Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Neurology ; 101(4): e455-e458, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37487758

ABSTRACT

While it was previously believed that neuromyelitis optic spectrum disorder (NMOSD) mostly affected the optic nerves and the spinal cord, it is increasingly recognized that NMOSD can involve any area of the CNS where aquaporin-4 is highly expressed. These other areas can include the hypothalamus and the circumventricular organs that surround the third and fourth ventricles, serving as osmoregulators. The syndrome of inappropriate antidiuretic hormone secretion (SIADH) is one of the most common causes of hyponatremia and has been associated with NMOSD due to these lesions. In this report, we present a case of a patient with known NMOSD, who presented with dizziness, fatigue, and generalized weakness and whose workup revealed hyponatremia in the setting of SIADH and hypothalamic demyelinating lesions. This case illustrates an atypical presentation of NMOSD and the importance of looking for syndromes, such as SIADH. This can guide diagnostic testing, such as getting thin MRI cuts through the hypothalamus and brainstem, as well as advanced management techniques such as immunotherapy.


Subject(s)
Hyponatremia , Inappropriate ADH Syndrome , Neuroinflammatory Diseases , Neuromyelitis Optica , Adult , Female , Humans , Dizziness/complications , Fatigue/complications , Hyponatremia/complications , Hyponatremia/diagnosis , Hyponatremia/therapy , Hypothalamus/pathology , Inappropriate ADH Syndrome/complications , Inappropriate ADH Syndrome/diagnosis , Inappropriate ADH Syndrome/therapy , Magnetic Resonance Imaging , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/pathology , Neuromyelitis Optica/complications , Neuromyelitis Optica/pathology , Immunotherapy
2.
Pharmacol Res ; 175: 105989, 2022 01.
Article in English | MEDLINE | ID: mdl-34800628

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.


Subject(s)
Brain Ischemia/drug therapy , Caffeic Acids/therapeutic use , Cognitive Dysfunction/drug therapy , Lactates/therapeutic use , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Caffeic Acids/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Chronic Disease , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Crystallins/metabolism , Glucose/metabolism , Humans , Lactates/pharmacology , Male , Microtubule-Associated Proteins/metabolism , NF-kappa B/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroprotective Agents/pharmacology , Rats, Wistar , Receptors, Dopamine D2/metabolism
3.
J Neuroinflammation ; 18(1): 256, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34740378

ABSTRACT

BACKGROUND: Neurotoxic microglia and astrocytes begin to activate and participate in pathological processes after spinal cord injury (SCI), subsequently causing severe secondary damage and affecting tissue repair. We have previously reported that photobiomodulation (PBM) can promote functional recovery by reducing neuroinflammation after SCI, but little is known about the underlying mechanism. Therefore, we aimed to investigate whether PBM ameliorates neuroinflammation by modulating the activation of microglia and astrocytes after SCI. METHODS: Male Sprague-Dawley rats were randomly divided into three groups: a sham control group, an SCI + vehicle group and an SCI + PBM group. PBM was performed for two consecutive weeks after clip-compression SCI models were established. The activation of neurotoxic microglia and astrocytes, the level of tissue apoptosis, the number of motor neurons and the recovery of motor function were evaluated at different days post-injury (1, 3, 7, 14, and 28 days post-injury, dpi). Lipocalin 2 (Lcn2) and Janus kinase-2 (JAK2)-signal transducer and activator of transcription-3 (STAT3) signaling were regarded as potential targets by which PBM affected neurotoxic microglia and astrocytes. In in vitro experiments, primary microglia and astrocytes were irradiated with PBM and cotreated with cucurbitacin I (a JAK2-STAT3 pathway inhibitor), an adenovirus (shRNA-Lcn2) and recombinant Lcn2 protein. RESULTS: PBM promoted the recovery of motor function, inhibited the activation of neurotoxic microglia and astrocytes, alleviated neuroinflammation and tissue apoptosis, and increased the number of neurons retained after SCI. The upregulation of Lcn2 and the activation of the JAK2-STAT3 pathway after SCI were suppressed by PBM. In vitro experiments also showed that Lcn2 and JAK2-STAT3 were mutually promoted and that PBM interfered with this interaction, inhibiting the activation of microglia and astrocytes. CONCLUSION: Lcn2/JAK2-STAT3 crosstalk is involved in the activation of neurotoxic microglia and astrocytes after SCI, and this process can be suppressed by PBM.


