Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 612
Filter
1.
J Neuroinflammation ; 21(1): 216, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39218899

ABSTRACT

Parkinson's disease (PD) is characterized by neuroinflammation, progressive loss of dopaminergic neurons, and accumulation of α-synuclein (α-Syn) into insoluble aggregates called Lewy pathology. The Line 61 α-Syn mouse is an established preclinical model of PD; Thy-1 is used to promote human α-Syn expression, and features of sporadic PD develop at 9-18 months of age. To accelerate the PD phenotypes, we injected sonicated human α-Syn preformed fibrils (PFFs) into the striatum, which produced phospho-Syn (p-α-Syn) inclusions in the substantia nigra pars compacta and significantly increased MHC Class II-positive immune cells. Additionally, there was enhanced infiltration and activation of innate and adaptive immune cells in the midbrain. We then used this new model, Line 61-PFF, to investigate the effect of inhibiting the JAK/STAT signaling pathway, which is critical for regulation of innate and adaptive immune responses. After administration of the JAK1/2 inhibitor AZD1480, immunofluorescence staining showed a significant decrease in p-α-Syn inclusions and MHC Class II expression. Flow cytometry showed reduced infiltration of CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, dendritic cells, macrophages, and endogenous microglia into the midbrain. Importantly, single-cell RNA-Sequencing analysis of CD45+ cells from the midbrain identified 9 microglia clusters, 5 monocyte/macrophage (MM) clusters, and 5 T-cell (T) clusters, in which potentially pathogenic MM4 and T3 clusters were associated with neuroinflammatory responses in Line 61-PFF mice. AZD1480 treatment reduced cell numbers and cluster-specific expression of the antigen-presentation genes H2-Eb1, H2-Aa, H2-Ab1, and Cd74 in the MM4 cluster and proinflammatory genes such as Tnf, Il1b, C1qa, and C1qc in the T3 cluster. Together, these results indicate that inhibiting the JAK/STAT pathway suppresses the activation and infiltration of innate and adaptive cells, reducing neuroinflammation in the Line 61-PFF mouse model.


Subject(s)
Disease Models, Animal , Neuroinflammatory Diseases , Parkinson Disease , STAT Transcription Factors , Signal Transduction , alpha-Synuclein , Animals , Mice , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , STAT Transcription Factors/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/immunology , Humans , Mice, Transgenic , Mice, Inbred C57BL , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/immunology , Pyrimidines/pharmacology
2.
FASEB J ; 38(17): e70030, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39221499

ABSTRACT

Citicoline, a compound produced naturally in small amounts in the human body, assumes a pivotal role in phosphatidylcholine synthesis, a dynamic constituent of membranes of neurons. Across diverse models of brain injury and neurodegeneration, citicoline has demonstrated its potential through neuroprotective and anti-inflammatory effects. This review aims to elucidate citicoline's anti-inflammatory mechanism and its clinical implications in conditions such as ischemic stroke, head trauma, glaucoma, and age-associated memory impairment. Citicoline's anti-inflammatory prowess is rooted in its ability to stabilize cellular membranes, thereby curbing the excessive release of glutamate-a pro-inflammatory neurotransmitter. Moreover, it actively diminishes free radicals and inflammatory cytokines productions, which could otherwise harm neurons and incite neuroinflammation. It also exhibits the potential to modulate microglia activity, the brain's resident immune cells, and hinder the activation of NF-κB, a transcription factor governing inflammatory genes. Clinical trials have subjected citicoline to rigorous scrutiny in patients grappling with acute ischemic stroke, head trauma, glaucoma, and age-related memory impairment. While findings from these trials are mixed, numerous studies suggest that citicoline could confer improvements in neurological function, disability reduction, expedited recovery, and cognitive decline prevention within these cohorts. Additionally, citicoline boasts a favorable safety profile and high tolerability. In summary, citicoline stands as a promising agent, wielding both neuroprotective and anti-inflammatory potential across a spectrum of neurological conditions. However, further research is imperative to delineate the optimal dosage, treatment duration, and underlying mechanisms. Moreover, identifying specific patient subgroups most likely to reap the benefits of citicoline as a new therapy remains a critical avenue for exploration.


