Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.733
Filter
1.
J Neuroinflammation ; 21(1): 143, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822367

ABSTRACT

The dysregulation of pro- and anti-inflammatory processes in the brain has been linked to the pathogenesis of major depressive disorder (MDD), although the precise mechanisms remain unclear. In this study, we discovered that microglial conditional knockout of Pdcd4 conferred protection against LPS-induced hyperactivation of microglia and depressive-like behavior in mice. Mechanically, microglial Pdcd4 plays a role in promoting neuroinflammatory responses triggered by LPS by inhibiting Daxx-mediated PPARγ nucleus translocation, leading to the suppression of anti-inflammatory cytokine IL-10 expression. Finally, the antidepressant effect of microglial Pdcd4 knockout under LPS-challenged conditions was abolished by intracerebroventricular injection of the IL-10 neutralizing antibody IL-10Rα. Our study elucidates the distinct involvement of microglial Pdcd4 in neuroinflammation, suggesting its potential as a therapeutic target for neuroinflammation-related depression.


Subject(s)
Co-Repressor Proteins , Interleukin-10 , Mice, Knockout , Microglia , Neuroinflammatory Diseases , PPAR gamma , Signal Transduction , Animals , Mice , Microglia/metabolism , Microglia/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Signal Transduction/physiology , Signal Transduction/drug effects , Neuroinflammatory Diseases/metabolism , Interleukin-10/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Depression/metabolism , Depression/etiology , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/deficiency , Mice, Inbred C57BL , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Male , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Lipopolysaccharides/toxicity
2.
Int J Med Sci ; 21(7): 1265-1273, 2024.
Article in English | MEDLINE | ID: mdl-38818478

ABSTRACT

This study investigated the effects of pregabalin on microglial differentiation in rats with neuropathic pain (NP) induced by sciatic nerve ligation and transection. After confirming NP, the rats were randomly allocated to either a pregabalin or control group. The pregabalin group received intraperitoneal injections of 10 mg/kg pregabalin, while the control group received an equivalent volume of normal saline following surgery. On postoperative day 28, neuronal damage, microglial activity, and microglial differentiation were assessed. The pregabalin group exhibited significantly less neuronal damage compared to the control group, along with a significant decrease in activated microglial expression in both the brain and spinal cord. Pregabalin treatment also significantly altered the microglial phenotype expression, with a decrease in the M1 phenotype percentage and an increase in the M2 phenotype percentage in both the brain (M1 phenotype: 43.52 ± 12.16% and 18.00 ± 8.57% in the control and pregabalin groups, respectively; difference: 27.26 [15.18-42.10], p = 0.002; M2 phenotype: 16.88 ± 6.47% and 39.63 ± 5.82% in the control and pregabalin groups, respectively; difference 22.04 [17.17-32.70], p < 0.001) and the spinal cord ipsilateral to nerve injury (M1 phenotype: 44.35 ± 12.12% and 13.78 ± 5.39% in the control and pregabalin groups, respectively; difference 30.46 [21.73-44.45], p < 0.001; M2 phenotype: 7.64 ± 3.91% and 33.66 ± 7.95% in the control and pregabalin groups, respectively; difference 27.41 [21.21-36.30], p < 0.001). Overall, pregabalin treatment significantly decreased the microglial M1 phenotype while increasing the microglial M2 phenotype in NP rats.


Subject(s)
Cell Differentiation , Microglia , Neuralgia , Pregabalin , Animals , Pregabalin/pharmacology , Pregabalin/therapeutic use , Microglia/drug effects , Microglia/pathology , Neuralgia/drug therapy , Neuralgia/pathology , Neuralgia/etiology , Rats , Cell Differentiation/drug effects , Male , Spinal Cord/drug effects , Spinal Cord/pathology , Disease Models, Animal , Analgesics/pharmacology , Analgesics/therapeutic use , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Rats, Sprague-Dawley , Humans , Brain/drug effects , Brain/pathology
3.
Int Immunopharmacol ; 134: 112191, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38759369

