1
|
Garbizu M, Aguado L, Martín A. Alpha7 nicotinic receptors as potential theranostic targets for experimental stroke. Neural Regen Res 2024; 19:939-940. [PMID: 37862178 PMCID: PMC10749633 DOI: 10.4103/1673-5374.385294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/17/2023] [Accepted: 07/27/2023] [Indexed: 10/22/2023] Open
Affiliation(s)
- Maider Garbizu
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Laura Aguado
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Abraham Martín
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
2
|
Bernardino PN, Luo AS, Andrew PM, Unkel CM, Gonzalez MI, Gelli A, Lein PJ. Evidence Implicating Blood-Brain Barrier Impairment in the Pathogenesis of Acquired Epilepsy following Acute Organophosphate Intoxication. J Pharmacol Exp Ther 2024; 388:301-312. [PMID: 37827702 PMCID: PMC10801776 DOI: 10.1124/jpet.123.001836] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Organophosphate (OP) poisoning can trigger cholinergic crisis, a life-threatening toxidrome that includes seizures and status epilepticus. These acute toxic responses are associated with persistent neuroinflammation and spontaneous recurrent seizures (SRS), also known as acquired epilepsy. Blood-brain barrier (BBB) impairment has recently been proposed as a pathogenic mechanism linking acute OP intoxication to chronic adverse neurologic outcomes. In this review, we briefly describe the cellular and molecular components of the BBB, review evidence of altered BBB integrity following acute OP intoxication, and discuss potential mechanisms by which acute OP intoxication may promote BBB dysfunction. We highlight the complex interplay between neuroinflammation and BBB dysfunction that suggests a positive feedforward interaction. Lastly, we examine research from diverse models and disease states that suggest mechanisms by which loss of BBB integrity may contribute to epileptogenic processes. Collectively, the literature identifies BBB impairment as a convergent mechanism of neurologic disease and justifies further mechanistic research into how acute OP intoxication causes BBB impairment and its role in the pathogenesis of SRS and potentially other long-term neurologic sequelae. Such research is critical for evaluating BBB stabilization as a neuroprotective strategy for mitigating OP-induced epilepsy and possibly seizure disorders of other etiologies. SIGNIFICANCE STATEMENT: Clinical and preclinical studies support a link between blood-brain barrier (BBB) dysfunction and epileptogenesis; however, a causal relationship has been difficult to prove. Mechanistic studies to delineate relationships between BBB dysfunction and epilepsy may provide novel insights into BBB stabilization as a neuroprotective strategy for mitigating epilepsy resulting from acute organophosphate (OP) intoxication and non-OP causes and potentially other adverse neurological conditions associated with acute OP intoxication, such as cognitive impairment.
Collapse
Affiliation(s)
- Pedro N Bernardino
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, California (P.N.B., A.S.L., P.M.A., C.M.U., P.J.L.); Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California (M.I.G.); and Department of Pharmacology, University of California, Davis, School of Medicine, Davis, California (A.G.)
| | - Audrey S Luo
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, California (P.N.B., A.S.L., P.M.A., C.M.U., P.J.L.); Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California (M.I.G.); and Department of Pharmacology, University of California, Davis, School of Medicine, Davis, California (A.G.)
| | - Peter M Andrew
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, California (P.N.B., A.S.L., P.M.A., C.M.U., P.J.L.); Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California (M.I.G.); and Department of Pharmacology, University of California, Davis, School of Medicine, Davis, California (A.G.)
| | - Chelsea M Unkel
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, California (P.N.B., A.S.L., P.M.A., C.M.U., P.J.L.); Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California (M.I.G.); and Department of Pharmacology, University of California, Davis, School of Medicine, Davis, California (A.G.)
| | - Marco I Gonzalez
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, California (P.N.B., A.S.L., P.M.A., C.M.U., P.J.L.); Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California (M.I.G.); and Department of Pharmacology, University of California, Davis, School of Medicine, Davis, California (A.G.)
| | - Angie Gelli
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, California (P.N.B., A.S.L., P.M.A., C.M.U., P.J.L.); Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California (M.I.G.); and Department of Pharmacology, University of California, Davis, School of Medicine, Davis, California (A.G.)
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, California (P.N.B., A.S.L., P.M.A., C.M.U., P.J.L.); Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California (M.I.G.); and Department of Pharmacology, University of California, Davis, School of Medicine, Davis, California (A.G.)
| |
Collapse
|
3
|
Taboada-Rosell K, Castro-García FA, Medina-Saldivar C, Cruz-Visalaya SR, Pacheco-Otalora LF. The novel FAAH inhibitor, MCH1, reduces the infarction area in the motor cortex-related region but does not affect the sensorimotor function or memory and spatial learning in rats exposed to transient middle cerebral artery occlusion. Brain Res 2024; 1822:148636. [PMID: 37865139 DOI: 10.1016/j.brainres.2023.148636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/23/2023]
Abstract
Macamides, amides of fatty acids first isolated from maca (Lepidium meyenii) are potentially responsible for the reduction of ischemic injury in the stroke animal model followed by maca extract administration. This deduction comes from its ability to inhibit the fatty acid amide hydrolase activity, an enzyme related to the endocannabinoid anandamide hydrolysis. However, no study about the effects of isolated macamides on in-vivo models has been published yet. Our objective was to evaluate the effect of a 10-day 30 mg/kg i.p. MCH1 administration, the macamide with the higher FAAH inhibition capability, on the neurological recovery and brain infarction area of Sprague-Dawley rats exposed to the transient middle cerebral artery occlusion (MCAO) model. Our results showed that the group receiving MCH1 for 10 days did not improve Garcia's neurological score compared to receiving the vehicle only. Likewise, the MCH1 group did not improve their sensorimotor dysfunction as indicated by the latency to detect and remove the tape from the contralateral forepaw in the adhesive removal test, and a similar number of errors with the contralateral forepaw in the foot fault test compared to the vehicle group at the 10th day. Evaluation of the spatial memory and learning using the Barnes test showed longer latency to reach the escape box in the Vehicle and MCH1 groups compared to the control group (no MCAO) only in the retrieval test, while no effect of MCAO procedure or MCH1 administration was observed in the reversal learning test. Despite the lack of behavioral effect of MCH1, analysis of the infarcted areas in the brain using the 2, 3, 5-Triphenyltetrazolium chloride (TTC) staining method in the seven consecutive coronal sections revealed that the infarcted area in the first (bregma + 4.2 mm) and fifth (bregma -3.8 mm) coronal sections of the MCAO + MCH1 group remained similar to the Control group. These results provide evidence that MCH1 can limit damage from ischemic stroke, although it is not reflected in neurological or sensorimotor behavior and spatial learning and memory.
