ROP's protective effects in reducing oxidative stress and modulating mitochondrial function might have a propensity in AD pathogenesis.
Collapse
Affiliation(s)
- Mohd Akram
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi, India
- Department of Tahaffuzi Wa Samaji Tibb, School of Unani Medicine, Jamia Hamdard, New Delhi, India
| | - Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Pinky
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Saqib
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Salman
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
18
|
Furgiuele A, Pereira FC, Martini S, Marino F, Cosentino M. Dopaminergic regulation of inflammation and immunity in Parkinson's disease: friend or foe? Clin Transl Immunology 2023; 12:e1469. [PMID: 37781343 PMCID: PMC10540835 DOI: 10.1002/cti2.1469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/11/2022] [Accepted: 09/16/2023] [Indexed: 10/03/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease affecting 7-10 million people worldwide. Currently, there is no treatment available to prevent or delay PD progression, partially due to the limited understanding of the pathological events which lead to the death of dopaminergic neurons in the substantia nigra in the brain, which is known to be the cause of PD symptoms. The current available treatments aim at compensating dopamine (DA) deficiency in the brain using its precursor levodopa, dopaminergic agonists and some indirect dopaminergic agents. The immune system is emerging as a critical player in PD. Therefore, immune-based approaches have recently been proposed to be used as potential antiparkinsonian agents. It has been well-known that dopaminergic pathways play a significant role in regulating immune responses in the brain. Although dopaminergic agents are the primary antiparkinsonian treatments, their immune regulatory effect has yet to be fully understood. The present review summarises the current available evidence of the immune regulatory effects of DA and its mimics and discusses dopaminergic agents as antiparkinsonian drugs. Based on the current understanding of their involvement in the regulation of neuroinflammation in PD, we propose that targeting immune pathways involved in PD pathology could offer a better treatment outcome for PD patients.
Collapse
Affiliation(s)
- Alessia Furgiuele
- Center for Research in Medical PharmacologyUniversity of InsubriaVareseItaly
| | - Frederico C Pereira
- Faculty of Medicine, Institute of Pharmacology and Experimental TherapeuticsUniversity of CoimbraCoimbraPortugal
- Faculty of Medicine, Institute for Clinical and Biomedical Research (iCBR)University of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
- Clinical Academic Center of Coimbra (CACC)CoimbraPortugal
| | - Stefano Martini
- Center for Research in Medical PharmacologyUniversity of InsubriaVareseItaly
| | - Franca Marino
- Center for Research in Medical PharmacologyUniversity of InsubriaVareseItaly
| | - Marco Cosentino
- Center for Research in Medical PharmacologyUniversity of InsubriaVareseItaly
| |
Collapse
|
19
|
Liu J, Mu D, Xu J, Liu Y, Zhang G, Tang Y, Wang D, Wang F, Liang D, Hou Y. Inhibition of TLR4 Signaling by Isorhapontigenin Targeting of the AHR Alleviates Cerebral Ischemia/Reperfusion Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:13270-13283. [PMID: 37624928 DOI: 10.1021/acs.jafc.3c00152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Abstract
Ischemic stroke is a major risk factor in human health, yet there are no drugs to cure cerebral ischemia/reperfusion injury (CIRI). Inflammation plays a fundamental role in the consequences of CIRI. Isorhapontigenin (ISOR) exhibits great anti-inflammatory activity; however, it is unclear whether ISOR can treat ischemic stroke through an anti-inflammation effect. Here, middle cerebral artery occlusion/reperfusion (MCAO/R) was used to investigate the effects of ISOR on CIRI. The in vitro activity was measured in BV-2 cells exposed to oxygen-glucose deprivation/reperfusion. As measured by neurological scores, brain water content, and infarction, neurological dysfunction was improved in the ISOR group. The neuronal death and microglial activation in the ipsilateral cortex were reduced by ISOR. TLR4 signaling was significantly inhibited by ISOR in vivo and in vitro. By reverse molecular docking, cellular thermal shift, and drug affinity-responsive target stability assays, an aryl hydrocarbon receptor (AHR) was found to be a target of ISOR. Furthermore, AHR knockdown blocked the effect of ISOR on TLR4 signaling, suggesting that ISOR may regulate TLR4-mediated inflammation through AHR, thereby protecting neurons from CIRI. This study demonstrated that ISOR is a promising drug candidate for the treatment of ischemic stroke and provided a theoretical basis for the development of the medicinal value of ISOR-derived foods, such as grapes.
Collapse
Affiliation(s)
- Jingyu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110167, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang 110167, China
| | - Danyang Mu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110167, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang 110167, China
| | - Jikai Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110167, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang 110167, China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guijie Zhang
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Yue Tang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110167, China
| | - Dequan Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110167, China
| | - Feng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110167, China
| | - Dong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110167, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang 110167, China
| |
Collapse
|
20
|
Liang T, Zhu Z, Gong F, Yang X, Lei X, Lu L. Galectin-3 promotes brain injury by modulating the phenotype of microglia via binding TLR-4 after intracerebral hemorrhage. Aging (Albany NY) 2023; 15:9041-9058. [PMID: 37698533 PMCID: PMC10522396 DOI: 10.18632/aging.205014] [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: 04/17/2023] [Accepted: 08/20/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a stroke subtype with high mortality and disability rate, and neuroinflammation is involved in secondary brain injury. Galectin-3 (Gal-3) is one of the scaffold proteins of Galectins. Studies have indicated that Gal-3 plays an important role in the physiological and pathological state of the nervous system. Here we focus on the role of Gal-3 in ICH, especially in neuroinflammation. METHODS Injection of autologous blood into the right basal ganglia was used to simulate ICH injury, and the level of Gal-3 in brain was regulated by related means. The changes of Gal-3 were detected by western blot and immunofluorescence, the level of neuroinflammation by immunofluorescence staining and ELISA. Apoptosis and neuron loss were detected by TUNEL staining FJB staining and Nissl staining, and neurological deficits were judged by neurobehavioral tests. RESULTS The protein level of Gal-3 increased at 24 h after ICH. Downregulation of Gal-3 level can reduce the infiltration of M1-type microglia and peripheral inflammatory cells, thus alleviating post-ICH neuroinflammation, and reducing cell apoptosis and neuron loss in brain tissue. ICH-induced neurological damage was rescued. Meanwhile, the promotion in the expression level of Gal-3 increased neuroinflammatory activation and nerve cell death, aggravating ICH-induced brain injury. CONCLUSIONS This study proves that Gal-3 is involved in neuroinflammation and nerve damage after ICH. Gal-3 expression should not be encouraged early on to prevent neuroinflammation. which provides a new possibility for clinical treatment for ICH patients.
Collapse
Affiliation(s)
- Tianyu Liang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zheng Zhu
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fangxiao Gong
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaobo Yang
- Center for General Practice Medicine, Department of Nursing, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoju Lei
- Center for General Practice Medicine, Department of Nursing, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ling Lu
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Ma P, Ou Y. Correlation between the dopaminergic system and inflammation disease: a review. Mol Biol Rep 2023; 50:7043-7053. [PMID: 37382774 DOI: 10.1007/s11033-023-08610-2] [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: 03/09/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023]
Abstract
The dopaminergic system is inextricably linked with neurological diseases and addiction. In recent years, many studies have found that the dopaminergic system involves in inflammatory diseases, particularly neuroinflammatory diseases development; This review summarizes the studies of dopaminergic system in inflammatory diseases, and specifically highlights the mechanisms of how dopaminergic system regulates inflammation; In addition, we speculate that there are some cavities in current research, including mixed usage of inhibitors, agonists and lack of systematic controls; We expect this review would provide directions to future research of dopaminergic system and inflammatory diseases.