ABSTRACT

Social behavior is inextricably linked to the immune system. Although IFN-γ is known to be involved in social behavior, yet whether and how it encodes social memory remains unclear. In the current study, we injected with IFN-γ into the lateral ventricle of male C57BL/6J mice, and three-chamber social test was used to examine the effects of IFN-γ on their social preference and social memory. The morphology of microglia in the hippocampus, prelimbic cortex and amygdala was examined using immunohistochemistry, and the phenotype of microglia were examined using immunohistochemistry and enzyme-linked immunosorbent assays. The IFN-γ-injected mice were treated with lipopolysaccharide, and effects of IFN-γ on behavior and microglial responses were evaluated. STAT1 pathway and microglia-neuron interactions were examined in vivo or in vitro using western blotting and immunohistochemistry. Finally, we use STAT1 inhibitor or minocycline to evaluated the role of STAT1 in mediating the microglial priming and effects of primed microglia in IFN-γ-induced social dysfunction. We demonstrated that 500 ng of IFN-γ injection results in significant decrease in social index and social novelty recognition index, and induces microglial priming in hippocampus, characterized by enlarged cell bodies, shortened branches, increased expression of CD68, CD86, CD74, CD11b, CD11c, CD47, IL-33, IL-1ß, IL-6 and iNOS, and decreased expression of MCR1, Arg-1, IGF-1 and BDNF. This microglia subpopulation is more sensitive to LPS challenge, which characterized by more significant morphological changes and inflammatory responses, as well as induced increased sickness behaviors in mice. IFN-γ upregulated pSTAT1 and STAT1 and promoted the nuclear translocation of STAT1 in the hippocampal microglia and in the primary microglia. Giving minocycline or STAT1 inhibitor fludarabin blocked the priming of hippocampal microglia induced by IFN-γ, ameliorated the dysfunction in hippocampal microglia-neuron interactions and synapse pruning by microglia, thereby improving social memory deficits in IFN-γ injected mice. IFN-γ initiates STAT1 pathway to induce priming of hippocampal microglia, thereby disrupts hippocampal microglia-neuron interactions and neural circuit link to social memory. Blocking STAT1 pathway or inhibiting microglial priming may be strategies to reduce the effects of IFN-γ on social behavior.


Subject(s)
Hippocampus , Interferon-gamma , Mice, Inbred C57BL , Microglia , STAT1 Transcription Factor , Signal Transduction , Social Behavior , Animals , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , STAT1 Transcription Factor/metabolism , Male , Interferon-gamma/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/immunology , Mice , Signal Transduction/drug effects , Lipopolysaccharides , Memory/drug effects , Cells, Cultured , Neurons/drug effects , Neurons/immunology , Neurons/metabolism
4.
J Neuroimmune Pharmacol ; 19(1): 23, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38775885

ABSTRACT

Hyperbilirubinemia is one of the most common occurrence in newborns and is toxic to the brain, resulting in neurological sequelae such as auditory impairment, with potential to evolve to chronic bilirubin encephalopathy and long-term cognitive impairment in adults. In the early postnatal period, neurogenesis is rigorous and neuroinflammation is detrimental to the brain. What are the alterations in neurogenesis and the underlying mechanisms of bilirubin encephalopathy during the early postnatal period? This study found that, there were a reduction in the number of neuronal stem/progenitor cells, an increase in microglia in the dentate gyrus (DG) and an inflammatory state in the hippocampus, characterized by increased levels of IL-6, TNF-α, and IL-1ß, as well as a decreased level of IL-10 in a rat model of bilirubin encephalopathy (BE). Furthermore, there was a significant decrease in the number of newborn neurons and the expression of neuronal differentiation-associated genes (NeuroD and Ascl1) in the BE group. Additionally, cognitive impairment was observed in this group. The administration of minocycline, an inhibitor of microglial activation, resulted in a reduction of inflammation in the hippocampus, an enhancement of neurogenesis, an increase in the expression of neuron-related genes (NeuroD and Ascl1), and an improvement in cognitive function in the BE group. These results demonstrate that microglia play a critical role in reduced neurogenesis and impaired brain function resulting from bilirubin encephalopathy model, which could inspire the development of novel pharmaceutical and therapeutic strategies.


Subject(s)
Hippocampus , Kernicterus , Microglia , Minocycline , Neurogenesis , Animals , Neurogenesis/drug effects , Neurogenesis/physiology , Microglia/drug effects , Microglia/metabolism , Rats , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Minocycline/pharmacology , Disease Models, Animal , Rats, Sprague-Dawley , Inflammation/metabolism , Inflammation/pathology , Neuroinflammatory Diseases/drug therapy
5.
Molecules ; 29(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731397

ABSTRACT

A chemical investigation of the arils of Torreya grandis led to the isolation of seven abietane-type diterpenoids (compounds 1-7) including three previously undescribed compounds, one unreported natural product, and three known analogs. The structures of these compounds were determined by means of spectroscopy, single-crystal X-ray diffraction, and ECD spectra. An antibacterial activity assay showed that compounds 5 and 6 had significant inhibitory effects on methicillin-resistant Staphylococcus aureus, with MIC values of 100 µM. Moreover, compounds 1, 3, 4, and 7 exhibited anti-neuroinflammatory activity in LPS-stimulated BV-2 microglia cells, with the IC50 values ranging from 38.4 to 67.9 µM.