Collapse
Affiliation(s)
- K Taboada-Rosell
- Laboratorio de Investigación en Neurociencia, Instituto Científico, Universidad Andina del Cusco, Cuzco, Peru
| | - F A Castro-García
- Laboratorio de Investigación en Neurociencia, Instituto Científico, Universidad Andina del Cusco, Cuzco, Peru
| | - C Medina-Saldivar
- Laboratorio de Investigación en Neurociencia, Instituto Científico, Universidad Andina del Cusco, Cuzco, Peru
| | - S R Cruz-Visalaya
- Laboratorio de Investigación en Neurociencia, Instituto Científico, Universidad Andina del Cusco, Cuzco, Peru
| | - L F Pacheco-Otalora
- Laboratorio de Investigación en Neurociencia, Instituto Científico, Universidad Andina del Cusco, Cuzco, Peru.
| |
Collapse
|
4
|
Cipriani R, Domerq M, Martín A, Matute C. Role of Microglia in Stroke. ADVANCES IN NEUROBIOLOGY 2024; 37:405-422. [PMID: 39207705 DOI: 10.1007/978-3-031-55529-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Ischemic stroke is a complex brain pathology caused by an interruption of blood supply to the brain. It results in neurological deficits which that reflect the localization and the size of the compromised brain area and are the manifestation of complex pathogenic events triggered by energy depletion. Inflammation plays a prominent role, worsening the injury in the early phase and influencing poststroke recovery in the late phase. Activated microglia are one of the most important cellular components of poststroke inflammation, appearing from the first few hours and persisting for days and weeks after stroke injury. In this chapter, we will discuss the nature of the inflammatory response in brain ischemia, the contribution of microglia to injury and regeneration after stroke, and finally, how ischemic stroke directly affects microglia functions and survival.
Collapse
Affiliation(s)
| | - Maria Domerq
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU) and CIBERNED, Leioa, Spain
| | - Abraham Martín
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Ikerbasque Basque Foundation for Science, Bilbao, Spain.
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU) and CIBERNED, Leioa, Spain.
| |
Collapse
|
5
|
Hu S, Wang X, Yang X, Ouyang S, Pan X, Fu Y, Wu S. Long-term iTBS Improves Neural Functional Recovery by Reducing the Inflammatory Response and Inhibiting Neuronal Apoptosis Via miR-34c-5p/p53/Bax Signaling Pathway in Cerebral Ischemic Rats. Neuroscience 2023; 527:37-51. [PMID: 37468029 DOI: 10.1016/j.neuroscience.2023.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
To investigate intermittent theta-burst stimulation (iTBS) effect on ischemic stroke and the underlying mechanism of neurorehabilitation, we developed an ischemia/reperfusion (I/R) injury model in Sprague-Dawley (SD) rats using the middle cerebral artery occlusion/reperfusion (MCAO/r) method. Next, using different behavioral studies, we compared the improvement of the whole organism with and without iTBS administration for 28 days. We further explored the morphological and molecular biological alterations associated with neuronal apoptosis and neuroinflammation by TTC staining, HE staining, Nissl staining, immunofluorescence staining, ELISA, small RNA sequencing, RT-PCR, and western blot assays. The results showed that iTBS significantly protected against neurological deficits and neurological damage induced by cerebral I/R injury. iTBS also significantly decreased brain infarct volume and increased the number of surviving neurons after 28 days. Additionally, it was observed that iTBS decreased synaptic loss, suppressed activation of astrocytes and M1-polarized microglia, and simultaneously promoted M2-polarized microglial activation. Furthermore, iTBS intervention inhibited neuronal apoptosis and exerted a positive impact on the neuronal microenvironment by reducing neuroinflammation in cerebral I/R injured rats. To further investigate the iTBS mechanism, this study was conducted using small RNA transcriptome sequencing of various groups of peri-infarcted tissues. Bioinformatics analysis and RT-PCR discovered the possible involvement of miR-34c-5p in the mechanism of action. The target genes prediction and detection of dual-luciferase reporter genes confirmed that miR-34c-5p could inhibit neuronal apoptosis in cerebral I/R injured rats by regulating the p53/Bax signaling pathway. We also confirmed by RT-PCR and western blotting that miR-34c-5p inhibited Bax expression. In conclusion, our study supports that iTBS is vital in inhibiting neuronal apoptosis in cerebral I/R injured rats by mediating the miR-34c-5p involvement in regulating the p53/Bax signaling pathway.