Collapse
Affiliation(s)
- Peng Ma
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, Jiangsu, China
| | - Yu Ou
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, Jiangsu, China.
| |
Collapse
|
22
|
Cervetto C, Maura G, Guidolin D, Amato S, Ceccoli C, Agnati LF, Marcoli M. Striatal astrocytic A2A-D2 receptor-receptor interactions and their role in neuropsychiatric disorders. Neuropharmacology 2023:109636. [PMID: 37321323 DOI: 10.1016/j.neuropharm.2023.109636] [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: 03/30/2023] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
It is now generally accepted that astrocytes are active players in synaptic transmission, so that a neurocentric perspective of the integrative signal communication in the central nervous system is shifting towards a neuro-astrocentric perspective. Astrocytes respond to synaptic activity, release chemical signals (gliotransmitters) and express neurotransmitter receptors (G protein-coupled and ionotropic receptors), thus behaving as co-actors with neurons in signal communication in the central nervous system. The ability of G protein-coupled receptors to physically interact through heteromerization, forming heteromers and receptor mosaics with new distinct signal recognition and transduction pathways, has been intensively studied at neuronal plasma membrane, and has changed the view of the integrative signal communication in the central nervous system. One of the best-known examples of receptor-receptor interaction through heteromerization, with relevant consequences for both the physiological and the pharmacological points of view, is given by adenosine A2A and dopamine D2 receptors on the plasma membrane of striatal neurons. Here we review evidence that native A2A and D2 receptors can interact through heteromerization at the plasma membrane of astrocytes as well. Astrocytic A2A-D2 heteromers were found able to control the release of glutamate from the striatal astrocyte processes. A2A-D2 heteromers on striatal astrocytes and astrocyte processes are discussed as far as their potential relevance in the control of glutamatergic transmission in striatum is concerned, including potential roles in glutamatergic transmission dysregulation in pathological conditions including schizophrenia or the Parkinson's disease.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy.
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Italy.
| | - Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Cristina Ceccoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Luigi F Agnati
- Department of Biochemical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy; Center of Excellence for Biomedical Research, University of Genova, Italy.
| |
Collapse
|
23
|
Bonilla H, Peluso MJ, Rodgers K, Aberg JA, Patterson TF, Tamburro R, Baizer L, Goldman JD, Rouphael N, Deitchman A, Fine J, Fontelo P, Kim AY, Shaw G, Stratford J, Ceger P, Costantine MM, Fisher L, O’Brien L, Maughan C, Quigley JG, Gabbay V, Mohandas S, Williams D, McComsey GA. Therapeutic trials for long COVID-19: A call to action from the interventions taskforce of the RECOVER initiative. Front Immunol 2023; 14:1129459. [PMID: 36969241 PMCID: PMC10034329 DOI: 10.3389/fimmu.2023.1129459] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/06/2023] [Indexed: 03/11/2023] Open
Abstract
Although most individuals recover from acute SARS-CoV-2 infection, a significant number continue to suffer from Post-Acute Sequelae of SARS-CoV-2 (PASC), including the unexplained symptoms that are frequently referred to as long COVID, which could last for weeks, months, or even years after the acute phase of illness. The National Institutes of Health is currently funding large multi-center research programs as part of its Researching COVID to Enhance Recover (RECOVER) initiative to understand why some individuals do not recover fully from COVID-19. Several ongoing pathobiology studies have provided clues to potential mechanisms contributing to this condition. These include persistence of SARS-CoV-2 antigen and/or genetic material, immune dysregulation, reactivation of other latent viral infections, microvascular dysfunction, and gut dysbiosis, among others. Although our understanding of the causes of long COVID remains incomplete, these early pathophysiologic studies suggest biological pathways that could be targeted in therapeutic trials that aim to ameliorate symptoms. Repurposed medicines and novel therapeutics deserve formal testing in clinical trial settings prior to adoption. While we endorse clinical trials, especially those that prioritize inclusion of the diverse populations most affected by COVID-19 and long COVID, we discourage off-label experimentation in uncontrolled and/or unsupervised settings. Here, we review ongoing, planned, and potential future therapeutic interventions for long COVID based on the current understanding of the pathobiological processes underlying this condition. We focus on clinical, pharmacological, and feasibility data, with the goal of informing future interventional research studies.
Collapse
Affiliation(s)
- Hector Bonilla
- Department of Medicine and Infectious Diseases, Stanford University, Palo Alto, CA, United States
| | - Michael J. Peluso
- Department of Medicine and Infectious Diseases, University of California, San Francisco, San Francisco, CA, United States
| | - Kathleen Rodgers
- Center for Innovations in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Judith A. Aberg
- Department of Medicine, Infectious Diseases, Icahn School of Medicine at Mount Sinai, Chief, Division of Infectious Disease, New York, NY, United States
| | - Thomas F. Patterson
- Department of Medicine, Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Robert Tamburro
- Division of Intramural Research, National Institute of Health, Bethesda, MD, United States
| | - Lawrence Baizer
- National Heart Lung and Blood Institute, Division of Lung Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jason D. Goldman
- Department of Medicine, Organ Transplant and Liver Center, Swedish Medical Center, Seattle, WA, United States
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Nadine Rouphael
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - Amelia Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Jeffrey Fine
- Department of Rehabilitation Medicine at New York University (NYU) Grossman School of Medicine, Physical Medicine and Rehabilitation Service, New York University (NYU), New York University Medical Center, New York, NY, United States
| | - Paul Fontelo
- Applied Clinical Informatics Branch, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States
| | - Arthur Y. Kim
- Department of Medicine at Harvard Medical School, Division of Infectious Disease, Boston, MA, United States
| | - Gwendolyn Shaw
- Research Triangle Institute (RTI), International, Durham, NC, United States
| | - Jeran Stratford
- Research Triangle Institute (RTI), International, Durham, NC, United States
| | - Patricia Ceger
- Research Triangle Institute (RTI), International, Durham, NC, United States
| | - Maged M. Costantine
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH, United States
| | - Liza Fisher
- Long COVID Families, Houston, TX, United States
| | - Lisa O’Brien
- Utah Covid-19 Long Haulers, Salt Lake City, UT, United States
| | | | - John G. Quigley
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Vilma Gabbay
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Sindhu Mohandas
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - David Williams
- Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Grace A. McComsey
- Department of Pediatrics and Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
24
|
Heteromerization of Dopamine D2 and Oxytocin Receptor in Adult Striatal Astrocytes. Int J Mol Sci 2023; 24:ijms24054677. [PMID: 36902106 PMCID: PMC10002782 DOI: 10.3390/ijms24054677] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
The ability of oxytocin (OT) to interact with the dopaminergic system through facilitatory D2-OT receptor (OTR) receptor-receptor interaction in the limbic system is increasingly considered to play roles in social or emotional behavior, and suggested to serve as a potential therapeutic target. Although roles of astrocytes in the modulatory effects of OT and dopamine in the central nervous system are well recognized, the possibility of D2-OTR receptor-receptor interaction in astrocytes has been neglected. In purified astrocyte processes from adult rat striatum, we assessed OTR and dopamine D2 receptor expression by confocal analysis. The effects of activation of these receptors were evaluated in the processes through a neurochemical study of glutamate release evoked by 4-aminopyridine; D2-OTR heteromerization was assessed by co-immunoprecipitation and proximity ligation assay (PLA). The structure of the possible D2-OTR heterodimer was estimated by a bioinformatic approach. We found that both D2 and OTR were expressed on the same astrocyte processes and controlled the release of glutamate, showing a facilitatory receptor-receptor interaction in the D2-OTR heteromers. Biochemical and biophysical evidence confirmed D2-OTR heterodimers on striatal astrocytes. The residues in the transmembrane domains four and five of both receptors are predicted to be mainly involved in the heteromerization. In conclusion, roles for astrocytic D2-OTR in the control of glutamatergic synapse functioning through modulation of astrocytic glutamate release should be taken into consideration when considering interactions between oxytocinergic and dopaminergic systems in striatum.
Collapse
|
25
|
Götz J, Wieters F, Fritz VJ, Käsgen O, Kalantari A, Fink GR, Aswendt M. Temporal and Spatial Gene Expression Profile of Stroke Recovery Genes in Mice. Genes (Basel) 2023; 14:454. [PMID: 36833381 PMCID: PMC9956317 DOI: 10.3390/genes14020454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Stroke patients show some degree of spontaneous functional recovery, but this is not sufficient to prevent long-term disability. One promising approach is to characterize the dynamics of stroke recovery genes in the lesion and distant areas. We induced sensorimotor cortex lesions in adult C57BL/6J mice using photothrombosis and performed qPCR on selected brain areas at 14, 28, and 56 days post-stroke (P14-56). Based on the grid walk and rotating beam test, the mice were classified into two groups. The expression of cAMP pathway genes Adora2a, Pde10a, and Drd2, was higher in poor- compared to well-recovered mice in contralesional primary motor cortex (cl-MOp) at P14&56 and cl-thalamus (cl-TH), but lower in cl-striatum (cl-Str) at P14 and cl-primary somatosensory cortex (cl-SSp) at P28. Plasticity and axonal sprouting genes, Lingo1 and BDNF, were decreased in cl-MOp at P14 and cl-Str at P28 and increased in cl-SSp at P28 and cl-Str at P14, respectively. In the cl-TH, Lingo1 was increased, and BDNF decreased at P14. Atrx, also involved in axonal sprouting, was only increased in poor-recovered mice in cl-MOp at P28. The results underline the gene expression dynamics and spatial variability and challenge existing theories of restricted neural plasticity.