Subject(s)
Abietanes , Anti-Bacterial Agents , Abietanes/chemistry , Abietanes/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Microglia/drug effects , Microglia/metabolism , Mice , Methicillin-Resistant Staphylococcus aureus/drug effects , Animals , Molecular Structure , Cell Line , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Diterpenes/pharmacology , Diterpenes/chemistry , Diterpenes/isolation & purification , Lipopolysaccharides/pharmacology
6.
Neuroreport ; 35(9): 549-557, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38739900

ABSTRACT

Neuroinflammation after traumatic brain injury (TBI) exhibits a strong correlation with neurological impairment, which is a crucial target for improving the prognosis of TBI patients. The involvement of CXCL5/CXCR2 signaling in the regulation of neuroinflammation in brain injury models has been documented. Therefore, the effects of CXCL5 on post-TBI neuroinflammation and its potential mechanisms need to be explored. Following TBI, C57BL/6 mice were administered intraperitoneal injections of a CXCL5 neutralizing antibody (Nab-CXCL5) (5 mg/kg, 2 times/day). Subsequently, the effects on neuroinflammation, nerve injury, and neurological function were assessed. Nab-CXCL5 significantly reduced the release of inflammatory factors, inhibited the formation of inflammatory microglia and astrocytes, and reduced the infiltration of peripheral immune cells in TBI mice. Additionally, this intervention led to a reduction in neuronal impairment and facilitated the restoration of sensorimotor abilities, as well as improvements in learning and memory functions. Peripheral administration of the Nab-CXCL5 to TBI mice could suppress neuroinflammation, reduce neurological damage, and improve neurological function. Our data suggest that neutralizing antibodies against CXCL5 (Nab-CXCL5) may be a promising agent for treating TBI.


Subject(s)
Brain Injuries, Traumatic , Chemokine CXCL5 , Mice, Inbred C57BL , Neuroinflammatory Diseases , Recovery of Function , Animals , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/drug therapy , Chemokine CXCL5/metabolism , Neuroinflammatory Diseases/drug therapy , Mice , Male , Recovery of Function/drug effects , Recovery of Function/physiology , Antibodies, Neutralizing/pharmacology , Microglia/drug effects , Microglia/metabolism
7.
J Neuroimmune Pharmacol ; 19(1): 20, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758335

ABSTRACT

Neuroinflammation has emerged as a crucial factor in the development of depression. Despite the well-known anti-inflammatory properties of 6-gingerol, its potential impact on depression remains poorly understood. This study aimed to investigate the antidepressant effects of 6-gingerol by suppressing microglial activation. In vivo experiments were conducted to evaluate the effect of 6-gingerol on lipopolysaccharide (LPS)-induced behavioral changes and neuroinflammation in rat models. In vitro studies were performed to examine the neuroprotective properties of 6-gingerol against LPS-induced microglial activation. Furthermore, a co-culture system of microglia and neurons was established to assess the influence of 6-gingerol on the expression of synaptic-related proteins, namely synaptophysin (SYP) and postsynaptic density protein 95 (PSD95), which are influenced by microglial activation. In the in vivo experiments, administration of 6-gingerol effectively alleviated LPS-induced depressive behavior in rats. Moreover, it markedly suppressed the activation of rat prefrontal cortex (PFC) microglia induced by LPS and the activation of the NF-κB/NLRP3 inflammatory pathway, while also reducing the levels of inflammatory cytokines IL-1ß and IL-18. In the in vitro experiments, 6-gingerol mitigated nuclear translocation of NF-κB p65, NLRP3 activation, and maturation of IL-1ß and IL-18, all of which were induced by LPS. Furthermore, in the co-culture system of microglia and neurons, 6-gingerol effectively restored the decreased expression of SYP and PSD95. The findings of this study demonstrate the neuroprotective effects of 6-gingerol in the context of LPS-induced depression-like behavior. These effects are attributed to the inhibition of microglial hyperactivation through the suppression of the NF-κB/NLRP3 inflammatory pathway.


Subject(s)
Catechols , Depression , Fatty Alcohols , Lipopolysaccharides , Microglia , Neuronal Plasticity , Rats, Sprague-Dawley , Animals , Fatty Alcohols/pharmacology , Microglia/drug effects , Microglia/metabolism , Rats , Lipopolysaccharides/toxicity , Male , Catechols/pharmacology , Neuronal Plasticity/drug effects , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Coculture Techniques , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Disease Models, Animal , Neuroprotective Agents/pharmacology , Cells, Cultured , Antidepressive Agents/pharmacology
8.
CNS Neurosci Ther ; 30(5): e14736, 2024 05.
Article in English | MEDLINE | ID: mdl-38739106

ABSTRACT

AIMS: Multiple sclerosis (MS) is a neuroinflammatory demyelinating disease. Microglia are reportedly involved in the pathogenesis of MS. However, the key molecules that control the inflammatory activity of microglia in MS have not been identified. METHODS: Experimental autoimmune encephalomyelitis (EAE) mice were randomized into CD22 blockade and control groups. The expression levels of microglial CD22 were measured by flow cytometry, qRT-PCR, and immunofluorescence. The effects of CD22 blockade were examined via in vitro and in vivo studies. RESULTS: We detected increased expression of microglial CD22 in EAE mice. In addition, an in vitro study revealed that lipopolysaccharide upregulated the expression of CD22 in microglia and that CD22 blockade modulated microglial polarization. Moreover, an in vivo study demonstrated that CD22 blockade aggravated EAE in mice and promoted microglial M1 polarization. CONCLUSION: Collectively, our study indicates that CD22 may be protective against EAE and may play a critical role in the maintenance of immune homeostasis in EAE mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Microglia , Sialic Acid Binding Ig-like Lectin 2 , Animals , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Microglia/drug effects , Microglia/metabolism , Mice , Female , Cell Polarity/drug effects , Cell Polarity/physiology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/toxicity , Cells, Cultured , Myelin-Oligodendrocyte Glycoprotein/toxicity , Myelin-Oligodendrocyte Glycoprotein/immunology
9.
Cells ; 13(9)2024 May 04.
Article in English | MEDLINE | ID: mdl-38727321