Collapse
Affiliation(s)
- Shouxing Hu
- Guizhou Medical University, 9 Beijing Street, Yunyan District, Guiyang, Guizhou, China
| | - Xianbin Wang
- Guizhou Medical University, 9 Beijing Street, Yunyan District, Guiyang, Guizhou, China; Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, China
| | - Xianglian Yang
- Guizhou Medical University, 9 Beijing Street, Yunyan District, Guiyang, Guizhou, China
| | - Shuai Ouyang
- Guizhou Medical University, 9 Beijing Street, Yunyan District, Guiyang, Guizhou, China
| | - Xiao Pan
- Guizhou Medical University, 9 Beijing Street, Yunyan District, Guiyang, Guizhou, China
| | - Yingxue Fu
- Guizhou Medical University, 9 Beijing Street, Yunyan District, Guiyang, Guizhou, China
| | - Shuang Wu
- Guizhou Medical University, 9 Beijing Street, Yunyan District, Guiyang, Guizhou, China; Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, China.
| |
Collapse
|
6
|
Ivanov IA, Siniavin AE, Palikov VA, Senko DA, Shelukhina IV, Epifanova LA, Ojomoko LO, Belukhina SY, Prokopev NA, Landau MA, Palikova YA, Kazakov VA, Borozdina NA, Bervinova AV, Dyachenko IA, Kasheverov IE, Tsetlin VI, Kudryavtsev DS. Analogs of 6-Bromohypaphorine with Increased Agonist Potency for α7 Nicotinic Receptor as Anti-Inflammatory Analgesic Agents. Mar Drugs 2023; 21:368. [PMID: 37367693 DOI: 10.3390/md21060368] [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: 04/20/2023] [Revised: 06/03/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
Hypaphorines, tryptophan derivatives, have anti-inflammatory activity, but their mechanism of action was largely unknown. Marine alkaloid L-6-bromohypaphorine with EC50 of 80 μM acts as an agonist of α7 nicotinic acetylcholine receptor (nAChR) involved in anti-inflammatory regulation. We designed the 6-substituted hypaphorine analogs with increased potency using virtual screening of their binding to the α7 nAChR molecular model. Fourteen designed analogs were synthesized and tested in vitro by calcium fluorescence assay on the α7 nAChR expressed in neuro 2a cells, methoxy ester of D-6-iodohypaphorine (6ID) showing the highest potency (EC50 610 nM), being almost inactive toward α9α10 nAChR. The macrophages cytometry revealed an anti-inflammatory activity, decreasing the expression of TLR4 and increasing CD86, similarly to the action of PNU282987, a selective α7 nAChR agonist. 6ID administration in doses 0.1 and 0.5 mg/kg decreased carrageenan-induced allodynia and hyperalgesia in rodents, in accord with its anti-inflammatory action. Methoxy ester of D-6-nitrohypaphorine demonstrated anti-oedemic and analgesic effects in arthritis rat model at i.p. doses 0.05-0.26 mg/kg. Tested compounds showed excellent tolerability with no acute in vivo toxicity in dosages up to 100 mg/kg i.p. Thus, combining molecular modelling and natural product-inspired drug design improved the desired activity of the chosen nAChR ligand.
Collapse
Affiliation(s)
- Igor A Ivanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Andrei E Siniavin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ivanovsky Institute of Virology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Victor A Palikov
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Dmitry A Senko
- Center Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Irina V Shelukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Lyubov A Epifanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Lucy O Ojomoko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Svetlana Y Belukhina
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Nikita A Prokopev
- Department of Biology, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Mariia A Landau
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Yulia A Palikova
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Vitaly A Kazakov
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Natalia A Borozdina
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Arina V Bervinova
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Igor A Dyachenko
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Igor E Kasheverov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Victor I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Denis S Kudryavtsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
7
|
Zhang Y, Jia P, Wang K, Zhang Y, Lv Y, Fan P, Yang L, Zhang S, Wang T, Zhao J, Lv H, Chen X, Liu Y, Wei H, Zhang P. Lactate modulates microglial inflammatory responses after oxygen-glucose deprivation through HIF-1α-mediated inhibition of NF-κB. Brain Res Bull 2023; 195:1-13. [PMID: 36746287 DOI: 10.1016/j.brainresbull.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
Metabolic adaption drives microglial inflammatory responses, and lactate shapes immunological and inflammatory states. However, whether lactate was involved in the regulation of microglial inflammatory responses after cerebral ischemia remains unclear. In this study, the expression of iNOS, arginase-1, phosphorylated NF-κB p65 and IκB-α, and HIF-1α in BV2 cells after oxygen-glucose deprivation (OGD) were detected by western blotting and immunofluorescence. The mRNA levels of microglial responsive markers and inflammatory factors were assessed by real-time-qPCR. The effect of lactate-treated BV2 cells on the survival of primary neurons was observed using transwell co-culture. The results showed that the protein levels of iNOS and arginase-1, the ratio of mRNA levels of iNOS/CD206, CD86/Ym1, IL-6/IL-10, TNF-α/IL-10 and the mRNA levels of IL-6 and TNF-α, as well as the protein levels of phosphorylated NF-κB p65 and IκB-α, were increased after OGD. Lactate treatment inhibited the OGD-induced increase in the protein levels of iNOS, phosphorylated NF-κB p65 and IκB-α, as well as iNOS/CD206, CD86/Ym1, IL-6/IL-10, TNF-α/IL-10, IL-6 and TNF-α mRNA levels in BV2 cells, while promoted arginase-1 protein expression as well as IL-10 and TGF-β mRNA level. Interestingly, lactate activated HIF-1α and the HIF-1α inhibitor YC-1 reversed the effect of lactate on levels of microglial responsive markers and phosphorylated NF-κB p65 and IκB-α in BV2 cells. Moreover, knockdown of HIF-1α by lentivirus-delivered shRNA also reversed the effect of lactate on phosphorylated NF-κB p65 and IκB-α in BV2 cells after OGD. Finally, and importantly, lactate-treated BV2 microglia increased the viability and decreased the apoptosis of neurons after OGD. These findings revealed that lactate inhibited NF-κB pathway and skewed BV2 microglia toward the protective response through activation of HIF-1α after OGD, thereby improving neuronal survival.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Pengyu Jia
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yuying Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Pei Fan
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Liufei Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shuyue Zhang