Subject(s)
Cytidine Diphosphate Choline , Neuroinflammatory Diseases , Cytidine Diphosphate Choline/therapeutic use , Cytidine Diphosphate Choline/pharmacology , Humans , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Animals , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Nootropic Agents/therapeutic use , Nootropic Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Clinical Relevance
3.
J Nanobiotechnology ; 22(1): 534, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227960

ABSTRACT

Ischemic stroke is a common cause of mortality and severe disability in human and currently lacks effective treatment. Neuronal activation and neuroinflammation are the major two causes of neuronal damage. However, little is known about the connection of these two phenomena. This study uses middle cerebral artery occlusion mouse model and chemogenetic techniques to study the underlying mechanisms of neuronal excitotoxicity and severe neuroinflammation after ischemic stroke. Chemogenetic inhibition of neuronal activity in ipsilesional M1 alleviates infarct area and neuroinflammation, and improves motor recovery in ischemia mice. This study identifies that ischemic challenge triggers neuron to produce unique small extracellular vesicles (EVs) to aberrantly activate adjacent neurons which enlarge the neuron damage range. Importantly, these EVs also drive microglia activation to exacerbate neuroinflammation. Mechanistically, EVs from ischemia-evoked neuronal activity induce neuronal apoptosis and innate immune responses by transferring higher miR-100-5p to adjacent neuron and microglia. MiR-100-5p can bind to and activate TLR7 through U18U19G20-motif, thereby activating NF-κB pathway. Furthermore, knock-down of miR-100-5p expression improves poststroke outcomes in mice. Taken together, this study suggests that the combination of inhibiting aberrant neuronal activity and the secretion of specific EVs-miRNAs may serve as novel methods for stroke treatment.


Subject(s)
Extracellular Vesicles , Mice, Inbred C57BL , MicroRNAs , Microglia , Neurons , Stroke , Animals , Extracellular Vesicles/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Microglia/metabolism , Neurons/metabolism , Mice , Male , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Disease Models, Animal , NF-kappa B/metabolism , Neuroinflammatory Diseases/metabolism , Infarction, Middle Cerebral Artery , Apoptosis , Immunity, Innate , Humans , Membrane Glycoproteins
4.
Mol Brain ; 17(1): 62, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223564

ABSTRACT

We previously demonstrated that felodipine, an L-type calcium channel blocker, inhibits LPS-mediated neuroinflammatory responses in BV2 microglial cells and wild-type mice. However, the effects of felodipine on tau pathology, a hallmark of Alzheimer's disease (AD), have not been explored yet. Therefore, in the present study, we determined whether felodipine affects neuroinflammation and tau hyperphosphorylation in 3-month-old P301S transgenic mice (PS19), an early phase AD mice model for tauopathy. Felodipine administration decreased tauopathy-mediated microglial activation and NLRP3 expression in PS19 mice but had no effect on tauopathy-associated astrogliosis. In addition, felodipine treatment significantly reduced tau hyperphosphorylation at S202/Thr205 and Thr212/Ser214 residues via inhibiting JNK/P38 signaling in PS19 mice. Collectively, our results suggest that felodipine significantly ameliorates tau hyper-phosphorylation and tauopathy-associated neuroinflammatory responses in AD mice model for tauopathy and could be a novel therapeutic agent for AD.


Subject(s)
Alzheimer Disease , Felodipine , Mice, Transgenic , Microglia , Neuroinflammatory Diseases , p38 Mitogen-Activated Protein Kinases , tau Proteins , Animals , tau Proteins/metabolism , Phosphorylation/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Felodipine/pharmacology , Felodipine/therapeutic use , Alzheimer Disease/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , MAP Kinase Signaling System/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Disease Models, Animal , Mice, Inbred C57BL
5.
Cell Commun Signal ; 22(1): 427, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223674

ABSTRACT

BACKGROUND: Depression is often linked to inflammation in the brain. Researchers have been exploring ways to reduce this inflammation to improve depression symptoms. One potential target is a protein called RIPK1, which is known to contribute to brain inflammation. However, it's unclear how RIPK1 influences depression. Our study aims to determine whether RIPK1 inhibition could alleviate neuroinflammation-associated depression and elucidate its underlying mechanisms. METHODS: To investigate our research objectives, we established a neuroinflammation mouse model by administering LPS. Behavioral and biochemical assessments were conducted on these mice. The findings were subsequently validated through in vitro experiments. RESULTS: Using LPS-induced depression models, we investigated RIPK1's role, observing depressive-like behaviors accompanied by elevated cytokines, IBA-1, GFAP levels, and increased inflammatory signaling molecules and NO/H2O2. Remarkably, Necrostatin (Nec-1 S), a RIPK1 inhibitor, mitigated these changes. We further found altered expression and phosphorylation of eIF4E, PI3K/AKT/mTOR, and synaptic proteins in hippocampal tissues, BV2, and N2a cells post-LPS treatment, which Nec-1 S also ameliorated. Importantly, eIF4E inhibition reversed some of the beneficial effects of Nec-1 S, suggesting a complex interaction between RIPK1 and eIF4E in LPS-induced neuroinflammation. Moreover, citronellol, a RIPK1 agonist, significantly altered eIF4E phosphorylation, indicating RIPK1's potential upstream regulatory role in eIF4E and its contribution to neuroinflammation-associated depression. CONCLUSION: These findings propose RIPK1 as a pivotal mediator in regulating neuroinflammation and neural plasticity, highlighting its significance as a potential therapeutic target for depression.