Collapse
Affiliation(s)
- Jan Götz
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Frederique Wieters
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Veronika J. Fritz
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Olivia Käsgen
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Aref Kalantari
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Gereon R. Fink
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, 52425 Juelich, Germany
| | - Markus Aswendt
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| |
Collapse
|
26
|
Santamaria-Anzures J, Pérez-Ramos J, Mendoza-Pérez F, Godínez-Chaparro B. Pramipexole inhibits formalin-induce acute and long-lasting mechanical hypersensitivity via NF-kB pathway in rats. Drug Dev Res 2023; 84:253-261. [PMID: 36651647 DOI: 10.1002/ddr.22029] [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] [Revised: 12/09/2022] [Accepted: 12/25/2022] [Indexed: 01/19/2023]
Abstract
Pain is one of the most frequent causes for patients to seek medical care. It interferes with daily functioning and affects the quality of life of the patient. There is a clear need to investigate nonopioid or non-nonsteroidal anti-inflammatory drug alternatives for the treatment of pain. In this study, we determined the effect of acute pre- and posttreatment with pramipexole (PPX), a dopamine D2/D3 selective agonist, on formalin 1%-induced acute and long-lasting nociceptive behavior sensitivity in rats. Moreover, we sought to investigate whether the antiallodynic and antihyperalgesic effect induced by PPX was mediated through the nuclear factor-κB (NF-kB) signaling pathway. Moreover, acute systemic pretreatment with PPX (1 and 3 mg/kg, ip) suppressed the formalin-induced nociceptive behavior during both phases of the formalin test and the development of formalin-induced secondary mechanical allodynia and hyperalgesia in both paws. Acute systemic posttreatment with PPX (3 mg/kg, ip) reverted the formalin-induced long-lasting secondary mechanical allodynia and hyperalgesia. Furthermore, PPX inhibits the protein expression of NF-κB-p65 and the levels of tumor necrosis factor-α and interleukin-1β in the spinal cord of animals with secondary mechanical allodynia and hyperalgesia induced by formalin. These data suggest that PPX has a potential role in producing anti-inflammatory activity. Moreover, the antiallodynic and antihyperalgesic effects induced by PPX can be mediated through the NF-kB signaling pathway.
Collapse
Affiliation(s)
- Jazmín Santamaria-Anzures
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Julia Pérez-Ramos
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Felipe Mendoza-Pérez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Beatriz Godínez-Chaparro
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| |
Collapse
|
27
|
Wenk D, Khan S, Ignatchenko V, Hübner H, Gmeiner P, Weikert D, Pischetsrieder M, Kislinger T. Phosphoproteomic Analysis of Dopamine D2 Receptor Signaling Reveals Interplay of G Protein- and β-Arrestin-Mediated Effects. J Proteome Res 2023; 22:259-271. [PMID: 36508580 PMCID: PMC9831068 DOI: 10.1021/acs.jproteome.2c00707] [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] [Indexed: 12/15/2022]
Abstract
Leveraging biased signaling of G protein-coupled receptors has been proposed as a promising strategy for the development of drugs with higher specificity. However, the consequences of selectively targeting G protein- or β-arrestin-mediated signaling on cellular functions are not comprehensively understood. In this study, we utilized phosphoproteomics to gain a systematic overview of signaling induced by the four biased and balanced dopamine D2 receptor (D2R) ligands MS308, BM138, quinpirole, and sulpiride in an in vitro D2R transfection model. Quantification of 14,160 phosphosites revealed a low impact of the partial G protein agonist MS308 on cellular protein phosphorylation, as well as surprising similarities between the balanced agonist quinpirole and the inverse agonist sulpiride. Analysis of the temporal profiles of ligand-induced phosphorylation events showed a transient impact of the G protein-selective agonist MS308, whereas the β-arrestin-preferring agonist BM138 elicited a delayed, but more pronounced response. Functional enrichment analysis of ligand-impacted phosphoproteins and treatment-linked kinases confirmed multiple known functions of D2R signaling while also revealing novel effects, for example of MS308 on sterol regulatory element-binding protein-related gene expression. All raw data were deposited in MassIVE (MSV000089457).
Collapse
Affiliation(s)
- Deborah Wenk
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada
| | - Shahbaz Khan
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada
| | - Vladimir Ignatchenko
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada
| | - Harald Hübner
- Medicinal
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Medicinal
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Dorothee Weikert
- Medicinal
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Monika Pischetsrieder
- Food
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Thomas Kislinger
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada,Department
of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada,
| |
Collapse
|
28
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
29
|
Song A, Yang H, Wu G, Ren S, Wang L, Qin G, Mao Y. Study of Intracranial Hematoma Removal and High Intracranial Pressure Reduction Using a Novel Three-Needle Brain Puncture Technique. Int J Gen Med 2022; 15:8797-8805. [PMID: 36605333 PMCID: PMC9809359 DOI: 10.2147/ijgm.s392149] [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: 10/04/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
Objective The present study aimed to evaluate the clinical value of minimally invasive surgery for intracranial hematoma removal and high intracranial pressure (ICP) reduction using a novel three-needle brain puncture technique. Methods A total of 202 cases with supratentorial hematoma were analyzed, 54 of whom received three-needle brain puncture (study group), and the remaining cases received single-needle (control groups 1 and 2) and two-needle brain puncture (control group 3). The amount of intracranial hematoma removed, changes in ICP, retention time of puncture needle, volume of residual blood, the National Institute of Health Stroke Scale (NIHSS) score, and postoperative survival rate were used as indexes to evaluate patient outcomes. Results We found that three-needle brain puncture (study group) can remove more intracranial hematoma (P < 0.05) and achieve lower ICP (P < 0.05) than single- and two-needle brain puncture (control group). The needle retention time and volume of residual blood significantly decreased in the study group. Additionally, a statistically significant difference was observed in the NIHSS scores and survival rates between the study and control groups (P < 0.05). Conclusion These data suggest that three-needle minimally invasive stereotactic puncture can effectively remove hematoma, reduce ICP, decrease the degree of brain damage, and improve prognosis.
Collapse
Affiliation(s)
- Anjun Song
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China,Correspondence: Anjun Song, Email
| | - Hui Yang
- Guiyang Public Health Treatment Center, Guiyang, Guizhou, People’s Republic of China
| | - Guofeng Wu
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Siying Ren
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Likun Wang
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Guannan Qin
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Yuanhong Mao
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| |
Collapse
|
30
|
Dopamine D2 receptor agonist Bromocriptine ameliorates Aβ 1-42-induced memory deficits and neuroinflammation in mice. Eur J Pharmacol 2022; 938:175443. [PMID: 36470446 DOI: 10.1016/j.ejphar.2022.175443] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
Alzheimer's Disease (AD) is the most common neurodegenerative disease, which lacks disease-modifying therapeutics so far. Studies have shown that the dysfunction of the dopaminergic system is related to a variety of pathophysiology of AD, and the expression of Dopamine D2 receptor (DRD2) in the brains of AD patients and animal models is significantly downregulated, suggesting that DRD2 may represent a therapeutic target for AD. However, the strategy of targeting DRD2 for AD treatment still lacks some key experimental evidences. Here we show that DRD2 agonist Bromocriptine improved Aβ1-42 induced neuroinflammation, neuronal apoptosis, and memory deficits in mice. For animal study, the mice have injected intracerebroventricularly (i.c.v.) with Aβ1-42(410 pmol/5 μl) to induced AD cognitive deficit model (Mazzola et al., 2003; van der Stelt et al., 2006). After 7 days, Bromocriptine (2.5 mg/kg, 5 mg/kg and 10 mg/kg) or normal saline was administered intragastrically once a day for 30 days. Behavioral tests about the Y maze and Morris water maze in mice were initiated on the twenty-fourth day of drug administration for 7 days. In vivo and in vitro mechanism research revealed that Bromocriptine, via activating DRD2, promoted the recruitment of PP2A and JNK by scaffold protein β-arrestin 2, that repressed JNK-mediated transcription of proinflammatory cytokines and activation of NLRP3 inflammasome in microglia. Collectively, our findings suggest that Bromocriptine can ameliorate Aβ1-42 induced neuroinflammation and memory deficits in mice through DRD2/β-arrestin 2/PP2A/JNK signaling axis, which provides an experimental basis for the development of Bromocriptine as a drug for AD.