Subject(s)
Astrocytes/radiation effects , Low-Level Light Therapy , Microglia/radiation effects , Recovery of Function/radiation effects , Spinal Cord Injuries/pathology , Animals , Astrocytes/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/radiation effects , Lipocalin-2/metabolism , Lipocalin-2/radiation effects , Male , Microglia/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/radiation effects , Signal Transduction/radiation effects , Spinal Cord Injuries/metabolism , Up-Regulation
4.
J Cell Mol Med ; 25(22): 10534-10542, 2021 11.
Article in English | MEDLINE | ID: mdl-34632701

ABSTRACT

The main mechanism of pyroptosis is Caspase-1-mediated GSDMD cleavage, and GSDMD is also the executive protein of pyroptosis. Our previous study has shown that mafenide can inhibit pyroptosis by inhibiting the GSDMD-Asp275 site to suppress cleavage. In this study, sulfonamide was used as the parent nucleus structure to synthesize sulfa-4 and sulfa-20. Screening of drug activity in the pyroptosis model of BV2 and iBMDM cell lines revealed the efficacy of five compounds were superior to mafenide, which exerted a better inhibitory effect on the occurrence of pyroptosis. For in vivo assay, Sulfa-4 and Sulfa-22 were intervened in the neuroinflammation APP/PS1 mice. As a result, the administration of Sulfa-4 and Sulfa-22 could significantly inhibit the activation of microglia, decrease the expression of inflammatory factors in the central nervous system and simultaneously suppress the production of p30-GSDMD as well as the expression of upstream NLRP3 inflammasome and Caspase-1 protein. Immunoprecipitation and Biotin-labelled assay confirmed the targeted binding relationship of Sulfa-4 and Sulfa-22 with GSDMD protein in the iBMDM model in vitro. In this study, we investigated a new type inhibitor of GSDMD cleavage, which exerted a good inhibitory effect on pyroptosis and provided new references for the development of inflammatory drugs in the future.


Subject(s)
Alzheimer Disease/complications , Anti-Inflammatory Agents/pharmacology , Mafenide/pharmacology , Neuroinflammatory Diseases/etiology , Pyroptosis/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Biomarkers , Cell Line , Cytokines/metabolism , Disease Management , Disease Models, Animal , Disease Susceptibility , Dose-Response Relationship, Drug , Drug Discovery , Drug Evaluation, Preclinical/methods , Inflammation Mediators , Mafenide/analogs & derivatives , Mafenide/chemistry , Mice , Microglia/drug effects , Microglia/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Structure-Activity Relationship
5.
Sci Rep ; 11(1): 19877, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615939

ABSTRACT

ATP-dependent P2X3 receptors play a crucial role in the sensitization of nerve fibers and pathological pain pathways. They are also involved in pathways triggering cough and may contribute to the pathophysiology of endometriosis and overactive bladder. However, despite the strong therapeutic rationale for targeting P2X3 receptors, preliminary antagonists have been hampered by off-target effects, including severe taste disturbances associated with blocking the P2X2/3 receptor heterotrimer. Here we present a P2X3 receptor antagonist, eliapixant (BAY 1817080), which is both highly potent and selective for P2X3 over other P2X subtypes in vitro, including P2X2/3. We show that eliapixant reduces inflammatory pain in relevant animal models. We also provide the first in vivo experimental evidence that P2X3 antagonism reduces neurogenic inflammation, a phenomenon hypothesised to contribute to several diseases, including endometriosis. To test whether eliapixant could help treat endometriosis, we confirmed P2X3 expression on nerve fibers innervating human endometriotic lesions. We then demonstrate that eliapixant reduces vaginal hyperalgesia in an animal model of endometriosis-associated dyspareunia, even beyond treatment cessation. Our findings indicate that P2X3 antagonism could alleviate pain, including non-menstrual pelvic pain, and modify the underlying disease pathophysiology in women with endometriosis. Eliapixant is currently under clinical development for the treatment of disorders associated with hypersensitive nerve fibers.