Subject(s)
Depression , Disease Models, Animal , Lipopolysaccharides , Mice, Inbred C57BL , Neuroinflammatory Diseases , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Lipopolysaccharides/pharmacology , Depression/drug therapy , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Mice , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Male , Behavior, Animal/drug effects , Indoles/pharmacology , Indoles/therapeutic use , Signal Transduction/drug effects , Imidazoles/pharmacology , Imidazoles/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/drug therapy , Inflammation/pathology
6.
Front Immunol ; 15: 1418422, 2024.
Article in English | MEDLINE | ID: mdl-39136022

ABSTRACT

Introduction: Alzheimer pathology (AD) is characterized by the deposition of amyloid beta (Aß) and chronic neuroinflammation, with the NLRP3 inflammasome playing a significant role. This study demonstrated that the OCD drug fluvoxamine maleate (FXN) can potently ameliorate AD pathology in 5XFAD mice by promoting autophagy-mediated clearance of Aß and inhibiting the NLRP3 inflammasome. Methods: We used mice primary astrocytes to establish the mechanism of action of FXN against NLRP3 inflammasome by using various techniques like ELISA, Western blotting, confocal microscopy, Immunofluorescence, etc. The anti-AD activity of FXN was validated in transgenic 5XFAD mice following two months of treatment. This was followed by behavior analysis, examination of inflammatory and autophagy proteins and immunohistochemistry analysis for Aß load in the hippocampi. Results: Our data showed that FXN, at a low concentration of 78 nM, induces autophagy to inhibit NF-κB and the NLRP3 inflammasome, apart from directly inhibiting NLRP3 inflammasome in primary astrocytes. FXN activated the PRKAA2 pathway through CAMKK2 signaling, leading to autophagy induction. It inhibited the ATP-mediated NLRP3 inflammasome activation by promoting the autophagic degradation of NF-κB, resulting in the downregulation of pro-IL-1ß and NLRP3. The anti-NLRP3 inflammasome effect of FXN was reversed when autophagy was inhibited by either genetic knockdown of the PRKAA2 pathway or pharmacological inhibition with bafilomycin A1. Furthermore, FXN treatment led to improved AD pathology in 5XFAD mice, resulting in significant improvements in various behavioral parameters such as working memory and neuromuscular coordination, making their behavior more similar to that of wild-type animals. FXN improved behavior in 5XFAD mice by clearing the Aß deposits from the hippocampi and significantly reducing multiple inflammatory proteins, including NF-κB, GFAP, IBA1, IL-1ß, TNF-α, and IL-6, which are associated with NF-κB and NLRP3 inflammasome in the brain. Moreover, these changes were accompanied by increased expression of autophagic proteins. Discussion: Our data suggest that FXN ameliorates AD pathology, by simultaneously targeting two key pathological features: Aß deposits and neuroinflammation. As an already approved drug, FXN holds potential as a candidate for human studies against AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Astrocytes , Autophagy , Disease Models, Animal , Fluvoxamine , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Mice , Fluvoxamine/pharmacology , Fluvoxamine/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Autophagy/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects
7.
Mol Biol Rep ; 51(1): 908, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141244

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by oxidative stress and neuroinflammation. Sofalcone (SFC), a chalcone derivative known for its antioxidative and anti-inflammatory properties, is widely used clinically as a gastric mucosa protective agent. However, its therapeutic potential in PD remains to be fully explored. In this study, we investigated the neuroprotective effects of SFC in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. METHODS AND RESULTS: We found that SFC ameliorated MPTP-induced motor impairments in mice, as assessed by the rotarod and wire tests. Moreover, SFC administration prevented the loss of dopaminergic neurons and striatal degeneration induced by MPTP. Subsequent investigations revealed that SFC reversed MPTP-induced downregulation of NRF2, reduced elevated levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and increased total antioxidant capacity (TAOC). Furthermore, SFC suppressed MPTP-induced activation of microglia and astrocytes, downregulated the pro-inflammatory cytokine TNF-α, and upregulated the anti-inflammatory cytokine IL-4. Additionally, SFC ameliorated the MPTP-induced downregulation of phosphorylation of Akt at Ser473. CONCLUSIONS: This study provides evidence for the neuroprotective effects of SFC, highlighting its antioxidative and anti-inflammatory properties and its role in Akt activation in the PD model. These findings underscore SFC's potential as a promising therapeutic candidate for PD, warranting further clinical investigation.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Chalcones , Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Animals , Oxidative Stress/drug effects , Mice , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Chalcones/pharmacology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Antioxidants/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Microglia/drug effects , Microglia/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , NF-E2-Related Factor 2/metabolism , Anti-Inflammatory Agents/pharmacology
8.
J Neurosci Res ; 102(8): e25370, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39158105