Collapse
|
31
|
Ren R, Fang Y, Sherchan P, Lu Q, Lenahan C, Zhang JH, Zhang J, Tang J. Kynurenine/Aryl Hydrocarbon Receptor Modulates Mitochondria-Mediated Oxidative Stress and Neuronal Apoptosis in Experimental Intracerebral Hemorrhage. Antioxid Redox Signal 2022; 37:1111-1129. [PMID: 35481813 PMCID: PMC9784632 DOI: 10.1089/ars.2021.0215] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 12/30/2022]
Abstract
Aims: Oxidative stress and neuronal apoptosis play crucial roles in the pathological processes of secondary injury after intracerebral hemorrhage (ICH). Aryl hydrocarbon receptor (AHR), together with its endogenous ligand kynurenine, is known to mediate free radical accumulation and neuronal excitotoxicity in central nervous systems. Herein, we investigate the pathological roles of kynurenine/AHR after ICH. Results: Endogenous AHR knockout alleviated reactive oxygen species accumulation and neuronal apoptosis in ipsilateral hemisphere at 48 h after ICH in mice. The ICH insult resulted in an increase of total and nucleus AHR protein levels and AHR transcriptional activity. Inhibition of AHR provided both short- and long- term neurological benefits by attenuating mitochondria-mediated oxidative stress and neuronal apoptosis after ICH in mice. RhoA-Bax signaling activated mitochondrial death pathway and participated in deleterious actions of AHR. Finally, we reported that exogenous kynurenine aggravated AHR activation and mediated the brain mentioned earlier. Male animals were used in the experiments. Innovation: We show for the first time that kynurenine/AHR mediates mitochondria death and free radical accumulation, at least partially via the RhoA/Bax signaling pathway. Pharmacological antagonists of AHR and kynurenine may ameliorate neurobehavioral function and improve the prognosis of patients with ICH. Conclusion: Kynurenine/AHR may serve as a potential therapeutic target to attenuate mitochondria-mediated oxidative stress and neuronal cells impairment in patients with ICH. Antioxid. Redox Signal. 37, 1111-1129.
Collapse
Affiliation(s)
- Reng Ren
- Department of Neurointensive Care Unit and The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Qin Lu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - John H. Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
- Department of Neurosurgery, and Loma Linda University School of Medicine, Loma Linda, California, USA
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Jianmin Zhang
- Department of Neurointensive Care Unit and The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
32
|
Yang Y, Zhong Z, Wang B, Wang Y. Xiaoyao San ameliorates high-fat diet-induced anxiety and depression via regulating gut microbiota in mice. Biomed Pharmacother 2022; 156:113902. [DOI: 10.1016/j.biopha.2022.113902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 11/02/2022] Open
|
33
|
Ding Z, Zhong Z, Wang J, Zhang R, Shao J, Li Y, Wu G, Tu H, Yuan W, Sun H, Wang Q. Inhibition of Dectin-1 Alleviates Neuroinflammatory Injury by Attenuating NLRP3 Inflammasome-Mediated Pyroptosis After Intracerebral Hemorrhage in Mice: Preliminary Study Results. J Inflamm Res 2022; 15:5917-5933. [PMID: 36274828 PMCID: PMC9579968 DOI: 10.2147/jir.s384020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/07/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Neuroinflammation plays an important role following intracerebral hemorrhage (ICH). NLRP3 inflammasome-mediated pyroptosis contributes to the mechanism of neuroinflammation. It has been reported that dendritic cell-associated C-type lectin-1 (Dectin-1) activation triggers inflammation in neurological diseases. However, the role of Dectin-1 on NLRP3 inflammasome-mediated pyroptosis after ICH remains unclear. Here, we aimed to explore the effect of Dectin-1 on NLRP3 inflammasome-mediated pyroptosis and neuroinflammation after ICH. METHODS Adult male C57BL/6 mice were used to establish the ICH model. Laminarin, an inhibitor of Dectin-1, was administered for intervention. Expression of Dectin-1 was evaluated by Western blot and immunofluorescence. Brain water content and neurobehavioral function were tested to assess brain edema and neurological performance. Western blot was conducted to evaluate the level of GSDMD-N. ELISA kits were used to measure the levels of IL-1β and IL-18. qRT-PCR and Western blot were performed to evaluate the expressions of NLRP3 inflammasome, IL-1β, and IL-18. RESULTS The expression of Dectin-1 increased following ICH, and Dectin-1 was expressed on microglia. In addition, inhibition of Dectin-1 by laminarin decreased brain edema and neurological impairment after ICH. Moreover, inhibition of Dectin-1 decreased the expression of pyroptosis-related protein, GSDMD-N, and inflammatory cytokines (IL-1β and IL-18). Mechanistically, Dectin-1 blockade inhibits NLRP3 inflammasome activation, thereby alleviating neuroinflammatory injury by attenuating NLRP3 inflammasome-mediated pyroptosis both in vivo and in vitro. CONCLUSION Our study indicates that the inhibition of Dectin-1 alleviates neuroinflammation by attenuating NLRP3 inflammasome-mediated pyroptosis after ICH.
Collapse
Affiliation(s)
- Zhiquan Ding
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Zhenzhong Zhong
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jun Wang
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Run Zhang
- Neurosurgery Center, Department of Neuro-oncological Surgery, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jinlian Shao
- Department of Emergency, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yulong Li
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Guiwei Wu
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Huiru Tu
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Wen Yuan
- Laboratory Animal Center, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Haitao Sun
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China,Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Qinghua Wang
- Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China,Department of Emergency, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China,Correspondence: Qinghua Wang; Haitao Sun, Neurosurgery Center, Department of Neurotrauma and Neurocritical Care Medicine, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China, Email ;
| |
Collapse
|
34
|
Ugalde-Muñiz P, Hernández-Luna MG, García-Velasco S, Lugo-Huitrón R, Murcia-Ramírez J, Martínez-Tapia RJ, Noriega-Navarro R, Navarro L. Activation of dopamine D2 receptors attenuates neuroinflammation and ameliorates the memory impairment induced by rapid eye movement sleep deprivation in a murine model. Front Neurosci 2022; 16:988167. [PMID: 36278007 PMCID: PMC9579422 DOI: 10.3389/fnins.2022.988167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
The proinflammatory state, which may be induced by sleep deprivation, seems to be a determining factor in the development of neurodegenerative processes. Investigations of mechanisms that help to mitigate the inflammatory effects of sleep disorders are important. A new proposal involves the neurotransmitter dopamine, which may modulate the progression of the immune response by activating receptors expressed on immune cells. This study aimed to determine whether dopamine D2 receptor (D2DR) activation attenuates the proinflammatory response derived from rapid eye movement (REM) sleep deprivation in mice. REM sleep deprivation (RSD) was induced in 2-month-old male CD1 mice using the multiple platform model for three consecutive days; during this period, the D2DR receptor agonist quinpirole (QUIN) was administered (2 mg/kg/day i.p.). Proinflammatory cytokine levels were assessed in serum and homogenates of the brain cortex, hippocampus, and striatum using ELISAs. Long-term memory deficits were identified using the Morris water maze (MWM) and novel object recognition (NOR) tests. Animals were trained until learning criteria were achieved; then, they were subjected to RSD and treated with QUIN for 3 days. Memory evocation was determined afterward. Moreover, we found RSD induced anhedonia, as measured by the sucrose consumption test, which is commonly related to the dopaminergic system. Our data revealed increased levels of proinflammatory cytokines (TNFα and IL-1β) in both the hippocampus and serum from RSD mice. However, QUIN attenuated the increased levels of these cytokines. Furthermore, RSD caused a long-term memory evocation deficit in both the MWM and NOR tests. In contrast, QUIN coadministration during the RSD period significantly improved the performance of the animals. On the other hand, QUIN prevented the anhedonic condition induced by RSD. Based on our results, D2DR receptor activation protects against memory impairment induced by disturbed REM sleep by inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Perla Ugalde-Muñiz
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - María Guadalupe Hernández-Luna
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Stephany García-Velasco
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Rafael Lugo-Huitrón