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tianyue Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jing Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Haixia Lv
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an 710061, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
8
|
Aguado L, Joya A, Garbizu M, Plaza-García S, Iglesias L, Hernández MI, Ardaya M, Mocha N, Gómez-Vallejo V, Cossio U, Higuchi M, Rodríguez-Antigüedad A, Freijo MM, Domercq M, Matute C, Ramos-Cabrer P, Llop J, Martín A. Therapeutic effect of α7 nicotinic receptor activation after ischemic stroke in rats. J Cereb Blood Flow Metab 2023:271678X231161207. [PMID: 36916034 PMCID: PMC10369150 DOI: 10.1177/0271678x231161207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
Nicotinic acetylcholine α7 receptors (α7 nAChRs) have a well-known modulator effect in neuroinflammation. Yet, the therapeutical effect of α7 nAChRs activation after stroke has been scarcely evaluated to date. The role of α7 nAChRs activation with PHA 568487 on inflammation after brain ischemia was assessed with positron emission tomography (PET) using [18F]DPA-714 and [18F]BR-351 radiotracers after transient middle cerebral artery occlusion (MCAO) in rats. The assessment of brain oedema, blood brain barrier (BBB) disruption and neurofunctional progression after treatment was evaluated with T2 weighted and dynamic contrast-enhanced magnetic resonance imaging (T2 W and DCE-MRI) and neurological evaluation. The activation of α7 nAChRs resulted in a decrease of ischemic lesion, midline displacement and cell neurodegeneration from days 3 to 7 after ischemia. Besides, the treatment with PHA 568487 improved the neurofunctional outcome. Treated ischemic rats showed a significant [18F]DPA-714-PET uptake reduction at day 7 together with a decrease of activated microglia/infiltrated macrophages. Likewise, the activation of α7 receptors displayed an increase of [18F]BR-351-PET signal in ischemic cortical regions, which resulted from the overactivation of MMP-2. Finally, the treatment with PHA 568487 showed a protective effect on BBB disruption and blood brain vessel integrity after cerebral ischemia.
Collapse
Affiliation(s)
- Laura Aguado
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Ana Joya
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | | | - Sandra Plaza-García
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Leyre Iglesias
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Neurovascular Group, Biocruces Health Research Institute, Barakaldo, Spain
| | | | - María Ardaya
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Donostia International Physics Center (DIPC), San Sebastian, Spain
| | - Naroa Mocha
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | | | - Unai Cossio
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Makoto Higuchi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | | | - Mari Mar Freijo
- Neurovascular Group, Biocruces Health Research Institute, Barakaldo, Spain.,Department of Neurology, Cruces University Hospital, Barakaldo, Spain
| | - María Domercq
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of Basque Country (UPV/EHU) and CIBERNED, Leioa, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of Basque Country (UPV/EHU) and CIBERNED, Leioa, Spain
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain.,Centro de Investigación Biomédica en Red - Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - Abraham Martín
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
9
|
Yu CC, Liu LB, Chen SY, Wang XF, Wang L, Du YJ. Ancient Chinese Herbal Recipe Huanglian Jie Du Decoction for Ischemic Stroke: An Overview of Current Evidence. Aging Dis 2022; 13:1733-1744. [PMID: 36465168 PMCID: PMC9662271 DOI: 10.14336/ad.2022.0311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/11/2022] [Indexed: 09/30/2023] Open
Abstract
Ischemic stroke is a major cause of mortality and neurological morbidity worldwide. The underlying pathophysiology of ischemic stroke is highly complicated and correlates with various pathological processes, including neuroinflammation, oxidative stress injury, altered cell apoptosis and autophagy, excitotoxicity, and acidosis. The current treatment for ischemic stroke is limited to thrombolytic therapy such as recombinant tissue plasminogen activator. However, tissue plasminogen activator is limited by a very narrow therapeutic time window (<4.5 hours), selective efficacy, and hemorrhagic complication. Hence, the development of novel therapies to prevent ischemic damage to the brain is urgent. Chinese herbal medicine has a long history in treating stroke and its sequela. In the past decades, extensive studies have focused on the neuroprotective effects of Huanglian Jie Du decoction (HLJDD), an ancient and classical Chinese herbal formula that can treat a wide spectrum of disorders including ischemic stroke. In this review, the current evidence of HLJDD and its bioactive components for ischemic stroke is comprehensively reviewed, and their potential application directions in ischemic stroke management are discussed.
Collapse
Affiliation(s)
- Chao-Chao Yu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
| | - Le-Bin Liu
- Department of Rehabilitation Medicine, Hubei Rongjun Hospital, Wuhan, Hubei, China.
| | - Shi-Yuan Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Xiao-Fei Wang
- Department of Rehabilitation Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Li Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Wei Z, Xie Y, Wei M, Zhao H, Ren K, Feng Q, Xu Y. New insights in ferroptosis: Potential therapeutic targets for the treatment of ischemic stroke. Front Pharmacol 2022; 13:1020918. [PMID: 36425577 PMCID: PMC9679292 DOI: 10.3389/fphar.2022.1020918] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/26/2022] [Indexed: 10/22/2023] Open
Abstract
Stroke is a common disease in clinical practice, which seriously endangers people's physical and mental health. The neurovascular unit (NVU) plays a key role in the occurrence and development of ischemic stroke. Different from other classical types of cell death such as apoptosis, necrosis, autophagy, and pyroptosis, ferroptosis is an iron-dependent lipid peroxidation-driven new form of cell death. Interestingly, the function of NVU and stroke development can be regulated by activating or inhibiting ferroptosis. This review systematically describes the NVU in ischemic stroke, provides a comprehensive overview of the regulatory mechanisms and key regulators of ferroptosis, and uncovers the role of ferroptosis in the NVU and the progression of ischemic stroke. We further discuss the latest progress in the intervention of ferroptosis as a therapeutic target for ischemic stroke and summarize the research progress and regulatory mechanism of ferroptosis inhibitors on stroke. In conclusion, ferroptosis, as a new form of cell death, plays a key role in ischemic stroke and is expected to become a new therapeutic target for this disease.