ABSTRACT

Resistance exercise training (RET) is considered an excellent tool for preventing diseases with an inflammatory background. Its neuroprotective, antioxidant, and anti-inflammatory properties are responsible for positively modulating cholinergic and oxidative systems, promoting neurogenesis, and improving memory. However, the mechanisms behind these actions are largely unknown. In order to investigate the pathways related to these effects of exercise, we conducted a 12-week long-term exercise training protocol and used lipopolysaccharide (LPS) to induce damage to the cortex and hippocampus of male Wistar rats. The cholinergic system, oxidative stress, and histochemical parameters were analyzed in the cerebral cortex and hippocampus, and memory tests were also performed. It was observed that LPS: (1) caused memory loss in the novel object recognition (NOR) test; (2) increased the activity of acetylcholinesterase (AChE) and Iba1 protein density; (3) reduced the protein density of brain-derived neurotrophic factor (BDNF) and muscarinic acetylcholine receptor M1 (CHRM1); (4) elevated the levels of lipid peroxidation (TBARS) and reactive species (RS); and (5) caused inflammatory damage to the dentate gyrus. RET, on the other hand, was able to prevent all alterations induced by LPS, as well as increase per se the protein density of the alpha-7 nicotinic acetylcholine receptor (nAChRα7) and Nestin, and the levels of protein thiols (T-SH). Overall, our study elucidates some mechanisms that support resistance physical exercise as a valuable approach against LPS-induced neuroinflammation and memory loss.


Subject(s)
Lipopolysaccharides , Memory Disorders , Neuroinflammatory Diseases , Physical Conditioning, Animal , Rats, Wistar , Animals , Male , Lipopolysaccharides/toxicity , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/methods , Rats , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Memory Disorders/chemically induced , Memory Disorders/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Resistance Training/methods , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Lipid Peroxidation/drug effects , Acetylcholinesterase/metabolism , Receptor, Muscarinic M1/metabolism
9.
Neurosci Lett ; 837: 137923, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39106918

ABSTRACT

Caffeine, a nonselective adenosine receptor antagonist, is the major component of coffee and the most consumed psychostimulant at nontoxic doses in the world. It has been identified that caffeine consumption reduces the risk of several neurological diseases. However, the mechanisms by which it impacts the pathophysiology of neurological diseases remain to be elucidated. In this study, we investigated whether caffeine exerts anti-inflammatory effects on lipopolysaccharide (LPS)-induced inflammation and depression in vivo and explored the potential mechanism of caffeine through LPS-induced brain injury. Adult male Sprague-Dawley (SD) rats were intraperitoneal injected with various concentrations of LPS to induce the neuroinflammation and depressive-like behavior. Then SD rats were treated with caffeine in the presence or absence of LPS. Open-filed and closed-field tests were applied to detect the behaviors of SD rats, while western blot was performed to measure the phosphorylation level of protein kinase B (p-AKT) and nuclear factor κB (NF-κB) in the cortex after caffeine was orally administered. Our findings indicated that caffeine markedly improved the neuroinflammation and depressive-like behavior of LPS-treated SD rats. Mechanistic investigations demonstrated that caffeine down-regulated the expression of p-AKT and NF-κB in LPS-induced SD rats cortex. Taken together, these results indicated that caffeine, a potential agent for preventing inflammatory diseases, may suppress LPS-induced inflammatory and depressive responses by regulating AKT phosphorylation and NF-κB.


Subject(s)
Caffeine , Depression , Lipopolysaccharides , NF-kappa B , Neuroinflammatory Diseases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Animals , NF-kappa B/metabolism , Male , Caffeine/pharmacology , Caffeine/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Rats , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Phosphorylation/drug effects , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/chemically induced
10.
CNS Neurosci Ther ; 30(8): e14913, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39123294

ABSTRACT

BACKGROUND: Hyperglycemia-induced neuroinflammation significantly contributes to diabetic neuropathic pain (DNP), but the underlying mechanisms remain unclear. OBJECTIVE: To investigate the role of Sirt3, a mitochondrial deacetylase, in hyperglycemia-induced neuroinflammation and DNP and to explore potential therapeutic interventions. METHOD AND RESULTS: Here, we found that Sirt3 was downregulated in spinal dorsal horn (SDH) of diabetic mice by RNA-sequencing, which was further confirmed at the mRNA and protein level. Sirt3 deficiency exacerbated hyperglycemia-induced neuroinflammation and DNP by enhancing microglial aerobic glycolysis in vivo and in vitro. Overexpression of Sirt3 in microglia alleviated inflammation by reducing aerobic glycolysis. Mechanistically, high-glucose stimulation activated Akt, which phosphorylates and inactivates FoxO1. The inactivation of FoxO1 diminished the transcription of Sirt3. Besides that, we also found that hyperglycemia induced Sirt3 degradation via the mitophagy-lysosomal pathway. Blocking Akt activation by GSK69093 or metformin rescued the degradation of Sirt3 protein and transcription inhibition of Sirt3 mRNA, which substantially diminished hyperglycemia-induced inflammation. Metformin in vivo treatment alleviated neuroinflammation and diabetic neuropathic pain by rescuing hyperglycemia-induced Sirt3 downregulation. CONCLUSION: Hyperglycemia induces metabolic reprogramming and inflammatory activation in microglia through the regulation of Sirt3 transcription and degradation. This novel mechanism identifies Sirt3 as a potential drug target for treating DNP.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Down-Regulation , Glycolysis , Hyperglycemia , Mice, Inbred C57BL , Microglia , Sirtuin 3 , Animals , Sirtuin 3/metabolism , Sirtuin 3/genetics , Mice , Glycolysis/drug effects , Glycolysis/physiology , Down-Regulation/drug effects , Down-Regulation/physiology , Hyperglycemia/metabolism , Microglia/metabolism , Microglia/drug effects , Male , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/metabolism , Inflammation/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Metformin/pharmacology
11.
J Neuroimmune Pharmacol ; 19(1): 46, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162886