Subject(s)
Nerve Fibers/drug effects , Nerve Fibers/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X3/metabolism , Somatosensory Disorders/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Disease Models, Animal , Female , Gene Expression , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Membrane Potentials/drug effects , Mice , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Rats , Receptors, Purinergic P2X3/genetics , Somatosensory Disorders/drug therapy , Somatosensory Disorders/etiology
6.
Int Immunopharmacol ; 101(Pt A): 108181, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607229

ABSTRACT

Demyelinating diseases of the central nervous system are characterized by recurrent demyelination and progressive neurodegeneration, but there are no clinical drugs targeting myelin regeneration or improving functional disability in the treatment of multiple sclerosis. Total flavone of Epimedium (TFE) is the main active components of Epimedium, which exhibits the beneficial biological activities in the treatment of diseases, but there is no report in the treatment of demyelinating disorder. The purpose of this study was to explore the therapeutic potential and possible mechanism of TFE in the treatment of demyelination. The results showed that TFE efficiently improved the behavioural performance and histological demyelination in cuprizone (CPZ)-induced demyelinating model. In terms of action, TFE increased astrocytes enrichment in corpus callosum, striatum and cortex, and promoted astrocytes to express neurotrophic factors. Furthermore, the expression of platelet-activating factor receptor (PAFR) in astrocytes was induced by CPZ feeding and LPS stimulation, accompanied by the increase of inflammatory cytokines TNF-α,IL-6 and IL-1ß. TFE declined the expression of PAFR, and inhibited inflammatory response. At the same time, TFE also antagonized PAFR activation and inflammatory response triggered by PAF, which further confirmed that TFE, as a new PAFR antagonist, inhibited the astrocyte-derived inflammatory response by antagonizing PAFR-neuroinflammation axis, thus contributing to myelin protection and regeneration.


Subject(s)
Demyelinating Diseases/drug therapy , Epimedium/chemistry , Neuroinflammatory Diseases/drug therapy , Plant Extracts/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Cuprizone/administration & dosage , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Flavones/pharmacology , Flavones/therapeutic use , Humans , Male , Mice , Myelin Sheath/drug effects , Myelin Sheath/immunology , Myelin Sheath/pathology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Plant Extracts/therapeutic use
7.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34497121

ABSTRACT

Alzheimer's disease (AD) is a progressive and fatal neurodegenerative disorder. Impaired neuronal bioenergetics and neuroinflammation are thought to play key roles in the progression of AD, but their interplay is not clear. Nicotinamide adenine dinucleotide (NAD+) is an important metabolite in all human cells in which it is pivotal for multiple processes including DNA repair and mitophagy, both of which are impaired in AD neurons. Here, we report that levels of NAD+ are reduced and markers of inflammation increased in the brains of APP/PS1 mutant transgenic mice with beta-amyloid pathology. Treatment of APP/PS1 mutant mice with the NAD+ precursor nicotinamide riboside (NR) for 5 mo increased brain NAD+ levels, reduced expression of proinflammatory cytokines, and decreased activation of microglia and astrocytes. NR treatment also reduced NLRP3 inflammasome expression, DNA damage, apoptosis, and cellular senescence in the AD mouse brains. Activation of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are associated with DNA damage and senescence. cGAS-STING elevation was observed in the AD mice and normalized by NR treatment. Cell culture experiments using microglia suggested that the beneficial effects of NR are, in part, through a cGAS-STING-dependent pathway. Levels of ectopic (cytoplasmic) DNA were increased in APP/PS1 mutant mice and human AD fibroblasts and down-regulated by NR. NR treatment induced mitophagy and improved cognitive and synaptic functions in APP/PS1 mutant mice. Our findings suggest a role for NAD+ depletion-mediated activation of cGAS-STING in neuroinflammation and cellular senescence in AD.


Subject(s)
Alzheimer Disease/complications , Cellular Senescence , Dietary Supplements , Membrane Proteins/metabolism , NAD/administration & dosage , Neuroinflammatory Diseases/drug therapy , Nucleotidyltransferases/metabolism , Animals , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Nucleotidyltransferases/genetics , Pyridinium Compounds/administration & dosage
8.
Neurotoxicology ; 87: 106-119, 2021 12.
Article in English | MEDLINE | ID: mdl-34509511

ABSTRACT

Organophosphate (OP) nerve agents and pesticides are a class of neurotoxic compounds that can cause status epilepticus (SE), and death following acute high-dose exposures. While the standard of care for acute OP intoxication (atropine, oxime, and high-dose benzodiazepine) can prevent mortality, survivors of OP poisoning often experience long-term brain damage and cognitive deficits. Preclinical studies of acute OP intoxication have primarily used rat models to identify candidate medical countermeasures. However, the mouse offers the advantage of readily available knockout strains for mechanistic studies of acute and chronic consequences of OP-induced SE. Therefore, the main objective of this study was to determine whether a mouse model of acute diisopropylfluorophosphate (DFP) intoxication would produce acute and chronic neurotoxicity similar to that observed in rat models and humans following acute OP intoxication. Adult male C57BL/6J mice injected with DFP (9.5 mg/kg, s.c.) followed 1 min later with atropine sulfate (0.1 mg/kg, i.m.) and 2-pralidoxime (25 mg/kg, i.m.) developed behavioral and electrographic signs of SE within minutes that continued for at least 4 h. Acetylcholinesterase inhibition persisted for at least 3 d in the blood and 14 d in the brain of DFP mice relative to vehicle (VEH) controls. Immunohistochemical analyses revealed significant neurodegeneration and neuroinflammation in multiple brain regions at 1, 7, and 28 d post-exposure in the brains of DFP mice relative to VEH controls. Deficits in locomotor and home-cage behavior were observed in DFP mice at 28 d post-exposure. These findings demonstrate that this mouse model replicates many of the outcomes observed in rats and humans acutely intoxicated with OPs, suggesting the feasibility of using this model for mechanistic studies and therapeutic screening.


Subject(s)
Brain/pathology , Isoflurophate/toxicity , Status Epilepticus/chemically induced , Acetylcholinesterase/metabolism , Animals , Brain/drug effects , Brain/enzymology , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Electroencephalography , Female , Male , Mice , Mice, Inbred C57BL , Nesting Behavior/drug effects , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/psychology , Open Field Test , Status Epilepticus/pathology , Status Epilepticus/psychology
9.
Eur J Pharmacol ; 909: 174362, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34297968

ABSTRACT

The functions and mechanisms of GPR40 receptor to ameliorating the Alzheimer's disease (AD) by external treatment of encephalopathy remain unknown. In present study, the typical Aß1-42 induced mice model was applied to explore the functions and mechanisms of GPR40 receptor by external treatment of encephalopathy in AD. GPR40 agonist GW9508 and antagonist GW1100 were given by i.g injection to activate/inhibit the GPR40 receptor respectively in the gut of AD mouse which illustrated the function and mechanism of GPR40 receptor in ameliorating AD symptoms by external treatment of encephalopathy. A series of behavioral experiments were used to investigate the cognitive function and memory ability of mice, while molecular biology experiments such as Western blot, ELISA, flow cytometry were used to detect the corresponding changes of signaling pathways. The results revealed that intragastric administrated GW9508 could significantly ameliorate cognitive deficits of AD mouse, up-regulate the expression levels of gut-brain peptides both in blood circulation and hypothalamus thus up-regulate the expression levels of α-MSH in hypothalamus, while the negative autophagy-related proteins and inflammation-related proteins were down-regulated correspondingly. Meanwhile, GW9508 could also inhibit the pathological process of neuroinflammation in microglia. GW1100 reversed the effects of GW9508 significantly. These results suggested that GPR40 was an underlying therapeutic target for the external treatment of encephalopathy related to AD and GPR40 agonist could be explored as the emerging AD therapeutic drug.


Subject(s)
Alzheimer Disease/drug therapy , Cognitive Dysfunction/drug therapy , Methylamines/administration & dosage , Neuroinflammatory Diseases/drug therapy , Propionates/administration & dosage , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Alzheimer Disease/complications , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/toxicity , Animals , Behavior Observation Techniques , Blood-Brain Barrier/metabolism , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/immunology , Cognitive Dysfunction/pathology , Down-Regulation/drug effects , Humans , Hypothalamus/drug effects , Hypothalamus/immunology , Hypothalamus/pathology , Male , Methylamines/pharmacokinetics , Mice , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Neuroinflammatory Diseases/diagnosis , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/toxicity , Propionates/pharmacokinetics , Receptors, G-Protein-Coupled/metabolism , Tissue Distribution
10.
Pharm Biol ; 59(1): 828-839, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34196587

ABSTRACT

CONTEXT: Pien-Tze-Huang (PTH) is traditionally applied to treat various inflammation-related diseases including stroke. However, literature regarding the anti-inflammatory effects and possible mechanisms of PTH in ischaemic stroke is unavailable. OBJECTIVE: This study investigates the anti-inflammatory effects and its underlying mechanism of PTH on ischaemic stroke. MATERIALS AND METHODS: Cerebral ischaemia-reperfusion injury was induced through 2 h middle cerebral artery occlusion (MCAO) followed by 24 h reperfusion in male Sprague-Dawley (SD) rats receiving oral pre-treatment with PTH (180 mg/kg) for 4 days. TLR4 antagonist TAK-242 (3 mg/kg) was injected intraperitoneally at 1.5 h after MCAO. MRI, HE staining, qRT-PCR, western blot, and immunofluorescence methods were employed. RESULTS: PTH treatment markedly reduced cerebral infarct volume (by 51%), improved neurological function (by 33%), and ameliorated brain histopathological damage in MCAO rats. It also reduced the levels of four inflammatory mediators including IL-1ß (by 70%), IL-6 (by 78%), TNF-α (by 60%) and MCP-1 (by 58%); inhibited microglia and astrocyte activation; and decreased protein expression of iNOS and COX-2 in injured brains. Moreover, PTH down-regulated the protein expressions of TLR4, MyD88, and TRAF6; reduced the expression and nuclear translocation of NF-κB; and lowered the protein expressions of p-ERK1/2, p-JNK, and p-p38. Similar effects were observed in MCAO rats with TAK-242 treatment. However, combined administration of PTH and TAK-242 did not significantly reinforce the anti-inflammatory effects of PTH. DISCUSSION AND CONCLUSION: PTH improved cerebral ischaemia-reperfusion injury by inhibiting neuroinflammation partly via the TLR4/NF-κB/MAPK signalling pathway, which will help guide its clinical application.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Ischemic Stroke/drug therapy , Neuroinflammatory Diseases/drug therapy , Animals , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery , Ischemic Stroke/pathology , MAP Kinase Signaling System/drug effects , Male , NF-kappa B/metabolism , Neuroinflammatory Diseases/pathology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Sulfonamides/pharmacology , Toll-Like Receptor 4/metabolism
11.
J Ethnopharmacol ; 279: 114309, 2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34119609

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ficus deltoidea Jack (FD) is widely consumed in traditional medicine as a treatment for various diseases in Malaysia. Each part of the plant such as its leave, stem, fruit and root are used traditionally to treat different types of diseases. Vitexin and isovitexin are bioactive compounds abundantly found in the leaves of FD that possessed many pharmacological properties including neuroprotection. Nonetheless, its effects on key events in neuroinflammation are unknown. AIM OF THE STUDY: To determine the inhibitory properties of FD aqueous extract on pro-inflammatory mediators involved in lipopolysaccharide (LPS)-induced microglial cells. METHODS: Vitexin and isovitexin in the extract were quantified via high performance liquid chromatography (HPLC). The extract was evaluated for its cytotoxicity activity via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Pre-treatment with the extract on LPS-induced microglial cells was done to determine its antioxidant and anti-neuroinflammatory properties by measuring the level of reactive oxygen species (ROS), nitric oxide (NO), tumour necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) via 2'-7'-dichlorofluorescin diacetate (DCFDA) assay, Griess assay and Western blot respectively. RESULTS: The extract at all tested concentrations (0.1 µg/mL, 1 µg/mL, 10 µg/mL, 100 µg/mL) were not cytotoxic as the percentage viability of microglial cells were all above ~80%. At the highest concentration (100 µg/mL), the extract significantly reduced the formation of ROS, NO, TNF-α, IL-1ß and IL-6 in microglial cells induced by LPS. CONCLUSION: The extract showed neuroprotective effects by attenuating the levels of pro-inflammatory and cytotoxic factors in LPS-induced microglial cells, possibly by mediating the nuclear factor-kappa B (NF-κB) signalling pathway.


Subject(s)
Ficus/chemistry , Microglia/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Antioxidants/administration & dosage , Antioxidants/isolation & purification , Antioxidants/pharmacology , Apigenin/isolation & purification , Apigenin/pharmacology , Cell Line , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Lipopolysaccharides , Mice , Microglia/pathology , NF-kappa B/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/isolation & purification , Plant Extracts/administration & dosage , Plant Leaves
12.
J Neuroinflammation ; 18(1): 137, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34130727

ABSTRACT

BACKGROUND: The secondary injury caused by traumatic brain injury (TBI), especially white matter injury (WMI), is highly sensitive to neuroinflammation, which further leads to unfavored long-term outcomes. Although the cross-talk between the three active events, immune cell infiltration, BBB breakdown, and proinflammatory microglial/macrophage polarization, plays a role in the vicious cycle, its mechanisms are not fully understood. It has been reported that cordycepin, an extract from Cordyceps militaris, can inhibit TBI-induced neuroinflammation although the long-term effects of cordycepin remain unknown. Here, we report our investigation of cordycepin's long-term neuroprotective function and its underlying immunological mechanism. METHODS: TBI mice model was established with a controlled cortical impact (CCI) method. Cordycepin was intraperitoneally administered twice daily for a week. Neurological outcomes were assessed by behavioral tests, including grid walking test, cylinder test, wire hang test, and rotarod test. Immunofluorescence staining, transmission electron microscopy, and electrophysiology recording were employed to assess histological and functional lesions. Quantitative-PCR and flow cytometry were used to detect neuroinflammation. The tracers of Sulfo-NHS-biotin and Evans blue were assessed for the blood-brain barrier (BBB) leakage. Western blot and gelatin zymography were used to analyze protein activity or expression. Neutrophil depletion in vivo was performed via using Ly6G antibody intraperitoneal injection. RESULTS: Cordycepin administration ameliorated long-term neurological deficits and reduced neuronal tissue loss in TBI mice. Meanwhile, the long-term integrity of white matter was also preserved, which was revealed in multiple dimensions, such as morphology, histology, ultrastructure, and electrical conductivity. Cordycepin administration inhibited microglia/macrophage pro-inflammatory polarization and promoted anti-inflammatory polarization after TBI. BBB breach was attenuated by cordycepin administration at 3 days after TBI. Cordycepin suppressed the activities of MMP-2 and MMP-9 and the neutrophil infiltration at 3 days after TBI. Moreover, neutrophil depletion provided a cordycepin-like effect, and cordycepin administration united with neutrophil depletion did not show a benefit of superposition. CONCLUSIONS: The long-term neuroprotective function of cordycepin via suppressing neutrophil infiltration after TBI, thereby preserving BBB integrity and changing microglia/macrophage polarization. These findings provide significant clinical potentials to improve the quality of life for TBI patients.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Deoxyadenosines/therapeutic use , Neuroinflammatory Diseases/prevention & control , Neuroprotection/drug effects , Neuroprotective Agents , Neutrophil Infiltration/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/drug effects , Brain/pathology , Deoxyadenosines/pharmacology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
13.
Nutrients ; 14(1)2021 Dec 30.
Article in English | MEDLINE | ID: mdl-35011052

ABSTRACT

Perinatal hypoxia-ischemia (HI) is a major cause of neonatal brain injury, leading to long-term neurological impairments. Medical nutrition can be rapidly implemented in the clinic, making it a viable intervention to improve neurodevelopment after injury. The omega-3 (n-3) fatty acids docosahexaenoic acid (DHA, 22:6n-3) and eicosapentaenoic acid (EPA, 20:5n-3), uridine monophosphate (UMP) and choline have previously been shown in rodents to synergistically enhance brain phospholipids, synaptic components and cognitive performance. The objective of this study was to test the efficacy of an experimental diet containing DHA, EPA, UMP, choline, iodide, zinc, and vitamin B12 in a mouse model of perinatal HI. Male and female C57Bl/6 mice received the experimental diet or an isocaloric control diet from birth. Hypoxic ischemic encephalopathy was induced on postnatal day 9 by ligation of the right common carotid artery and systemic hypoxia. To assess the effects of the experimental diet on long-term motor and cognitive outcome, mice were subjected to a behavioral test battery. Lesion size, neuroinflammation, brain fatty acids and phospholipids were analyzed at 15 weeks after HI. The experimental diet reduced lesion size and neuroinflammation specifically in males. In both sexes, brain n-3 fatty acids were increased after receiving the experimental diet. The experimental diet also improved novel object recognition, but no significant effects on motor performance were observed. Current data indicates that early life nutritional supplementation with a combination of DHA, EPA, UMP, choline, iodide, zinc, and vitamin B12 may provide neuroprotection after perinatal HI.


Subject(s)
Choline/administration & dosage , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Hypoxia-Ischemia, Brain/diet therapy , Neuroinflammatory Diseases/diet therapy , Uridine Monophosphate/administration & dosage , Animals , Brain/metabolism , Disease Models, Animal , Fatty Acids, Omega-3/metabolism , Female , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Male , Mice, Inbred C57BL , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Sex Characteristics
SELECTION OF CITATIONS
SEARCH DETAIL