Collapse
Affiliation(s)
- Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingze Wei
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Xu XJ, Yang MS, Zhang B, Ge QQ, Niu F, Dong JQ, Zhuang Y, Liu BY. Genome-wide interrogation of transfer RNA-derived small RNAs in a mouse model of traumatic brain injury. Neural Regen Res 2022; 17:386-394. [PMID: 34269214 PMCID: PMC8463968 DOI: 10.4103/1673-5374.314315] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transfer RNA (tRNA)-derived small RNAs (tsRNAs) are a recently established family of regulatory small non-coding RNAs that modulate diverse biological processes. Growing evidence indicates that tsRNAs are involved in neurological disorders and play a role in the pathogenesis of neurodegenerative disease. However, whether tsRNAs are involved in traumatic brain injury-induced secondary injury remains poorly understood. In this study, a mouse controlled cortical impact model of traumatic brain injury was established, and integrated tsRNA and messenger RNA (mRNA) transcriptome sequencing were used. The results revealed that 103 tsRNAs were differentially expressed in the mouse model of traumatic brain injury at 72 hours, of which 56 tsRNAs were upregulated and 47 tsRNAs were downregulated. Based on microRNA-like seed matching and Pearson correlation analysis, 57 differentially expressed tsRNA-mRNA interaction pairs were identified, including 29 tsRNAs and 26 mRNAs. Moreover, Gene Ontology annotation of target genes revealed that the significantly enriched terms were primarily associated with inflammation and synaptic function. Collectively, our findings suggest that tsRNAs may be associated with traumatic brain injury-induced secondary brain injury, and are thus a potential therapeutic target for traumatic brain injury. The study was approved by the Beijing Neurosurgical Institute Animal Care and Use Committee (approval No. 20190411) on April 11, 2019.
Collapse
Affiliation(s)
- Xiao-Jian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Meng-Shi Yang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qian-Qian Ge
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jin-Qian Dong
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhuang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bai-Yun Liu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University; Nerve Injury and Repair Center of Beijing Institute for Brain Disorders; China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
12
|
Winek K, Soreq H, Meisel A. Regulators of cholinergic signaling in disorders of the central nervous system. J Neurochem 2021; 158:1425-1438. [PMID: 33638173 PMCID: PMC8518971 DOI: 10.1111/jnc.15332] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/23/2021] [Accepted: 02/21/2021] [Indexed: 12/14/2022]
Abstract
Cholinergic signaling is crucial in cognitive processes, and degenerating cholinergic projections are a pathological hallmark in dementia. Use of cholinesterase inhibitors is currently the main treatment option to alleviate symptoms of Alzheimer's disease and has been postulated as a therapeutic strategy in acute brain damage (stroke and traumatic brain injury). However, the benefits of this treatment are still not clear. Importantly, cholinergic receptors are expressed both by neurons and by astrocytes and microglia, and binding of acetylcholine to the α7 nicotinic receptor in glial cells results in anti-inflammatory response. Similarly, the brain fine-tunes the peripheral immune response over the cholinergic anti-inflammatory axis. All of these processes are of importance for the outcome of acute and chronic neurological disease. Here, we summarize the main findings about the role of cholinergic signaling in brain disorders and provide insights into the complexity of molecular regulators of cholinergic responses, such as microRNAs and transfer RNA fragments, both of which may fine-tune the orchestra of cholinergic mRNAs. The available data suggest that these small noncoding RNA regulators may include promising biomarkers for predicting disease course and assessing treatment responses and might also serve as drug targets to attenuate signaling cascades during overwhelming inflammation and to ameliorate regenerative capacities of neuroinflammation.
Collapse
Affiliation(s)
- Katarzyna Winek
- The Edmond and Lily Safra Center for Brain SciencesThe Hebrew University of JerusalemJerusalemIsrael
- The Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalemIsrael
| | - Hermona Soreq
- The Edmond and Lily Safra Center for Brain SciencesThe Hebrew University of JerusalemJerusalemIsrael
- The Alexander Silberman Institute of Life SciencesThe Hebrew University of JerusalemJerusalemIsrael
| | - Andreas Meisel
- Department of Neurology with Experimental NeurologyCenter for Stroke Research BerlinNeuroCure Clinical Research CenterCharité‐Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
13
|
Wang JH, Huang J, Guo FQ, Wang F, Yang S, Yu NW, Zheng B, Wang J. Circulating Neurofilament Light Predicts Cognitive Decline in Patients With Post-stroke Subjective Cognitive Impairment. Front Aging Neurosci 2021; 13:665981. [PMID: 34079450 PMCID: PMC8165181 DOI: 10.3389/fnagi.2021.665981] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/01/2021] [Indexed: 02/05/2023] Open
Abstract
Background Subjective cognitive impairment (SCI) is common after acute ischemic stroke and adversely affects the quality of life. SCI is associated with an increased risk of developing mild cognitive impairment and dementia. Identifying biomarkers which could predict long-term cognitive outcomes of post-stroke SCI is of importance for early intervention. This study aims to investigate the association between circulating neurofilament light (NfL) and long-term cognitive function in patients with post-stroke SCI. Methods This longitudinal study recruited 304 patients with post-stroke SCI, and serum NfL levels were determined at baseline. These patients were followed up for 12 months for the observation of cognitive change. Cognitive performances were assessed by a Chinese version of the Telephone Interview of Cognitive Status-40 (TICS-40) scale. Results The patients were divided into a progression group (as determined by decreased TICS-40 scores) and a stable group (as determined by increased or unchanged TICS-40 scores). The progression group had significantly higher serum NfL levels than the stable group at baseline. Serum NfL levels were predictive for longitudinal cognitive decline during follow-up. Conclusion These findings imply that circulating NfL could predict the long-term cognitive change of patients with post-stroke SCI.