ABSTRACT

The mechanisms for neuropathic pain amelioration by sigma-1 receptor inhibition are not fully understood. We studied genome-wide transcriptomic changes (RNAseq) in the dorsal root ganglia (DRG) from wild-type and sigma-1 receptor knockout mice prior to and following Spared Nerve Injury (SNI). In wildtype mice, most of the transcriptomic changes following SNI are related to the immune function or neurotransmission. Immune function transcripts contain cytokines and markers for immune cells, including macrophages/monocytes and CD4 + T cells. Many of these immune transcripts were attenuated by sigma-1 knockout in response to SNI. Consistent with this we found, using flow cytometry, that sigma-1 knockout mice showed a reduction in macrophage/monocyte recruitment as well as an absence of CD4 + T cell recruitment in the DRG after nerve injury. Sigma-1 knockout mice showed a reduction of neuropathic (mechanical and cold) allodynia and spontaneous pain-like responses (licking of the injured paw) which accompany the decreased peripheral neuroinflammatory response after nerve injury. Treatment with maraviroc (a CCR5 antagonist which preferentially inhibits CD4 + T cells in the periphery) of neuropathic wild-type mice only partially replicated the sigma-1 knockout phenotype, as it did not alter cold allodynia but attenuated spontaneous pain-like responses and mechanical hypersensitivity. Therefore, modulation of peripheral CD4 + T cell activity might contribute to the amelioration of spontaneous pain and neuropathic tactile allodynia seen in the sigma-1 receptor knockout mice, but not to the effect on cold allodynia. We conclude that sigma-1 receptor inhibition decreases DRG neuroinflammation which might partially explain its anti-neuropathic effect.


Subject(s)
Mice, Inbred C57BL , Mice, Knockout , Neuralgia , Receptors, sigma , Sigma-1 Receptor , Transcriptome , Animals , Receptors, sigma/genetics , Receptors, sigma/metabolism , Receptors, sigma/antagonists & inhibitors , Neuralgia/metabolism , Mice , Female , Neuroinflammatory Diseases/metabolism , Ganglia, Spinal/metabolism , Peripheral Nerve Injuries/metabolism
12.
Cell Biol Toxicol ; 40(1): 65, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110292

ABSTRACT

The primary aim of this research was to explore the functions of Wtap and Ythdf1 in regulating neuronal Lipocalin-2 (Lcn2) through m6A modification in traumatic brain injury (TBI). By employing transcriptome sequencing and enrichment analysis, we identified the Wtap/Ythdf1-mediated Lcn2 m6A modification pathway as crucial in TBI. In our in vitro experiments using primary cortical neurons, knockout of Wtap and Ythdf1 led to the inhibition of Lcn2 m6A modification, resulting in reduced neuronal death and inflammation. Furthermore, overexpression of Lcn2 in cortical neurons induced the activation of reactive astrocytes and M1-like microglial cells, causing neuronal apoptosis. In vivo experiments confirmed the activation of reactive astrocytes and microglial cells in TBI and importantly demonstrated that Wtap knockdown improved neuroinflammation and functional impairment. These findings underscore the significance of Wtap/Ythdf1-mediated Lcn2 regulation in TBI secondary injury and suggest potential therapeutic implications for combating TBI-induced neuroinflammation and neuronal damage.


Subject(s)
Brain Injuries, Traumatic , Lipocalin-2 , Neurons , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Lipocalin-2/metabolism , Lipocalin-2/genetics , Animals , Neurons/metabolism , Neurons/pathology , Mice , Microglia/metabolism , Microglia/pathology , Astrocytes/metabolism , Astrocytes/pathology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Male , Mice, Inbred C57BL , Apoptosis , Mice, Knockout , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology
13.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39117458