ABSTRACT

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by deficiency of the survival motor neuron (SMN) protein. Although SMA is a genetic disease, environmental factors contribute to disease progression. Common pathogen components such as lipopolysaccharides (LPS) are considered significant contributors to inflammation and have been associated with muscle atrophy, which is considered a hallmark of SMA. In this study, we used the SMNΔ7 experimental mouse model of SMA to scrutinize the effect of systemic LPS administration, a strong pro-inflammatory stimulus, on disease outcome. Systemic LPS administration promoted a reduction in SMN expression levels in CNS, peripheral lymphoid organs, and skeletal muscles. Moreover, peripheral tissues were more vulnerable to LPS-induced damage compared to CNS tissues. Furthermore, systemic LPS administration resulted in a profound increase in microglia and astrocytes with reactive phenotypes in the CNS of SMNΔ7 mice. In conclusion, we hereby show for the first time that systemic LPS administration, although it may not precipitate alterations in terms of deficits of motor functions in a mouse model of SMA, it may, however, lead to a reduction in the SMN protein expression levels in the skeletal muscles and the CNS, thus promoting synapse damage and glial cells' reactive phenotype.


Subject(s)
Disease Models, Animal , Lipopolysaccharides , Muscular Atrophy, Spinal , Animals , Lipopolysaccharides/pharmacology , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/metabolism , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 1 Protein/genetics , Mice, Inbred C57BL , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Inflammation/pathology
10.
Cell Mol Life Sci ; 81(1): 215, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38739166

ABSTRACT

Down syndrome (DS) is a genetic disease characterized by a supernumerary chromosome 21. Intellectual deficiency (ID) is one of the most prominent features of DS. Central nervous system defects lead to learning disabilities, motor and language delays, and memory impairments. At present, a prenatal treatment for the ID in DS is lacking. Subcutaneous administration of synthetic preimplantation factor (sPIF, a peptide with a range of biological functions) in a model of severe brain damage has shown neuroprotective and anti-inflammatory properties by directly targeting neurons and microglia. Here, we evaluated the effect of PIF administration during gestation and until weaning on Dp(16)1Yey mice (a mouse model of DS). Possible effects at the juvenile stage were assessed using behavioral tests and molecular and histological analyses of the brain. To test the influence of perinatal sPIF treatment at the adult stage, hippocampus-dependent memory was evaluated on postnatal day 90. Dp(16)1Yey pups showed significant behavioral impairment, with impaired neurogenesis, microglial cell activation and a low microglial cell count, and the deregulated expression of genes linked to neuroinflammation and cell cycle regulation. Treatment with sPIF restored early postnatal hippocampal neurogenesis, with beneficial effects on astrocytes, microglia, inflammation, and cell cycle markers. Moreover, treatment with sPIF restored the level of DYRK1A, a protein that is involved in cognitive impairments in DS. In line with the beneficial effects on neurogenesis, perinatal treatment with sPIF was associated with an improvement in working memory in adult Dp(16)1Yey mice. Perinatal treatment with sPIF might be an option for mitigating cognitive impairments in people with DS.


Subject(s)
Disease Models, Animal , Down Syndrome , Neurogenesis , Animals , Down Syndrome/drug therapy , Down Syndrome/pathology , Down Syndrome/metabolism , Down Syndrome/complications , Down Syndrome/genetics , Neurogenesis/drug effects , Mice , Female , Pregnancy , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/drug effects , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Dyrk Kinases , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Male , Cognition Disorders/drug therapy , Cognition Disorders/pathology
11.
Mol Med ; 30(1): 59, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745316

ABSTRACT

Microglial activation and polarization play a central role in poststroke inflammation and neuronal damage. Modulating microglial polarization from pro-inflammatory to anti-inflammatory phenotype is a promising therapeutic strategy for the treatment of cerebral ischemia. Polyphyllin I (PPI), a steroidal saponin, shows multiple bioactivities in various diseases, but the potential function of PPI in cerebral ischemia is not elucidated yet. In our study, the influence of PPI on cerebral ischemia-reperfusion injury was evaluated. Mouse middle cerebral artery occlusion (MCAO) model and oxygen-glucose deprivation and reoxygenation (OGD/R) model were constructed to mimic cerebral ischemia-reperfusion injury in vivo and in vitro. TTC staining, TUNEL staining, RT-qPCR, ELISA, flow cytometry, western blot, immunofluorescence, hanging wire test, rotarod test and foot-fault test, open-field test and Morris water maze test were performed in our study. We found that PPI alleviated cerebral ischemia-reperfusion injury and neuroinflammation, and improved functional recovery of mice after MCAO. PPI modulated microglial polarization towards anti-inflammatory M2 phenotype in MCAO mice in vivo and post OGD/R in vitro. Besides, PPI promoted autophagy via suppressing Akt/mTOR signaling in microglia, while inhibition of autophagy abrogated the effect of PPI on M2 microglial polarization after OGD/R. Furthermore, PPI facilitated autophagy-mediated ROS clearance to inhibit NLRP3 inflammasome activation in microglia, and NLRP3 inflammasome reactivation by nigericin abolished the effect of PPI on M2 microglia polarization. In conclusion, PPI alleviated post-stroke neuroinflammation and tissue damage via increasing autophagy-mediated M2 microglial polarization. Our data suggested that PPI had potential for ischemic stroke treatment.


Subject(s)
Autophagy , Disease Models, Animal , Microglia , Neuroinflammatory Diseases , Reperfusion Injury , Animals , Microglia/drug effects , Microglia/metabolism , Mice , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/etiology , Autophagy/drug effects , Male , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Diosgenin/analogs & derivatives , Diosgenin/pharmacology , Diosgenin/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Signal Transduction/drug effects , Infarction, Middle Cerebral Artery/drug therapy , TOR Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL , Cell Polarity/drug effects
12.
J Neuroimmune Pharmacol ; 19(1): 17, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717643

ABSTRACT

In our previous study, we concluded that sirtuin 5 (SIRT5) was highly expressed in microglia following ischaemic stroke, which induced excessive neuroinflammation and neuronal injury. Therefore, SIRT5-targeting interventions should reduce neuroinflammation and protect against ischaemic brain injury. Here, we showed that treatment with a specific SIRT5 inhibitor, MC3482, alleviated microglia-induced neuroinflammation and improved long-term neurological function in a mouse model of stroke. The mice were administrated with either vehicle or 2 mg/kg MC3482 daily for 7 days via lateral ventricular injection following the onset of middle cerebral artery occlusion. The outcome was assessed by a panel of tests, including a neurological outcome score, declarative memory, sensorimotor tests, anxiety-like behavior and a series of inflammatory factors. We observed a significant reduction of infarct size and inflammatory factors, and the improvement of long-term neurological function in the early stages during ischaemic stroke when the mice were treated with MC3482. Mechanistically, the administration of MC3482 suppressed the desuccinylation of annexin-A1, thereby promoting its membrane recruitment and extracellular secretion, which in turn alleviated neuroinflammation during ischaemic stroke. Based on our findings, MC3482 offers promise as an anti-ischaemic stroke treatment that targets directly the disease's underlying factors.


Subject(s)
Annexin A1 , Ischemic Stroke , Mice, Inbred C57BL , Microglia , Neuroinflammatory Diseases , Up-Regulation , Animals , Mice , Microglia/drug effects , Microglia/metabolism , Male , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Annexin A1/metabolism , Up-Regulation/drug effects , Sirtuins/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism
13.
Front Immunol ; 15: 1366841, 2024.
Article in English | MEDLINE | ID: mdl-38711521

ABSTRACT

Introduction: Age-related macular degeneration (AMD) is a prevalent, chronic and progressive retinal degenerative disease characterized by an inflammatory response mediated by activated microglia accumulating in the retina. In this study, we demonstrate the therapeutically effects and the underlying mechanisms of microglial repopulation in the laser-induced choroidal neovascularization (CNV) model of exudative AMD. Methods: The CSF1R inhibitor PLX3397 was used to establish a treatment paradigm for microglial repopulation in the retina. Neovascular leakage and neovascular area were examined by fundus fluorescein angiography (FFA) and immunostaining of whole-mount RPE-choroid-sclera complexes in CNV mice receiving PLX3397. Altered cellular senescence was measured by beta-galactosidase (SA-ß-gal) activity and p16INK4a expression. The effect and mechanisms of repopulated microglia on leukocyte infiltration and the inflammatory response in CNV lesions were analyzed. Results: We showed that ten days of the CSF1R inhibitor PLX3397 treatment followed by 11 days of drug withdrawal was sufficient to stimulate rapid repopulation of the retina with new microglia. Microglial repopulation attenuated pathological choroid neovascularization and dampened cellular senescence in CNV lesions. Repopulating microglia exhibited lower levels of activation markers, enhanced phagocytic function and produced fewer cytokines involved in the immune response, thereby ameliorating leukocyte infiltration and attenuating the inflammatory response in CNV lesions. Discussion: The microglial repopulation described herein are therefore a promising strategy for restricting inflammation and choroidal neovascularization, which are important players in the pathophysiology of AMD.