Collapse
Affiliation(s)
- Jian-Hong Wang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Jie Huang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Fu-Qiang Guo
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Fang Wang
- Innovation Center of Nursing Research, West China Hospital, Nursing Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Shu Yang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Neng-Wei Yu
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Bo Zheng
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
| | - Jian Wang
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
| |
Collapse
|
14
|
Purinergic signaling in nervous system health and disease: Focus on pannexin 1. Pharmacol Ther 2021; 225:107840. [PMID: 33753132 DOI: 10.1016/j.pharmthera.2021.107840] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
Purinergic signaling encompasses the cycle of adenosine 5' triphosphate (ATP) release and its metabolism into nucleotide and nucleoside derivatives, the direct release of nucleosides, and subsequent receptor-triggered downstream intracellular pathways. Since the discovery of nerve terminal and glial ATP release into the neuropil, purinergic signaling has been implicated in the modulation of nervous system development, function, and disease. In this review, we detail our current understanding of the roles of the pannexin 1 (PANX1) ATP-release channel in neuronal development and plasticity, glial signaling, and neuron-glial-immune interactions. We additionally provide an overview of PANX1 structure, activation, and permeability to orientate readers and highlight recent research developments. We identify areas of convergence between PANX1 and purinergic receptor actions. Additional highlights include data on PANX1's participation in the pathophysiology of nervous system developmental, degenerative, and inflammatory disorders. Our aim in combining this knowledge is to facilitate the movement of our current understanding of PANX1 in the context of other nervous system purinergic signaling mechanisms one step closer to clinical translation.
Collapse
|
15
|
Cytochrome P450 CYP2E1 Suppression Ameliorates Cerebral Ischemia Reperfusion Injury. Antioxidants (Basel) 2021; 10:antiox10010052. [PMID: 33466250 PMCID: PMC7824747 DOI: 10.3390/antiox10010052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022] Open
Abstract
Despite existing strong evidence on oxidative markers overproduction following ischemia/reperfusion (I/R), the mechanism by which oxidative enzyme Cytochrome P450-2E1 (CYP2E1) contributes to I/R outcomes is not clear. In this study, we sought to evaluate the functional significance of CYP2E1 in I/R. CYP2E1 KO mice and controls were subjected to middle cerebral artery occlusion (MCAo-90 min) followed by 24 h of reperfusion to induce focal I/R injury as an acute stage model. Then, histological and chemical analyses were conducted to investigate the role of CYP2E1 in lesion volume, oxidative stress, and inflammation exacerbation. Furthermore, the role of CYP2E1 on the blood-brain barrier (BBB) integrity was investigated by measuring 20-hydroxyecosatetraenoic acid (20-HETE) activity, as well as, in vivo BBB transfer rate. Following I/R, the CYP2E1 KO mice exhibited a significantly lower lesion volume, and neurological deficits compared to controls (p < 0.005). Moreover, reactive oxygen species (ROS) production, apoptosis, and neurodegeneration were significantly lower in the CYP2E1(−/−) I/R group (p < 0.001). The BBB damage was significantly lower in CYP2E1(−/−) mice compared to wild-type (WT) (p < 0.001), while 20-HETE production was increased by 41%. Besides, inflammatory cytokines expression and the number of activated microglia were significantly lower in CYP2E1(−/−) mice following I/R. CYP2E1 suppression ameliorates I/R injury and protects BBB integrity by reducing both oxidative stress and inflammation.
Collapse
|
16
|
Deletion of muscarinic acetylcholine receptor 3 in microglia impacts brain ischemic injury. Brain Behav Immun 2021; 91:89-104. [PMID: 32927021 DOI: 10.1016/j.bbi.2020.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 01/17/2023] Open
Abstract
Microglia are the immune cells of the brain and become activated during any type of brain injury. In the middle cerebral artery occlusion (MCAo) model, a mouse model for ischemic stroke, we have previously shown that microglia and invaded monocytes upregulate the expression of the muscarinic acetylcholine receptor 3 (M3R) in the ischemic lesion. Here we tested whether this upregulation has an impact on the pathogenesis of MCAo. We depleted the m3R receptor in microglia, but not in circulating monocytes by giving tamoxifen to CX3CR1-CreERT+/+M3Rflox/flox (M3RKOmi) animals 3 weeks prior to MCAo. We found that M3RKOmi male mice had bigger lesions, more pronounced motor deficits after one week and cognitive deficits after about one month compared to control males. The density of Iba1+ cells was lower in the lesions of M3RKO male mice in the early, but not in the late disease phase. In females, these differences were not significant. By giving tamoxifen 1 week prior to MCAo, we depleted m3R in microglia and in circulating monocytes (M3RKOmi/mo). Male M3RKOmi/mo did not differ in lesion size, but had a lower survival rate, showed motor deficits and a reduced accumulation of Iba1+ positive cells into the lesion site. In conclusion, our data suggest that the upregulation of m3R in microglia and monocytes in stroke has a beneficial effect on the clinical outcome in male mice.
Collapse
|
17
|
Joya A, Ardaya M, Montilla A, Garbizu M, Plaza-García S, Gómez-Vallejo V, Padro D, Gutiérrez JJ, Rios X, Ramos-Cabrer P, Cossío U, Pulagam KR, Higuchi M, Domercq M, Cavaliere F, Matute C, Llop J, Martín A. In vivo multimodal imaging of adenosine A 1 receptors in neuroinflammation after experimental stroke. Theranostics 2021; 11:410-425. [PMID: 33391483 PMCID: PMC7681082 DOI: 10.7150/thno.51046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/24/2020] [Indexed: 01/21/2023] Open
Abstract
Adenosine A1 receptors (A1ARs) are promising imaging biomarkers and targets for the treatment of stroke. Nevertheless, the role of A1ARs on ischemic damage and its subsequent neuroinflammatory response has been scarcely explored so far. Methods: In this study, the expression of A1ARs after transient middle cerebral artery occlusion (MCAO) was evaluated by positron emission tomography (PET) with [18F]CPFPX and immunohistochemistry (IHC). In addition, the role of A1ARs on stroke inflammation using pharmacological modulation was assessed with magnetic resonance imaging (MRI), PET imaging with [18F]DPA-714 (TSPO) and [18F]FLT (cellular proliferation), as well as IHC and neurofunctional studies. Results: In the ischemic territory, [18F]CPFPX signal and IHC showed the overexpression of A1ARs in microglia and infiltrated leukocytes after cerebral ischemia. Ischemic rats treated with the A1AR agonist ENBA showed a significant decrease in both [18F]DPA-714 and [18F]FLT signal intensities at day 7 after cerebral ischemia, a feature that was confirmed by IHC results. Besides, the activation of A1ARs promoted the reduction of the brain lesion, as measured with T2W-MRI, and the improvement of neurological outcome including motor, sensory and reflex responses. These results show for the first time the in vivo PET imaging of A1ARs expression after cerebral ischemia in rats and the application of [18F]FLT to evaluate glial proliferation in response to treatment. Conclusion: Notably, these data provide evidence for A1ARs playing a key role in the control of both the activation of resident glia and the de novo proliferation of microglia and macrophages after experimental stroke in rats.
Collapse
|
18
|
Yuan Y, Shan X, Men W, Zhai H, Qiao X, Geng L, Li C. The effect of crocin on memory, hippocampal acetylcholine level, and apoptosis in a rat model of cerebral ischemia. Biomed Pharmacother 2020; 130:110543. [PMID: 32738637 DOI: 10.1016/j.biopha.2020.110543] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/05/2020] [Accepted: 07/20/2020] [Indexed: 11/17/2022] Open
Abstract
Although the memory- improving effect of crocin has been suggested by previous evidences, the association between this effect and hippocampal acetylcholine (Ach) level and apoptosis is not well investigated. This study aimed to determine the protective effects of crocin on memory, hippocampal Ach level, and apoptosis in a rat model of cerebral ischemia. Male Wistar rats were divided into sham group received saline, and other 3 groups underwent 4-vessel occlusion brain ischemia (4VOI), received oral administration of either saline or crocin in doses of 30 mg/day and 60 mg/day for 7 days. Outcomes were memory, determined by radial eight-arm maze (RAM) task and Morris water maze (MWM) test, Ach release in the dorsal hippocampus (evaluated by microdialysis-HPLC) and apoptosis (investigated by TUNEL assay). 4VOI impaired memory reduced dorsal hippocampus Ach level, and induced apoptosis. Crocin, significantly improved the memory (F = 343.20; P < 0.001 for RAM error choices and F = 182.5; P < 0.0001 for MWM), increased Ach level (F = 115.1; P < 0.001) and prevented hippocampal neuronal apoptosis (W = 183.50; P < 0.001) as compared statistically by ANOVA test. Crocin can be suggested as a promising therapy for ischemic cerebrovascular accidents by its memory preserving, Ach-increasing, and neuroprotective effects.
Collapse
Affiliation(s)
- Yu Yuan
- Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, China
| | - Xiaosong Shan
- Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, China
| | - Weidong Men
- Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, China
| | - Hexin Zhai
- Emergency Department, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, China
| | - Xiaoxia Qiao
- Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, China
| | - Lianting Geng
- Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, China
| | - Chunhui Li
- Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, China.
| |
Collapse
|
19
|
Abstract
Stroke remains a major unmet clinical need that warrants novel therapies. Following an ischemic insult, the cerebral vasculature secretes inflammatory molecules, creating the stroke vasculome profile. The present study evaluated the therapeutic effects of endothelial cells on the inflammation-associated stroke vasculome. qRT-PCR analysis revealed that specific inflammation-related vasculome genes BRM, IκB, Foxf1, and ITIH-5 significantly upregulated by oxygen glucose deprivation (OGD. Interestingly, co-culture of human endothelial cells (HEN6) with human endothelial cells (EPCs) during OGD significantly blocked the elevations of BRM, IκB, and Foxf1, but not ITIH-5. Next, employing the knockdown/antisense technology, silencing the inflammation-associated stroke vasculome gene, IκB, as opposed to scrambled knockdown, blocked the EPC-mediated protection of HEN6 against OGD. In vivo, stroke animals transplanted with intracerebral human EPCs (300,000 cells) into the striatum and cortex 4 h post ischemic stroke displayed significant behavioral recovery up to 30 days post-transplantation compared to vehicle-treated stroke animals. At 7 days post-transplantation, quantification of the fluorescent staining intensity in the cortex and striatum revealed significant upregulation of the endothelial marker RECA1 and a downregulation of the stroke-associated vasculome BRM, IKB, Foxf1, ITIH-5 and PMCA2 in the ipsilateral side of cortex and striatum of EPC-transplanted stroke animals relative to vehicle-treated stroke animals. Altogether, these results demonstrate that EPCs exert therapeutic effects in experimental stroke possibly by modulating the inflammation-plagued vasculome.
Collapse
|
20
|
Kim DY, Zhang H, Park S, Kim Y, Bae CR, Kim YM, Kwon YG. CU06-1004 (endothelial dysfunction blocker) ameliorates astrocyte end-feet swelling by stabilizing endothelial cell junctions in cerebral ischemia/reperfusion injury. J Mol Med (Berl) 2020; 98:875-886. [PMID: 32415357 PMCID: PMC7297708 DOI: 10.1007/s00109-020-01920-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/27/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023]
Abstract
Cerebral ischemia, or stroke, is widespread leading cause of death and disability. Surgical and pharmacological interventions that recover blood flow are the most effective treatment strategies for stroke patients. However, restoring the blood supply is accompanied by severe reperfusion injury, with edema and astrocyte end-feet disruption. Here, we report that the oral administration of CU06-1004 (previously Sac-1004), immediately after onset of ischemia/reperfusion (I/R), ameliorated cerebral damage. CU06-1004 stabilized blood‑brain barrier by inhibiting the disruption of the tight junction-related protein zona occludens-1 and the cortical actin ring in endothelial cells (ECs) after I/R. Interestingly, CU06-1004 significantly suppressed astrocyte end-feet swelling following I/R, by reducing aquaporin 4 and connexin 43 levels, which mediates swelling. Furthermore, the degradation of β1-integrin and β-dystroglycan, which anchors to the cortical actin ring in ECs, was inhibited by CU06-1004 administration after I/R. Consistently, CU06-1004 administration following I/R also suppressed the loss of laminin and collagen type IV, which bind to the cortical actin ring anchoring proteins. Unlike the protective effects of CU06-1004 in ECs, astrocyte viability and proliferation were not directly affected. Taken together, our observations suggest that CU06-1004 inhibits I/R-induced cerebral edema and astrocyte end-feet swelling by maintaining EC junction stability. KEY MESSAGES: • CU06-1004 ameliorates I/R-induced cerebral injury. • EC junction integrity was stabilized by CU06-1004 treatment after I/R. • CU06-1004 reduces astrocyte end-feet swelling following I/R. • EC junction stability affects astrocyte end-feet structure maintenance after I/R.
Collapse
Affiliation(s)
- Dong Young Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Haiying Zhang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- CURACLE Co., Ltd, Gyeonggi-do, Seongnam-si, Republic of Korea
| | - Songyi Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yeaji Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Cho-Rong Bae
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Gangwon-do, Chuncheon-si, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
21
|
Bao SC, Khan A, Song R, Kai-yu Tong R. Rewiring the Lesioned Brain: Electrical Stimulation for Post-Stroke Motor Restoration. J Stroke 2020; 22:47-63. [PMID: 32027791 PMCID: PMC7005350 DOI: 10.5853/jos.2019.03027] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 02/06/2023] Open
Abstract
Electrical stimulation has been extensively applied in post-stroke motor restoration, but its treatment mechanisms are not fully understood. Stimulation of neuromotor control system at multiple levels manipulates the corresponding neuronal circuits and results in neuroplasticity changes of stroke survivors. This rewires the lesioned brain and advances functional improvement. This review addresses the therapeutic mechanisms of different stimulation modalities, such as noninvasive brain stimulation, peripheral electrical stimulation, and other emerging techniques. The existing applications, the latest progress, and future directions are discussed. The use of electrical stimulation to facilitate post-stroke motor recovery presents great opportunities in terms of targeted intervention and easy applicability. Further technical improvements and clinical studies are required to reveal the neuromodulatory mechanisms and to enhance rehabilitation therapy efficiency in stroke survivors and people with other movement disorders.
Collapse
Affiliation(s)
- Shi-chun Bao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Ahsan Khan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Rong Song
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, China
| | - Raymond Kai-yu Tong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
22
|
Barrera-Sandoval AM, Osorio E, Cardona-Gómez GP. Microglial-targeting induced by intranasal linalool during neurological protection postischemia. Eur J Pharmacol 2019; 857:172420. [DOI: 10.1016/j.ejphar.2019.172420] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022]
|
23
|
|
24
|
Khasanova LT, Stakhovskaya LV, Koltsova EA, Shamalov NA. [Genetic characteristics of stroke]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:65-72. [PMID: 32207720 DOI: 10.17116/jnevro201911912265] [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: 11/17/2022]
Abstract
In the recent years there is a growing interest in identification of additional genetic factors of stroke. A growing body of evidence supports the role of genetic factors in determining the risk of both hemorrhagic and ischemic stroke. The article considers the main genes associated with susceptibility to stroke and genetic polymorphisms associated with the disease. Genetic factors, modulating inflammation process, coagulation, lipid metabolism, NO formation, renin-angiotensin-aldosterone system and homeostasis play a significant role in stroke development. A comprehensive analysis of different genes associated with stroke may help to detect individuals with extremely high risk of stroke and implement timely preventive measures to decrease stroke burden.
Collapse
Affiliation(s)
- L T Khasanova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - L V Stakhovskaya
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - E A Koltsova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - N A Shamalov
- Federal Center for Cerebrovascular Pathology and Stroke, Moscow, Russia
| |
Collapse
|
25
|
Lurie DI. An Integrative Approach to Neuroinflammation in Psychiatric disorders and Neuropathic Pain. J Exp Neurosci 2018; 12:1179069518793639. [PMID: 30127639 PMCID: PMC6090491 DOI: 10.1177/1179069518793639] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is a complex process involving both the peripheral circulation
and the Central Nervous System (CNS) and is considered to underlie many CNS
disorders including depression, anxiety, schizophrenia, and pain. Stressors
including early-life adversity, psychosocial stress, and infection appear to
prime microglia toward a pro-inflammatory phenotype. Subsequent inflammatory
challenges then drive an exaggerated neuroinflammatory response involving the
upregulation of pro-inflammatory mediators that is associated with CNS
dysfunction. Several pharmacologic inhibitors of pro-inflammatory cytokines
including TNF-α and IL-1β show good clinical efficacy in terms of ameliorating
neuroinflammatory processes. Mind/body and plant-based interventions such as
yoga, breathing exercises, meditation, and herbs/spices have also been
demonstrated to reduce pro-inflammatory cytokines and have a positive impact on
depression, anxiety, cognition, and pain. As the intricate connections between
the immune system and the nervous system continue to be elucidated, successful
therapies for reducing neuroinflammation will likely involve an integrated
approach combining drug therapy with nonpharmacologic interventions.
Collapse
Affiliation(s)
- Diana I Lurie
- Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, College of Health Professions & Biomedical Sciences, The University of Montana, Missoula, MT, USA
| |
Collapse
|