ABSTRACT

Neuroinflammation, aging, and neurodegenerative disorders are associated with excessive accumulation of neutral lipids in lipid droplets (LDs) in microglia. Type 2 diabetes mellitus (T2DM) may cause neuroinflammation and is a risk factor for neurodegenerative disorders. Here, we show that hippocampal pyramidal neurons contain smaller, more abundant LDs than their neighboring microglia. The density of LDs varied between pyramidal cells in adjacent subregions, with CA3 neurons containing more LDs than CA1 neurons. Within the CA3 region, a gradual increase in the LD content along the pyramidal layer from the hilus toward CA2 was observed. Interestingly, the high neuronal LD content correlated with less ramified microglial morphotypes. Using the db/db model of T2DM, we demonstrated that diabetes increased the number of LDs per microglial cell without affecting the neuronal LD density. High-intensity interval exercise induced smaller changes in the number of LDs in microglia but was not sufficient to counteract the diabetes-induced changes in LD accumulation. The changes observed in response to T2DM may contribute to the cerebral effects of T2DM and provide a mechanistic link between T2DM and neurodegenerative disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Hippocampus , Lipid Droplets , Microglia , Neurons , Microglia/metabolism , Animals , Lipid Droplets/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Neurons/metabolism , Neurons/pathology , Male , Mice , Physical Conditioning, Animal , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Mice, Inbred C57BL , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Lipid Metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology
14.
Cells ; 13(15)2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39120330

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder that lacks effective treatment strategies to halt or delay its progression. The homeostasis of Ca2+ ions is crucial for ensuring optimal cellular functions and survival, especially for neuronal cells. In the context of PD, the systems regulating cellular Ca2+ are compromised, leading to Ca2+-dependent synaptic dysfunction, impaired neuronal plasticity, and ultimately, neuronal loss. Recent research efforts directed toward understanding the pathology of PD have yielded significant insights, particularly highlighting the close relationship between Ca2+ dysregulation, neuroinflammation, and neurodegeneration. However, the precise mechanisms driving the selective loss of dopaminergic neurons in PD remain elusive. The disruption of Ca2+ homeostasis is a key factor, engaging various neurodegenerative and neuroinflammatory pathways and affecting intracellular organelles that store Ca2+. Specifically, impaired functioning of mitochondria, lysosomes, and the endoplasmic reticulum (ER) in Ca2+ metabolism is believed to contribute to the disease's pathophysiology. The Na+-Ca2+ exchanger (NCX) is considered an important key regulator of Ca2+ homeostasis in various cell types, including neurons, astrocytes, and microglia. Alterations in NCX activity are associated with neurodegenerative processes in different models of PD. In this review, we will explore the role of Ca2+ dysregulation and neuroinflammation as primary drivers of PD-related neurodegeneration, with an emphasis on the pivotal role of NCX in the pathology of PD. Consequently, NCXs and their interplay with intracellular organelles may emerge as potentially pivotal players in the mechanisms underlying PD neurodegeneration, providing a promising avenue for therapeutic intervention aimed at halting neurodegeneration.


Subject(s)
Calcium , Neuroinflammatory Diseases , Parkinson Disease , Sodium-Calcium Exchanger , Humans , Parkinson Disease/metabolism , Parkinson Disease/pathology , Calcium/metabolism , Sodium-Calcium Exchanger/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Animals , Organelles/metabolism , Homeostasis , Mitochondria/metabolism , Mitochondria/pathology
15.
Sci Rep ; 14(1): 18002, 2024 08 03.
Article in English | MEDLINE | ID: mdl-39097642

ABSTRACT

Zika virus (ZIKV) infection was first reported in 2015 in Brazil as causing microcephaly and other developmental abnormalities in newborns, leading to the identification of Congenital Zika Syndrome (CZS). Viral infections have been considered an environmental risk factor for neurodevelopmental disorders outcome, such as Autism Spectrum Disorder (ASD). Moreover, not only the infection per se, but maternal immune system activation during pregnancy, has been linked to fetal neurodevelopmental disorders. To understand the impact of ZIKV vertical infection on brain development, we derived induced pluripotent stem cells (iPSC) from Brazilian children born with CZS, some of the patients also being diagnosed with ASD. Comparing iPSC-derived neurons from CZS with a control group, we found lower levels of pre- and postsynaptic proteins and reduced functional synapses by puncta co-localization. Furthermore, neurons and astrocytes derived from the CZS group showed decreased glutamate levels. Additionally, the CZS group exhibited elevated levels of cytokine production, one of which being IL-6, already associated with the ASD phenotype. These preliminary findings suggest that ZIKV vertical infection may cause long-lasting disruptions in brain development during fetal stages, even in the absence of the virus after birth. These disruptions could contribute to neurodevelopmental disorders manifestations such as ASD. Our study contributes with novel knowledge of the CZS outcomes and paves the way for clinical validation and the development of potential interventions to mitigate the impact of ZIKV vertical infection on neurodevelopment.