Subject(s)
Aminopyridines , Choroidal Neovascularization , Disease Models, Animal , Microglia , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Microglia/metabolism , Microglia/drug effects , Mice , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Mice, Inbred C57BL , Macular Degeneration/pathology , Macular Degeneration/metabolism , Macular Degeneration/drug therapy , Inflammation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Pyrroles/pharmacology , Pyrroles/therapeutic use , Cellular Senescence/drug effects
14.
Discov Med ; 36(184): 936-945, 2024 May.
Article in English | MEDLINE | ID: mdl-38798253

ABSTRACT

BACKGROUND: Inflammation is a key pathological process in bacterial meningitis, and the transforming growth factor-beta-activated kinase 1 (TAK1)/nuclear factor-kappa B (NF-κB) pathway is implicated in the activation of microglia and the production of inflammatory factors. Interleukin (IL)-10 is an anti-inflammatory cytokine acting in an autocrine fashion in macrophages to limit inflammatory responses by decreasing the production of pro-inflammatory cytokines. This paper investigates how IL-10 can inhibit microglia activation and reduce the inflammatory response of nervous system diseases. METHODS: This study used a pneumococcal-induced in Pneumococcal meningitis (PM) C57BL/6 mice and BV-2 cells model of microglial activation, assessing the effects of IL-10 on the TAK1/NF-κB pathway. The impact of IL-10 on microglial autophagy was investigated through western blot and immunofluorescence. The effects of IL-10 were evaluated by examining cellular activation markers and the activity of molecular signaling pathways (such as phosphorylation levels of TAK1 and NF-κB). RESULTS: Pneumococcus induced the activation of microglia and reduced IL-10. IL-10 inhibited the TAK1/NF-κB pathway, reducing the pneumococcal-induced inflammatory response in microglia. IL-10 ameliorated pneumococcal infection-induced microglial injury by inhibiting autophagy. Animal experiment results also showed that IL-10 inhibited inflammation and autophagy during Pneumococcal meningitis in mice. CONCLUSION: Our study demonstrates that IL-10 reduces the inflammatory response of microglia by inhibiting the TAK1/NF-κB pathway. Additionally, IL-10 ameliorates pneumococcal infection-induced microglial injury by inhibiting the process of autophagy. These results provide a new theoretical basis and offer new insights for developing strategies to treat bacterial meningitis.


Subject(s)
Interleukin-10 , MAP Kinase Kinase Kinases , Meningitis, Pneumococcal , Mice, Inbred C57BL , Microglia , NF-kappa B , Animals , Interleukin-10/metabolism , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Mice , Meningitis, Pneumococcal/drug therapy , Meningitis, Pneumococcal/immunology , Meningitis, Pneumococcal/pathology , NF-kappa B/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Inflammation/pathology , Autophagy/drug effects , Disease Models, Animal , Cell Line , Streptococcus pneumoniae
15.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791517

ABSTRACT

Maternal immune activation (MIA) is a risk factor for multiple neurodevelopmental disorders; however, animal models developed to explore MIA mechanisms are sensitive to experimental factors, which has led to complexity in previous reports of the MIA phenotype. We sought to characterize an MIA protocol throughout development to understand how prenatal immune insult alters the trajectory of important neurodevelopmental processes, including the microglial regulation of synaptic spines and complement signaling. We used polyinosinic:polycytidylic acid (polyI:C) to induce MIA on gestational day 9.5 in CD-1 mice, and measured their synaptic spine density, microglial synaptic pruning, and complement protein expression. We found reduced dendritic spine density in the somatosensory cortex starting at 3-weeks-of-age with requisite increases in microglial synaptic pruning and phagocytosis, suggesting spine density loss was caused by increased microglial synaptic pruning. Additionally, we showed dysregulation in complement protein expression persisting into adulthood. Our findings highlight disruptions in the prenatal environment leading to alterations in multiple dynamic processes through to postnatal development. This could potentially suggest developmental time points during which synaptic processes could be measured as risk factors or targeted with therapeutics for neurodevelopmental disorders.


Subject(s)
Complement System Proteins , Dendritic Spines , Microglia , Poly I-C , Animals , Microglia/metabolism , Microglia/drug effects , Microglia/immunology , Mice , Female , Pregnancy , Dendritic Spines/metabolism , Poly I-C/pharmacology , Complement System Proteins/metabolism , Complement System Proteins/immunology , Prenatal Exposure Delayed Effects , Phagocytosis , Disease Models, Animal , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Synapses/metabolism , Synapses/drug effects , Neuronal Plasticity/drug effects
16.
Transl Psychiatry ; 14(1): 209, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796504