- Laboratory of Behavioral Neurobiology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Jimena Murcia-Ramírez
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Ricardo Jesus Martínez-Tapia
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Roxana Noriega-Navarro
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Luz Navarro
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
- *Correspondence: Luz Navarro,
| |
Collapse
|
35
|
Guo X, Zhang Y, Liu C, Ren L, Gao S, Bi J, Liang J, Wang P. Intranasal administration of β‐1, 3‐galactosyltransferase 2 confers neuroprotection against ischemic stroke by likely inhibiting oxidative stress and
NLRP3
inflammasome activation. FASEB J 2022; 36:e22542. [DOI: 10.1096/fj.202200456rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/10/2022] [Accepted: 08/29/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Xun Guo
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province Jinzhou Medical University Jinzhou China
| | - Yang Zhang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province Jinzhou Medical University Jinzhou China
| | - Chang Liu
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province Jinzhou Medical University Jinzhou China
| | - Lili Ren
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province Jinzhou Medical University Jinzhou China
| | - Shuang Gao
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province Jinzhou Medical University Jinzhou China
| | - Jing Bi
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province Jinzhou Medical University Jinzhou China
| | - Jia Liang
- Institute of Life Science Jinzhou Medical University Jinzhou China
| | - Peng Wang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province Jinzhou Medical University Jinzhou China
| |
Collapse
|
36
|
Hong H, Lu X, Lu Q, Huang C, Cui Z. Potential therapeutic effects and pharmacological evidence of sinomenine in central nervous system disorders. Front Pharmacol 2022; 13:1015035. [PMID: 36188580 PMCID: PMC9523510 DOI: 10.3389/fphar.2022.1015035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
Sinomenine is a natural compound extracted from the medicinal plant Sinomenium acutum. Its supplementation has been shown to present benefits in a variety of animal models of central nervous system (CNS) disorders, such as cerebral ischemia, intracerebral hemorrhage, traumatic brain injury (TBI), Alzheimer’s disease (AD), Parkinson’s disease (PD), epilepsy, depression, multiple sclerosis, morphine tolerance, and glioma. Therefore, sinomenine is now considered a potential agent for the prevention and/or treatment of CNS disorders. Mechanistic studies have shown that inhibition of oxidative stress, microglia- or astrocyte-mediated neuroinflammation, and neuronal apoptosis are common mechanisms for the neuroprotective effects of sinomenine. Other mechanisms, including activation of nuclear factor E2-related factor 2 (Nrf2), induction of autophagy in response to inhibition of protein kinase B (Akt)-mammalian target of rapamycin (mTOR), and activation of cyclic adenosine monophosphate-response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF), may also mediate the anti-glioma and neuroprotective effects of sinomenine. Sinomenine treatment has also been shown to enhance dopamine receptor D2 (DRD2)-mediated nuclear translocation of αB-crystallin (CRYAB) in astrocytes, thereby suppressing neuroinflammation via inhibition of Signal Transducer and Activator of Transcription 3 (STAT3). In addition, sinomenine supplementation can suppress N-methyl-D-aspartate (NMDA) receptor-mediated Ca2+ influx and induce γ-aminobutyric acid type A (GABAA) receptor-mediated Cl− influx, each of which contributes to the improvement of morphine dependence and sleep disturbance. In this review, we outline the pharmacological effects and possible mechanisms of sinomenine in CNS disorders to advance the development of sinomenine as a new drug for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Hongxiang Hong
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, Nantong, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Zhiming Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- *Correspondence: Zhiming Cui,
| |
Collapse
|
37
|
Fatima S, Ali M, Quadri SN, Beg S, Samim M, Parvez S, Abdin MZ, Mishra P, Ahmad FJ. Crafting ɣ-L-Glutamyl-l-Cysteine layered Human Serum Albumin-nanoconstructs for brain targeted delivery of ropinirole to attenuate cerebral ischemia/reperfusion injury via "3A approach". Biomaterials 2022; 289:121805. [PMID: 36162213 DOI: 10.1016/j.biomaterials.2022.121805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/23/2022] [Accepted: 09/11/2022] [Indexed: 11/17/2022]
Abstract
Treatment of Ischemic Stroke is inordinately challenging due to its complex aetiology and constraints in shuttling therapeutics across blood-brain barrier. Ropinirole hydrochloride (Rp), a propitious neuroprotectant with anti-oxidant, anti-inflammatory, and anti-apoptotic properties (3A) is repurposed for remedying ischemic stroke and reperfusion (I/R) injury. The drug's low bioavailability in brain however, limits its therapeutic efficacy. The current research work has reported sub-100 nm gamma-L-Glutamyl-L-Cysteine coated Human Serum Albumin nanoparticles encapsulating Rp (C-Rp-NPs) for active targeting in ischemic brain to encourage in situ activity and reduce unwanted toxicities. Confocal microscopy and brain distribution studies confirmed the enhanced targeting potentiality of optimized C-Rp-NPs. The pharmacokinetics elucidated that C-Rp-NPs could extend Rp retention in systemic circulation and escalate bioavailability compared with free Rp solution (Rp-S). Additionally, therapeutic assessment in transient middle cerebral occlusion (tMCAO) model suggested that C-Rp-NPs attenuated the progression of I/R injury with boosted therapeutic index at 1000 times less concentration compared to Rp-S via reinstating neurological and behavioral deficits, while reducing ischemic neuronal damage. Moreover, C-Rp-NPs blocked mitochondrial permeability transition pore (mtPTP), disrupted apoptotic mechanisms, curbed oxidative stress and neuroinflammation, and elevated dopamine levels post tMCAO. Thus, our work throws light on fabrication of rationally designed C-Rp-NPs with enormous clinical potential.
Collapse
Affiliation(s)
- Saman Fatima
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110062, India
| | - Mubashshir Ali
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Syed Naved Quadri
- Centre for Transgenic Plant Development (CTPD), Department of Biotechnology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Sarwar Beg
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, United Kingdom
| | - M Samim
- Department of Chemistry, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Malik Zainul Abdin
- Centre for Transgenic Plant Development (CTPD), Department of Biotechnology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Prashant Mishra
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
38
|
Yin X, Wang J, Yang S, Li H, Shen H, Wang H, Li X, Chen G. Sam50 exerts neuroprotection by maintaining the mitochondrial structure during experimental cerebral ischemia/reperfusion injury in rats. CNS Neurosci Ther 2022; 28:2230-2244. [PMID: 36074556 PMCID: PMC9627377 DOI: 10.1111/cns.13967] [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: 04/25/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the role of Sam50, a barrel protein on the surface of the mitochondrial outer membrane, in cerebral ischemia-reperfusion (I/R) injury and its underlying mechanisms. METHODS A middle cerebral artery occlusion/reperfusion (MCAO/R) model in adult male Sprague-Dawley rats was established in vivo, and cultured neurons were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) to simulate I/R injury in vitro. Lentiviral vector encoding Sam50 or Sam50 shRNA was constructed and administered to rats by intracerebroventricular injection to overexpress and knockdown Sam50, respectively. RESULTS First, after MCAO/R induction, the mitochondrial structure was damaged, and Sam50 protein levels were increased responsively both in vivo and in vitro. Then, it was found that Sam50 overexpression could reduce infarction size, inhibit neuronal cell death, improve neurobehavioral disability, protect mitochondrial structure integrity, and ameliorate mitochondrial dysfunction, which was induced by I/R injury both in vivo and in vitro. However, Sam50 downregulation showed the opposite results and aggravated I/R injury by inducing neuronal cell death, neurobehavioral disability, and mitochondrial dysfunction. Moreover, we found that the interaction between Sam50 and Mic19 was broken off after OGD/R, showing that the Sam50-Mic19-Mic60 axis was breakage in neurons, which would be a reason for mitochondrial structure and function abnormalities induced by I/R injury. CONCLUSION Sam50 played a vital role in the protection of neurons and mitochondria in cerebral I/R injury, which could be a novel target for mitochondrial protection and ameliorating I/R injury.