Subject(s)
Brain , Induced Pluripotent Stem Cells , Infectious Disease Transmission, Vertical , Synapses , Zika Virus Infection , Zika Virus , Humans , Zika Virus Infection/virology , Zika Virus Infection/pathology , Female , Zika Virus/pathogenicity , Synapses/pathology , Synapses/metabolism , Brain/virology , Brain/pathology , Pregnancy , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/virology , Neurons/virology , Neurons/metabolism , Neurons/pathology , Male , Astrocytes/virology , Astrocytes/metabolism , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/pathology , Brazil , Infant, Newborn , Autism Spectrum Disorder/virology , Child
16.
J Cell Mol Med ; 28(15): e18554, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39103747

ABSTRACT

Alzheimer's disease (AD) is a progressive disease that causes an impairment of learning and memory. Despite the highly complex pathogenesis of AD, amyloid beta (Aß) deposition and neurofibrillary tangles (NFTs) formation are the main hallmarks of AD. Neuroinflammation also has a crucial role in the development of AD. As the central nervous system's innate immune cells, microglial cells are activated in AD and induce inflammation by producing pro-inflammatory mediators. However, microglial activation is not always deleterious. M2-activated microglial cells are considered anti-inflammatory cells, which develop neuroprotection. Various approaches are proposed for managing AD, yet no effective therapy is available for this disorder. Considering the potential protective role of M2 microglia in neurodegenerative disorders and the improvement of these disorders by preconditioning approaches, it can be suggested that preconditioning of microglial cells may be beneficial for managing AD progression. Therefore, this study review microglial preconditioning approaches for preventing and improving AD.


Subject(s)
Alzheimer Disease , Microglia , Microglia/metabolism , Microglia/pathology , Alzheimer Disease/therapy , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Humans , Animals , Amyloid beta-Peptides/metabolism , Neuroinflammatory Diseases/metabolism
17.
J Neuroinflammation ; 21(1): 208, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169375

ABSTRACT

BACKGROUND: Obstructive sleep apnoea (OSA) is a sleep-disordered breathing characterized by intermittent hypoxia (IH) that may cause cognitive dysfunction. However, the impact of IH on molecular processes involved in cognitive function remains unclear. METHODS: C57BL / 6 J mice were exposed to either normoxia (control) or IH for 6 weeks. DNA hydroxymethylation was quantified by hydroxymethylated DNA immunoprecipitation (hMeDIP) sequencing. ten-eleven translocation 1 (Tet1) was knocked down by lentivirus. Specifically, cognitive function was assessed by behavioral experiments, pathological features were assessed by HE staining, the hippocampal DNA hydroxymethylation was examined by DNA dot blot and immunohistochemical staining, while the Wnt signaling pathway and its downstream effects were studied using qRT-PCR, immunofluorescence staining, and Luminex liquid suspension chip analysis. RESULTS: IH mice showed pathological changes and cognitive dysfunction in the hippocampus. Compared with the control group, IH mice exhibited global DNA hydroxylmethylation in the hippocampus, and the expression of three hydroxylmethylases increased significantly. The Wnt signaling pathway was activated, and the mRNA and 5hmC levels of Wnt3a, Ccnd2, and Prickle2 were significantly up-regulated. Further caused downstream neurogenesis abnormalities and neuroinflammatory activation, manifested as increased expression of IBA1 (a marker of microglia), GFAP (a marker of astrocytes), and DCX (a marker of immature neurons), as well as a range of inflammatory cytokines (e.g. TNFa, IL3, IL9, and IL17A). After Tet1 knocked down, the above indicators return to normal. CONCLUSION: Activation of Wnt signaling pathway by hippocampal Tet1 is associated with cognitive dysfunction induced by IH.


Subject(s)
Cognitive Dysfunction , Hippocampus , Mice, Inbred C57BL , Proto-Oncogene Proteins , Sleep Apnea, Obstructive , Wnt Signaling Pathway , Animals , Hippocampus/metabolism , Hippocampus/pathology , Mice , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Wnt Signaling Pathway/physiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/etiology , Sleep Apnea, Obstructive/metabolism , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/pathology , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , DNA Methylation , DNA-Binding Proteins
18.
CNS Neurosci Ther ; 30(8): e70006, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39169794

ABSTRACT

INTRODUCTION: Trigeminal neuralgia (TN), marked by chronic pain from neural damage, is closely associated with inflammation. The role of OTULIN, a key regulator in inflammation and autophagy, is not fully understood in TN. The regulatory mechanism of OTULIN, a key protein involved in modulating inflammatory responses and autophagy processes, remains incompletely elucidated, particularly in the context of TN and neuroinflammation. METHODS: An infraorbital nerve ligation-induced rat model of TN was used. OTULIN's expression was modulated using adenovirus vectors and short hairpin RNA. The impact on pain and inflammatory responses was assessed via quantitative real-time polymerase chain reaction, western blot, immunofluorescence, and transcriptomic analysis. RESULTS: Enhanced OTULIN expression significantly increased head withdrawal thresholds and reduced pain sensitivity and neuroinflammatory markers in the model. Conversely, silencing OTULIN exacerbated pain and inflammation. Transcriptomic data revealed OTULINs influence on both inflammatory and autophagy pathways, specifically in suppressing NLR family pyrin domain containing 3 (NLRP3) inflammasome and promoting autophagy. In vitro experiments demonstrated OTULIN's inhibition of inflammatory markers in microglia and neurons. CONCLUSION: OTULIN is crucial in modulating TN, reducing neuropathic pain and neuroinflammation by activating the autophagy pathway and inhibiting the NLRP3 inflammasome.