ABSTRACT

Stress affects the brain and alters its neuroarchitecture and function; these changes can be severe and lead to psychiatric disorders. Recent evidence suggests that astrocytes and microglia play an essential role in the stress response by contributing to the maintenance of cerebral homeostasis. These cells respond rapidly to all stimuli that reach the brain, including stressors. Here, we used a recently validated rodent model of post-traumatic stress disorder in which rats can be categorized as resilient or vulnerable after acute inescapable footshock stress. We then investigated the functional, molecular, and morphological determinants of stress resilience and vulnerability in the prefrontal cortex, focusing on glial and neuronal cells. In addition, we examined the effects of a single subanesthetic dose of ketamine, a fast-acting antidepressant recently approved for the treatment of resistant depression and proposed for other stress-related psychiatric disorders. The present results suggest a prompt glial cell response and activation of the NF-κB pathway after acute stress, leading to an increase in specific cytokines such as IL-18 and TNF-α. This response persists in vulnerable individuals and is accompanied by a significant change in the levels of critical glial proteins such as S100B, CD11b, and CX43, brain trophic factors such as BDNF and FGF2, and proteins related to dendritic arborization and synaptic architecture such as MAP2 and PSD95. Administration of ketamine 24 h after the acute stress event rescued many of the changes observed in vulnerable rats, possibly contributing to support brain homeostasis. Overall, our results suggest that pivotal events, including reactive astrogliosis, changes in brain trophic factors, and neuronal damage are critical determinants of vulnerability to acute traumatic stress and confirm the therapeutic effect of acute ketamine against the development of stress-related psychiatric disorders.


Subject(s)
Astrocytes , Disease Models, Animal , Ketamine , Microglia , Stress Disorders, Post-Traumatic , Animals , Ketamine/pharmacology , Ketamine/administration & dosage , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Microglia/drug effects , Microglia/metabolism , Male , Rats , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Stress, Psychological/metabolism , Rats, Sprague-Dawley , NF-kappa B/metabolism
17.
Phytochemistry ; 223: 114121, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38697242

ABSTRACT

In this study, twenty-three ent-eudesmane sesquiterpenoids (1-23) including fifteen previously undescribed ones, named eutypelides A-O (1-15) were isolated from the marine-derived fungus Eutypella sp. F0219. Their planar structures and relative configurations were established by HR-ESIMS and extensive 1D and 2D NMR investigations. The absolute configurations of the previously undescribed compounds were determined by single-crystal X-ray diffraction analyses, modified Mosher's method, and ECD calculations. Structurally, eutypelide A (1) is a rare 1,10-seco-ent-eudesmane, whereas 2-15 are typically ent-eudesmanes with 6/6/-fused bicyclic carbon nucleus. The anti-neuroinflammatory activity of all isolated compounds (1-23) was accessed based on their ability to NO production in LPS-stimulated BV2 microglia cells. Compound 16 emerged as the most potent inhibitor. Further mechanistic investigation revealed that compound 16 modulated the inflammatory response by decreasing the protein levels of iNOS and increasing ARG 1 levels, thereby altering the iNOS/ARG 1 ratio and inhibiting macrophage polarization. qRT-PCR analysis showed that compound 16 reversed the LPS-induced upregulation of pro-inflammatory cytokines, including iNOS, TNF-α, IL-6, and IL-1ß, at both the transcriptional and translational levels. These effects were linked to the inhibition of the NF-κB pathway, a key regulator of inflammation. Our findings suggest that compound 16 may be a potential structure basis for developing neuroinflammation-related disease therapeutic agents.


Subject(s)
Anti-Inflammatory Agents , Lipopolysaccharides , Microglia , Sesquiterpenes, Eudesmane , Animals , Mice , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Sesquiterpenes, Eudesmane/pharmacology , Sesquiterpenes, Eudesmane/chemistry , Sesquiterpenes, Eudesmane/isolation & purification , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Microglia/drug effects , Molecular Structure , Nitric Oxide/biosynthesis , Nitric Oxide/antagonists & inhibitors , Structure-Activity Relationship , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Dose-Response Relationship, Drug , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification
18.
Free Radic Biol Med ; 220: 56-66, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38697489

ABSTRACT

Apart from dopaminergic neurotoxicity, exposure to rotenone, a commonly used insecticide in agriculture, also adversely affects hippocampal and cortical neurons, resulting in cognitive impairments in mice. We recently established a role of microglia-mediated neuroinflammation in rotenone-elicited deficits of cognition, yet the mechanisms remain elusive. Here, we investigated the involvement of NADPH oxidase 2 (NOX2) catalytic subunit gp91phox in rotenone-induced cognitive deficits and the associated mechanisms. Our study demonstrated that rotenone exposure elevated expression of gp91phox and phosphorylation of the NOX2 cytosolic subunit p47phox, along with NADPH depletion in the hippocampus and cortex of mice, indicating NOX2 activation. Specific knockdown of gp91phox in microglia via adeno-associated virus delivery resulted in reduced microglial activation, proinflammatory gene expression and improved learning and memory capacity in rotenone-intoxicated mice. Genetic deletion of gp91phox also reversed rotenone-elicited cognitive dysfunction in mice. Furthermore, microglial gp91phox knockdown attenuated neuronal damage and synaptic loss in mice. This intervention also suppressed iron accumulation, disruption of iron-metabolism proteins and iron-dependent lipid peroxidation and restored the balance of ferroptosis-related parameters, including GPX4, SLC711, PTGS2, and ACSL4 in rotenone-lesioned mice. Intriguingly, pharmacological inhibition of ferroptosis with liproxstatin-1 conferred protection against rotenone-induced neurodegeneration and cognitive dysfunction in mice. In summary, our findings underscored the contribution of microglial gp91phox-dependent neuroinflammation and ferroptosis to learning and memory dysfunction in rotenone-lesioned mice. These results provided valuable insights into the pathogenesis of cognitive deficits associated with pesticide-induced Parkinsonism, suggesting potential therapeutic avenues for intervention.


Subject(s)
Ferroptosis , Memory Disorders , Microglia , NADPH Oxidase 2 , Neuroinflammatory Diseases , Rotenone , Animals , Mice , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Rotenone/toxicity , Ferroptosis/drug effects , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/genetics , Memory Disorders/chemically induced , Memory Disorders/metabolism , Memory Disorders/genetics , Memory Disorders/pathology , Male , Mice, Inbred C57BL , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Mice, Knockout
19.
Biomolecules ; 14(5)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38785932

ABSTRACT

Augmenting the natural melanocortin pathway in mouse eyes with uveitis or diabetes protects the retinas from degeneration. The retinal cells are protected from oxidative and apoptotic signals of death. Therefore, we investigated the effects of a therapeutic application of the melanocortin alpha-melanocyte-stimulating hormone (α-MSH) on an ischemia and reperfusion (I/R) model of retinal degenerative disease. Eyes were subjected to an I/R procedure and were treated with α-MSH. Retinal sections were histopathologically scored. Also, the retinal sections were immunostained for viable ganglion cells, activated Muller cells, microglial cells, and apoptosis. The I/R caused retinal deformation and ganglion cell loss that was significantly reduced in I/R eyes treated with α-MSH. While α-MSH treatment marginally reduced the number of GFAP-positive Muller cells, it significantly suppressed the density of Iba1-positive microglial cells in the I/R retinas. Within one hour after I/R, there was apoptosis in the ganglion cell layer, and by 48 h, there was apoptosis in all layers of the neuroretina. The α-MSH treatment significantly reduced and delayed the onset of apoptosis in the retinas of I/R eyes. The results demonstrate that therapeutically augmenting the melanocortin pathways preserves retinal structure and cell survival in eyes with progressive neuroretinal degenerative disease.


Subject(s)
Apoptosis , Homeostasis , Reperfusion Injury , Retina , Retinal Ganglion Cells , alpha-MSH , Animals , alpha-MSH/pharmacology , alpha-MSH/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Mice , Apoptosis/drug effects , Retina/metabolism , Retina/drug effects , Retina/pathology , Homeostasis/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Mice, Inbred C57BL , Microglia/metabolism , Microglia/drug effects , Male , Ependymoglial Cells/metabolism , Ependymoglial Cells/drug effects , Ependymoglial Cells/pathology , Disease Models, Animal , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/drug therapy
20.
Biomolecules ; 14(5)2024 May 10.
Article in English | MEDLINE | ID: mdl-38785974

ABSTRACT

Diabetic retinopathy (DR) affects over 140 million people globally. The mechanisms that lead to blindness are still enigmatic but there is evidence that sustained inflammation and hypoxia contribute to vascular damage. Despite efforts to understand the role of inflammation and microglia in DR's pathology, the contribution of astrocytes to hypoxic responses is less clear. To investigate the role of astrocytes in hypoxia-induced retinopathy, we utilized a 7-day systemic hypoxia model using the GFAP-CreERT2:Rosa26iDTR transgenic mouse line. This allows for the induction of inflammatory reactive astrogliosis following tamoxifen and diphtheria toxin administration. We hypothesize that DTx-induced astrogliosis is neuroprotective during hypoxia-induced retinopathy. Glial, neuronal, and vascular responses were quantified using immunostaining, with antibodies against GFAP, vimentin, IBA-1, NeuN, fibrinogen, and CD31. Cytokine responses were measured in both the brain and serum. We report that while both DTx and hypoxia induced a phenotype of reduced microglia morphological activation, DTx, but not hypoxia, induced an increase in the Müller glia marker vimentin. We did not observe that the combination of DTx and hypoxic treatments exacerbated the signs of reactive glial cells, nor did we observe a significant change in the expression immunomodulatory mediators IL-1ß, IL2, IL-4, IL-5, IL-6, IL-10, IL-18, CCL17, TGF-ß1, GM-CSF, TNF-α, and IFN-γ. Overall, our results suggest that, in this hypoxia model, reactive astrogliosis does not alter the inflammatory responses or cause vascular damage in the retina.


Subject(s)
Disease Models, Animal , Ependymoglial Cells , Gliosis , Mice, Transgenic , Microglia , Animals , Gliosis/pathology , Gliosis/metabolism , Gliosis/chemically induced , Mice , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Ependymoglial Cells/drug effects , Retina/metabolism , Retina/pathology , Retina/drug effects , Hypoxia/metabolism , Hypoxia/pathology , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/drug effects , Glial Fibrillary Acidic Protein/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Cytokines/metabolism , Vimentin/metabolism , Vimentin/genetics , Diphtheria Toxin
SELECTION OF CITATIONS
SEARCH DETAIL
...