Collapse
Affiliation(s)
- Xulong Yin
- Department of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina,Institute of Stroke ResearchSoochow UniversitySuzhouChina
| | - Jiahe Wang
- Institute of Stroke ResearchSoochow UniversitySuzhouChina,Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Siyuan Yang
- Institute of Stroke ResearchSoochow UniversitySuzhouChina,Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Haiying Li
- Institute of Stroke ResearchSoochow UniversitySuzhouChina,Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Haitao Shen
- Institute of Stroke ResearchSoochow UniversitySuzhouChina,Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hui Wang
- Department of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina,Institute of Stroke ResearchSoochow UniversitySuzhouChina
| | - Xiang Li
- Institute of Stroke ResearchSoochow UniversitySuzhouChina,Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Gang Chen
- Institute of Stroke ResearchSoochow UniversitySuzhouChina,Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
39
|
Cincotta AH, Cersosimo E, Alatrach M, Ezrokhi M, Agyin C, Adams J, Chilton R, Triplitt C, Chamarthi B, Cominos N, DeFronzo RA. Bromocriptine-QR Therapy Reduces Sympathetic Tone and Ameliorates a Pro-Oxidative/Pro-Inflammatory Phenotype in Peripheral Blood Mononuclear Cells and Plasma of Type 2 Diabetes Subjects. Int J Mol Sci 2022; 23:ijms23168851. [PMID: 36012132 PMCID: PMC9407769 DOI: 10.3390/ijms23168851] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Bromocriptine-QR is a sympatholytic dopamine D2 agonist for the treatment of type 2 diabetes that has demonstrated rapid (within 1 year) substantial reductions in adverse cardiovascular events in this population by as yet incompletely delineated mechanisms. However, a chronic state of elevated sympathetic nervous system activity and central hypodopaminergic function has been demonstrated to potentiate an immune system pro-oxidative/pro-inflammatory condition and this immune phenotype is known to contribute significantly to the advancement of cardiovascular disease (CVD). Therefore, the possibility exists that bromocriptine-QR therapy may reduce adverse cardiovascular events in type 2 diabetes subjects via attenuation of this underlying chronic pro-oxidative/pro-inflammatory state. The present study was undertaken to assess the impact of bromocriptine-QR on a wide range of immune pro-oxidative/pro-inflammatory biochemical pathways and genes known to be operative in the genesis and progression of CVD. Inflammatory peripheral blood mononuclear cell biology is both a significant contributor to cardiovascular disease and also a marker of the body’s systemic pro-inflammatory status. Therefore, this study investigated the effects of 4-month circadian-timed (within 2 h of waking in the morning) bromocriptine-QR therapy (3.2 mg/day) in type 2 diabetes subjects whose glycemia was not optimally controlled on the glucagon-like peptide 1 receptor agonist on (i) gene expression status (via qPCR) of a wide array of mononuclear cell pro-oxidative/pro-inflammatory genes known to participate in the genesis and progression of CVD (OXR1, NRF2, NQO1, SOD1, SOD2, CAT, GSR, GPX1, GPX4, GCH1, HMOX1, BiP, EIF2α, ATF4, PERK, XBP1, ATF6, CHOP, GSK3β, NFkB, TXNIP, PIN1, BECN1, TLR2, TLR4, TLR10, MAPK8, NLRP3, CCR2, GCR, L-selectin, VCAM1, ICAM1) and (ii) humoral measures of sympathetic tone (norepinephrine and normetanephrine), whole-body oxidative stress (nitrotyrosine, TBARS), and pro-inflammatory factors (IL-1β, IL-6, IL-18, MCP-1, prolactin, C-reactive protein [CRP]). Relative to pre-treatment status, 4 months of bromocriptine-QR therapy resulted in significant reductions of mRNA levels in PBMC endoplasmic reticulum stress-unfolded protein response effectors [GRP78/BiP (34%), EIF2α (32%), ATF4 (29%), XBP1 (25%), PIN1 (14%), BECN1 (23%)], oxidative stress response proteins [OXR1 (31%), NRF2 (32%), NQO1 (39%), SOD1 (52%), CAT (26%), GPX1 (33%), GPX4 (31%), GCH1 (30%), HMOX1 (40%)], mRNA levels of TLR pro-inflammatory pathway proteins [TLR2 (46%), TLR4 (20%), GSK3β (19%), NFkB (33%), TXNIP (18%), NLRP3 (32%), CCR2 (24%), GCR (28%)], mRNA levels of pro-inflammatory cellular receptor proteins CCR2 and GCR by 24% and 28%, and adhesion molecule proteins L-selectin (35%) and VCAM1 (24%). Relative to baseline, bromocriptine-QR therapy also significantly reduced plasma levels of norepinephrine and normetanephrine by 33% and 22%, respectively, plasma pro-oxidative markers nitrotyrosine and TBARS by 13% and 10%, respectively, and pro-inflammatory factors IL-18, MCP1, IL-1β, prolactin, and CRP by 21%,13%, 12%, 42%, and 45%, respectively. These findings suggest a unique role for circadian-timed bromocriptine-QR sympatholytic dopamine agonist therapy in reducing systemic low-grade sterile inflammation to thereby reduce cardiovascular disease risk.
Collapse
Affiliation(s)
- Anthony H. Cincotta
- VeroScience LLC, Tiverton, RI 02878, USA
- Correspondence: ; Tel.: +1-401-816-0525
| | - Eugenio Cersosimo
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Mariam Alatrach
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | - Christina Agyin
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - John Adams
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Robert Chilton
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Curtis Triplitt
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | | | - Ralph A. DeFronzo
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
40
|
Li X, Wang B, Yu N, Yang L, Nan C, Sun Z, Guo L, Zhao Z. Gabapentin Alleviates Brain Injury in Intracerebral Hemorrhage Through Suppressing Neuroinflammation and Apoptosis. Neurochem Res 2022; 47:3063-3075. [PMID: 35809188 DOI: 10.1007/s11064-022-03657-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/29/2022]
Abstract
Neuroinflammation plays an important role in brain tissue injury during intracerebral hemorrhage. Gabapentin can reduce inflammation and oxidative stress through inhibiting nuclear factor κB (NFκB) signals. Here, we showed that gabapentin reduced brain tissue injury in ICH through suppressing NFκB-mediated neuroinflammation. ICH was induced by injecting collagenase IV into the right striatum of Sprague-Dawley rats. PC12 and BV2 cells injury induced by Hemin were used to simulate ICH in vitro. Inflammation and apoptosis were assessed in rat brain tissue and in vitro cells. The neurobehavioral scores were significantly decreased in ICH rats compared with sham rats. Phosphorylated IκB-α and cleaved caspase3, and apoptosis rate were significantly higher in tissue surrounding the hematoma than in brain tissues from rats subjected to sham surgery. Furthermore, serum IL-6 levels in ICH rats were higher than in sham rats. Gabapentin treatment significantly improved the behavioral scores, decreased levels of phosphorylated IκB-α and cleaved caspase3, apoptosis rate, and serum IL-6 level in ICH rats. Hemin-treated BV2 cells displayed higher levels of phosphorylated IκB-α, cleaved caspase3, and IL-6 in the supernatant compared with vehicle-treated cells. Hemin treatment induced a significantly lower level of peroxisome proliferator-activated receptor γ (PPARγ) in BV2 cells. BV2-PC12 co-culture cells treated by hemin displayed higher levels of cleaved caspase3 in PC12 cells. Furthermore, gabapentin treatment could reduce these effects induced by hemin and the protective effects of gabapentin were significantly attenuated by PPARγ inhibitor. Therefore, gabapentin may reduce inflammation and apoptosis induced by the ICH through PPARγ-NFκB pathway.
Collapse
Affiliation(s)
- Xiaopeng Li
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.,Department of Neurosurgery, The First Hospital of Handan City, Handan, 056000, HeBei, China
| | - Bingqian Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.,Department of Neurosurgery, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, 054000, HeBei, China
| | - Ning Yu
- Department of Anesthesiology and Intensive Care Unit, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, HeBei, China
| | - Liang Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China
| | - Zhimin Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.,Department of Neurosurgery, The Third Hospital of Shijiazhuang City, Shijiazhuang, 050000, HeBei, China
| | - Lisi Guo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.
| |
Collapse
|
41
|
Intervention of neuroinflammation in the traumatic brain injury trajectory: In vivo and clinical approaches. Int Immunopharmacol 2022; 108:108902. [PMID: 35729835 DOI: 10.1016/j.intimp.2022.108902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/25/2022] [Accepted: 05/24/2022] [Indexed: 12/11/2022]
|
42
|
Zhang Y, Guo X, Peng Z, Liu C, Ren L, Liang J, Wang P. Nicotinamide Mononucleotide Adenylyltransferase 1 Regulates Cerebral Ischemia-Induced Blood-Brain Barrier Disruption Through NAD +/SIRT1 Signaling Pathway. Mol Neurobiol 2022; 59:4879-4891. [PMID: 35657458 DOI: 10.1007/s12035-022-02903-6] [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: 11/09/2021] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
The molecular mechanisms of blood-brain barrier (BBB) disruption in the early stage after ischemic stroke are poorly understood. In the present study, we investigated the potential role of nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) in ischemia-induced BBB damage using an animal middle cerebral artery occlusion (MCAO) model of ischemic stroke. Recombinant human NMNAT1 (rh-NMNAT1) was administered intranasally and Sirtuin 1 (SIRT1) siRNA was administered by intracerebroventricular injection. Our results indicate that rh-NMNAT1 reduced infarct volume, improved functional outcome, and decreased BBB permeability in mice after ischemic stroke. Furthermore, rh-NMNAT1 prevented the loss of tight junction proteins (occludin and claudin-5) and reduced cell apoptosis in ischemic microvessels. NMNAT1-mediated BBB permeability was correlated with the elevation of nicotinamide adenine dinucleotide (NAD+)/NADH ratio and SIRT1 level in brain microvascular endothelial cells. In addition, rh-NMNAT1 treatment significantly decreased the levels of acetylated nuclear factor-κB, acetylated p53, and matrix metalloproteinase-9 in ischemic microvessels. Moreover, the protective effects of rh-NMNAT1 could be reversed by SIRT1 siRNA. In conclusion, these findings indicate that rh-NMNAT1 protects BBB integrity after cerebral ischemia via the NAD+/SIRT1 signaling pathway in brain microvascular endothelial cells. NMNAT1 may be a novel potential therapeutic target for reducing BBB disruption after ischemic stroke.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Xun Guo
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Zhifeng Peng
- Department of Physiology, Shanxi Datong University, Datong, 037009, Shanxi, China
| | - Chang Liu
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Lili Ren
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Jia Liang
- Institute of Life Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| | - Peng Wang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| |
Collapse
|
43
|
Du X, Luo L, Huang Q, Zhang J. Cortex metabolome and proteome analysis reveals chronic arsenic exposure via drinking water induces developmental neurotoxicity through hnRNP L mediated mitochondrial dysfunction in male rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 820:153325. [PMID: 35074374 DOI: 10.1016/j.scitotenv.2022.153325] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/09/2022] [Accepted: 01/18/2022] [Indexed: 06/14/2023]
Abstract
Lots of people are at the risk of arsenic-contaminated drinking water. Arsenic exposure was confirmed to be closely linked to neurocognitive deficits, particularly during childhood. The multi-omics approaches are known be well suitable for toxicological research. Thus, this study aimed to explore the molecular mechanisms of arsenic-induced learning and memory function impairments through the integrative proteome and metabolome analysis of cortex in rats. The weaned rats were exposed to arsenic-contaminated drinking water for six months to mimic the developmental exposure. 220 differential proteins and 19 differential metabolites were identified in the cortex, and nine potential biomarkers were found to be related to impaired Morris water maze (MWM) indicators. Chronic arsenic exposure affected the cognitive function by inducing the overproduction of amyloid-β (Aβ) peptides and the redox imbalance in the mitochondria. Glycolysis and tricarboxylic acid (TCA) cycle enhancement driven by the increased heterogeneous nuclear ribonucleoprotein L (hnRNP L) is a low-dose protective mechanism against arsenic-induced ATP deficiency and oxidative stress. Moreover, apoptosis is another important pathway of arsenic-induced neurotoxicity. This study provides new evidence about the alterations of proteins and metabolites in the cortex of the exposed rats under arsenic toxicity. These findings suggest hnRNP L could be a potential target for the treatment of arsenic-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiaoyan Du
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China; Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Lianzhong Luo
- Department of Pharmacy, Xiamen Medical College, China
| | - Qingyu Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Jie Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China.
| |
Collapse
|
44
|
Li M, Zhang C, Zhou L, Sun X, Wang T, Fu F. Continuous Activation of Dopamine Receptors Alleviates LPS-Induced Liver Injury in Mice via β-arrestin2 Dependent Akt/NF-κB Pathway. Front Pharmacol 2022; 13:853834. [PMID: 35359858 PMCID: PMC8963954 DOI: 10.3389/fphar.2022.853834] [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: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 11/14/2022] Open
Abstract
Many studies showed that dopamine receptors (DRs) agonists have anti-inflammatory effects. Rotigotine, a non-ergot dopamine receptor agonist, mainly actives DRD2/DRD3/DRD1. Rotigotine extended-release microspheres (RoMS) are a sustained-release formulation that can release sustainably rotigotine for more than 7 days after a single dose of RoMS. This study aimed to investigate whether RoMS can attenuate the lipopolysaccharide (LPS)-induced liver injury of mice. The liver injury was evaluated by assaying serum transaminase and observing histopathological changes. The levels of pro-inflammatory cytokines in serum were also detected. Western blot was employed to assay the expression of proteins in the Akt/NF-κB pathway. The results showed that pre-administration with a single dose of RoMS could inhibit the increase of serum transaminase induced by LPS, alleviate the pathological damage of liver tissue, and decrease the levels of tumor necrosis factor-α and interleukin-6. In addition, RoMS decreased Toll-like receptor 4 protein expression in liver tissue. RoMS mitigated liver injury by activating DRs and negatively regulating the β-arrestin2-dependent Akt/NF-κB signaling pathway. The effects of RoMS could be weakened or abolished by the specific DRD2 antagonist, R121. In conclusion, activation of DRs inhibited the releases of pro-inflammatory cytokines and alleviated the immune-mediated liver injury induced by LPS in mice. The anti-inflammatory mechanism of RoMS may be related to the regulation of the β-arrestin2-dependent Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Mingan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Ce Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Lin Zhou
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Xiaohui Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
45
|
Marchetti B, Giachino C, Tirolo C, Serapide MF. "Reframing" dopamine signaling at the intersection of glial networks in the aged Parkinsonian brain as innate Nrf2/Wnt driver: Therapeutical implications. Aging Cell 2022; 21:e13575. [PMID: 35262262 PMCID: PMC9009237 DOI: 10.1111/acel.13575] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/11/2022] [Accepted: 02/06/2022] [Indexed: 11/30/2022] Open
Abstract
Dopamine (DA) signaling via G protein-coupled receptors is a multifunctional neurotransmitter and neuroendocrine-immune modulator. The DA nigrostriatal pathway, which controls the motor coordination, progressively degenerates in Parkinson's disease (PD), a most common neurodegenerative disorder (ND) characterized by a selective, age-dependent loss of substantia nigra pars compacta (SNpc) neurons, where DA itself is a primary source of oxidative stress and mitochondrial impairment, intersecting astrocyte and microglial inflammatory networks. Importantly, glia acts as a preferential neuroendocrine-immune DA target, in turn, counter-modulating inflammatory processes. With a major focus on DA intersection within the astrocyte-microglial inflammatory network in PD vulnerability, we herein first summarize the characteristics of DA signaling systems, the propensity of DA neurons to oxidative stress, and glial inflammatory triggers dictating the vulnerability to PD. Reciprocally, DA modulation of astrocytes and microglial reactivity, coupled to the synergic impact of gene-environment interactions, then constitute a further level of control regulating midbrain DA neuron (mDAn) survival/death. Not surprisingly, within this circuitry, DA converges to modulate nuclear factor erythroid 2-like 2 (Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling, a key pathway for mDAn neurogenesis, neuroprotection, and immunomodulation, adding to the already complex "signaling puzzle," a novel actor in mDAn-glial regulatory machinery. Here, we propose an autoregulatory feedback system allowing DA to act as an endogenous Nrf2/Wnt innate modulator and trace the importance of DA receptor agonists applied to the clinic as immune modifiers.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology SectionMedical SchoolUniversity of CataniaCataniaItaly
- OASI Research Institute‐IRCCS, Troina (EN), ItalyTroinaItaly
| | | | - Cataldo Tirolo
- OASI Research Institute‐IRCCS, Troina (EN), ItalyTroinaItaly
| | - Maria F. Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology SectionMedical SchoolUniversity of CataniaCataniaItaly
| |
Collapse
|
46
|
Fei X, Dou YN, Wang L, Wu X, Huan Y, Wu S, He X, Lv W, Wei J, Fei Z. Homer1 promotes the conversion of A1 astrocytes to A2 astrocytes and improves the recovery of transgenic mice after intracerebral hemorrhage. J Neuroinflammation 2022; 19:67. [PMID: 35287697 PMCID: PMC8922810 DOI: 10.1186/s12974-022-02428-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background Inflammation induced by intracerebral hemorrhage (ICH) is one of the main causes of the high mortality and poor prognosis of patients with ICH. A1 astrocytes are closely associated with neuroinflammation and neurotoxicity, whereas A2 astrocytes are neuroprotective. Homer scaffolding protein 1 (Homer1) plays a protective role in ischemic encephalopathy and neurodegenerative diseases. However, the role of Homer1 in ICH-induced inflammation and the effect of Homer1 on the phenotypic conversion of astrocytes remain unknown. Methods Femoral artery autologous blood from C57BL/6 mice was used to create an ICH model. We use the A1 phenotype marker C3 and A2 phenotype marker S100A10 to detect astrocyte conversion after ICH. Homer1 overexpression/knock-down mice were constructed by adeno-associated virus (AAV) infection to explore the role of Homer1 and its mechanism of action after ICH. Finally, Homer1 protein and selumetinib were injected into in situ hemorrhage sites in the brains of Homer1flox/flox/Nestin-Cre+/− mice to study the efficacy of Homer1 in the treatment of ICH by using a mouse cytokine array to explore the potential mechanism. Results The expression of Homer1 peaked on the third day after ICH and colocalized with astrocytes. Homer1 promotes A1 phenotypic conversion in astrocytes in vivo and in vitro. Overexpression of Homer1 inhibits the activation of MAPK signaling, whereas Homer1 knock-down increases the expression of pathway-related proteins. The Homer1 protein and selumetinib, a non-ATP competitive MEK1/2 inhibitor, improved the outcome in ICH in Homer1flox/flox/Nestin-Cre+/− mice. The efficacy of Homer1 in the treatment of ICH is associated with reduced expression of the inflammatory factor TNFSF10 and increased expression of the anti-inflammatory factors activin A, persephin, and TWEAK. Conclusions Homer1 plays an important role in inhibiting inflammation after ICH by suppressing the A1 phenotype conversion in astrocytes. In situ injection of Homer1 protein may be a novel and effective method for the treatment of inflammation after ICH.
Collapse
Affiliation(s)
- Xiaowei Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Ya-Nan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Li Wang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Yu Huan
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Shuang Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Xin He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Weihao Lv
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China.
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
47
|
Berg SIT, Knapp J, Braunstein M, Shirriff C. The small heat shock protein HSPB5 attenuates the severity of lupus nephritis in lupus-prone mice. Autoimmunity 2022; 55:192-202. [PMID: 35137667 DOI: 10.1080/08916934.2022.2027921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Lupus nephritis (LN) is a common and serious complication of systemic lupus erythematosus. The current treatments for LN are accompanied with severe immunotoxicity and have limits of effectiveness. Since our in vitro experiments demonstrated that a small heat shock protein (HSP), alpha-B crystallin (HSPB5; CRYAB), selectively modulates myeloid cells towards anti-inflammatory and tolerogenic phenotypes, the aim of this study was to investigate whether HSPB5 can attenuate the severity of LN. MRL/lpr mice were treated intravenously with HSPB5 at 2.5 or 10 μg/dose twice per week after disease onset, from 11 to 21 weeks of age. Disease progression was monitored by weekly measurements of proteinuria, and sera, spleens, and kidneys were collected for assessment at the terminal time point. Treatment with 10 μg HSPB5 substantially reduced endocapillary proliferation and tubular atrophy, which significantly reduced proteinuria and blood urea nitrogen (BUN). Compared to vehicle, 10 μg HSPB5 treatment substantially decreased activation/proliferation of splenocytes, increased IL-10+ macrophages, T and B regulatory cells (Treg, Breg), increased serum IL-10, and lowered expression of IL-6 in kidneys, which correlated with improved kidney function and pathology. This study demonstrated the utility of exogenous human HSPB5 to attenuate severe nephropathy in MRL/lpr mice and provides evidence in favour of a novel therapeutic approach for lupus nephritis.
Collapse
|
48
|
LI T, WANG X, DONG K. Extraction and purification of oridonin from Rabdosia rubescens and its protective effect on intracerebral hemorrhage-induced brain injury in rats. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.24921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tie LI
- Tianjin University of Traditional Chinese Medicine, China
| | - Xueyan WANG
- Tianjin University of Traditional Chinese Medicine, China
| | - Kun DONG
- Tianjin University of Traditional Chinese Medicine, China
| |
Collapse
|
49
|
Yoshioka Y, Sugino Y, Yamamuro A, Ishimaru Y, Maeda S. Dopamine inhibits the expression of proinflammatory cytokines of microglial cells through the formation of dopamine quinone in the mouse striatum. J Pharmacol Sci 2022; 148:41-50. [PMID: 34924128 DOI: 10.1016/j.jphs.2021.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/18/2021] [Accepted: 10/06/2021] [Indexed: 12/28/2022] Open
Abstract
We previously reported that dopamine (DA) attenuated lipopolysaccharide (LPS)-induced expression of proinflammatory cytokines through the formation of DA quinone (DAQ) in murine microglial cell line BV-2 and primary murine microglial cells. To reveal whether DA inhibits the expression of proinflammatory cytokines of microglial cells through the formation of DAQ in the central nervous system (CNS), in this study, we examined the effect of DAQ on LPS-induced mRNA expression of proinflammatory cytokines in C57BL/6 mouse brain under two experimental conditions: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration and l-dopa/carbidopa administration. Acute MPTP administration reduced the number of tyrosine hydroxylase-positive cells in the substantia nigra, and decreased the level of quinoprotein, an indicator of DAQ formation, in the striatum. Real-time RT-PCR analysis revealed that intraperitoneal administration of LPS increased the mRNA levels of proinflammatory cytokines, including tumor-necrosis factor-α and interleukin-1β, in the striatum. These increases were enhanced in MPTP-treated mice. On the other hand, l-dopa/carbidopa administration increased the level of quinoprotein, attenuated the LPS-induced mRNA expression of proinflammatory cytokines, and reduced the LPS-induced increase in the number of microglial cells in the striatum. These results suggest that DA attenuate the expression of proinflammatory cytokines in microglia through the formation of DAQ in the CNS.
Collapse
Affiliation(s)
- Yasuhiro Yoshioka
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka, 573-0101, Japan.
| | - Yuta Sugino
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka, 573-0101, Japan
| | - Akiko Yamamuro
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka, 573-0101, Japan
| | - Yuki Ishimaru
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka, 573-0101, Japan
| | - Sadaaki Maeda
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka, 573-0101, Japan
| |
Collapse
|
50
|
Yang Y, Song J, Liu N, Wei G, Liu S, Zhang S, Jiang N, Yang H, Du G. Salvianolic acid A relieves cognitive disorder after chronic cerebral ischemia: Involvement of Drd2/Cryab/NF-κB pathway. Pharmacol Res 2022; 175:105989. [PMID: 34800628 DOI: 10.1016/j.phrs.2021.105989] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/03/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022]
Abstract
Chronic cerebral ischemia (CCI) refers to long-term hypoperfusion of cerebral blood flow with the main clinical manifestations of progressive cognitive impairment. The pathological mechanism of CCI is complex, and there is a lack of effective treatments. Salvianolic acid A (SalA) is a neuroprotective extract of Salvia miltiorrhiza with the effects of anti-inflammation and anti-apoptosis. In this study, the effect of SalA on cognitive function and Drd2/Cryab/NF-κB signaling pathway in rats with CCI was investigated. Morris water maze and open field test were used to observe the effects of SalA on the cognitive function of CCI rats. The pathological changes in the brain were observed by HE, Nissl, and LFB staining. TUNEL staining, enzyme-linked immunosorbent assay, and western blot analysis were used to detect the inflammatory and apoptosis in the cortex and hippocampus. The expression of Drd2/Cryab/NF-κB pathway-related molecules and Drd2 localization were detected by western blotting and dual immunofluorescence, respectively. SH-SY5Y cells were exposed to chronic hypoglycemic and hypoxic injury in vitro, and Drd2 inhibitor haloperidol was used to verify the involved pathway. The results showed that SalA could improve the cognitive function of CCI rats, reduce pathological damage of cortex and hippocampus, inhibit neuroinflammation and apoptosis, and suppress the activation of NF-κB by regulating Drd2/Cryab pathway. And SalA inhibited NF-κB activation and nuclear translocation in SH-SY5Y cells by upregulating Drd2/Cryab pathway, which was reversed by haloperidol interference. In conclusion, SalA could relieve CCI-induced cognitive impairment in rats, at least partly through the Drd2/Cryab/NF-κB pathway.
Collapse
Affiliation(s)
- Yujiao Yang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, PR China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Junke Song
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - NanNan Liu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 280 Waihuan East Road, Panyu District, Guangdong 510006, PR China
| | - Guangyi Wei
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 280 Waihuan East Road, Panyu District, Guangdong 510006, PR China
| | - Shan Liu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 280 Waihuan East Road, Panyu District, Guangdong 510006, PR China
| | - Sen Zhang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Nan Jiang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; School of Pharmacy, Henan University, North Section of Jinming Avenue, Kaifeng 475004, Henan, PR China
| | - Haiguang Yang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Guanhua Du
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, PR China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|