Subject(s)
Neuroinflammatory Diseases , Rats, Sprague-Dawley , Trigeminal Neuralgia , Animals , Trigeminal Neuralgia/metabolism , Rats , Neuroinflammatory Diseases/metabolism , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Autophagy/physiology , Microglia/metabolism , Inflammation/metabolism
19.
Biomed Pharmacother ; 178: 117277, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39126772

ABSTRACT

Alzheimer's disease (AD) is a progressive degenerative neurological condition characterized by cognitive decline, primarily affecting memory and logical thinking, attributed to amyloid-ß plaques and tau protein tangles in the brain, leading to neuronal loss and brain atrophy. Neuroinflammation, a hallmark of AD, involves the activation of microglia and astrocytes in response to pathological changes, potentially exacerbating neuronal damage. The gut-brain axis is a bidirectional communication pathway between the gastrointestinal and central nervous systems, crucial for maintaining brain health. Phytochemicals, natural compounds found in plants with antioxidant and anti-inflammatory properties, such as flavonoids, curcumin, resveratrol, and quercetin, have emerged as potential modulators of this axis, suggesting implications for AD prevention. Intake of phytochemicals influences the gut microbial composition and its metabolites, thereby impacting neuroinflammation and oxidative stress in the brain. Consumption of phytochemical-rich foods may promote a healthy gut microbiota, fostering the production of anti-inflammatory and neuroprotective substances. Early dietary incorporation of phytochemicals offers a non-invasive strategy for modulating the gut-brain axis and potentially reducing AD risk or delaying its onset. The exploration of interventions targeting the gut-brain axis through phytochemical intake represents a promising avenue for the development of preventive or therapeutic strategies against AD initiation and progression.


Subject(s)
Alzheimer Disease , Anti-Inflammatory Agents , Brain-Gut Axis , Gastrointestinal Microbiome , Neuroinflammatory Diseases , Phytochemicals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Humans , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Animals , Gastrointestinal Microbiome/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/prevention & control , Brain-Gut Axis/drug effects , Brain-Gut Axis/physiology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Brain/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Antioxidants/pharmacology , Oxidative Stress/drug effects
20.
Neuropharmacology ; 259: 110120, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39159835

ABSTRACT

A large portion of neuropathic pain suffering patients may also concurrently experience neuropathic itch, with a negative impact on the quality of life. The limited understanding of neuropathic itch and the low efficacy of current anti-itch therapies dictate the urgent need of a better comprehension of molecular mechanisms involved and development of relevant animal models. This study was aimed to characterize the itching phenotype in a model of trauma-induced peripheral neuropathy, the spared nerve injury (SNI), and the molecular events underlying the overlap with the nociceptive behavior. SNI mice developed hyperknesis and spontaneous itch 7-14 days after surgery that was prevented by gabapentin treatment. Itch was associated with pain hypersensitivity, loss of intraepidermal nerve fiber (IENF) density and increased epidermal thickness. In coincidence with the peak of scratching behavior, SNI mice showed a spinal overexpression of IBA1 and GFAP, microglia and astrocyte markers respectively. An increase of the itch neuropeptide B-type natriuretic peptide (BNP) in NeuN+ cells, of its downstream effector interleukin 17 (IL17) along with increased pERK1/2 levels occurred in the spinal cord dorsal horn and DRG. A raise in BNP and IL17 was also detected at skin level. Stimulation of HaCat cells with conditioned medium from BV2-stimulated SH-SY5Y cells produced a dramatic reduction of HaCat cell viability. This study showed that SNI mice might represent a model for neuropathic itch and pain. Collectively, our finding suggest that neuropathic itch might initiate at spinal level, then affecting skin epidermis events, through a glia-mediated neuroinflammation-evoked BNP/IL17 mechanism.


Subject(s)
Disease Models, Animal , Neuralgia , Neuroinflammatory Diseases , Pruritus , Animals , Pruritus/metabolism , Pruritus/pathology , Neuralgia/metabolism , Neuralgia/etiology , Mice , Male , Neuroinflammatory Diseases/metabolism , Humans , Gabapentin/pharmacology , Interleukin-17/metabolism , Mice, Inbred C57BL , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , HaCaT Cells , Microglia/metabolism , Microglia/drug effects , Hyperalgesia/metabolism , Microfilament Proteins/metabolism , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/drug effects , Calcium-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL