Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 382
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.
J Neuroinflammation ; 21(1): 145, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824526

ABSTRACT

BACKGROUND: Recent experimental studies of neuroinflammation in glaucoma pointed to cFLIP as a molecular switch for cell fate decisions, mainly regulating cell type-specific caspase-8 functions in cell death and inflammation. This study aimed to determine the importance of cFLIP for regulating astroglia-driven neuroinflammation in experimental glaucoma by analyzing the outcomes of astroglia-targeted transgenic deletion of cFLIP or cFLIPL. METHODS: Glaucoma was modeled by anterior chamber microbead injections to induce ocular hypertension in mouse lines with or without conditional deletion of cFLIP or cFLIPL in astroglia. Morphological analysis of astroglia responses assessed quantitative parameters in retinal whole mounts immunolabeled for GFAP and inflammatory molecules or assayed for TUNEL. The molecular analysis included 36-plexed immunoassays of the retina and optic nerve cytokines and chemokines, NanoString-based profiling of inflammation-related gene expression, and Western blot analysis of selected proteins in freshly isolated samples of astroglia. RESULTS: Immunoassays and immunolabeling of retina and optic nerve tissues presented reduced production of various proinflammatory cytokines, including TNFα, in GFAP/cFLIP and GFAP/cFLIPL relative to controls at 12 weeks of ocular hypertension with no detectable alteration in TUNEL. Besides presenting a similar trend of the proinflammatory versus anti-inflammatory molecules displayed by immunoassays, NanoString-based molecular profiling detected downregulated NF-κB/RelA and upregulated RelB expression of astroglia in ocular hypertensive samples of GFAP/cFLIP compared to ocular hypertensive controls. Analysis of protein expression also revealed decreased phospho-RelA and increased phospho-RelB in parallel with an increase in caspase-8 cleavage products. CONCLUSIONS: A prominent response limiting neuroinflammation in ocular hypertensive eyes with cFLIP-deletion in astroglia values the role of cFLIP in the molecular regulation of glia-driven neuroinflammation during glaucomatous neurodegeneration. The molecular responses accompanying the lessening of neurodegenerative inflammation also seem to maintain astroglia survival despite increased caspase-8 cleavage with cFLIP deletion. A transcriptional autoregulatory response, dampening RelA but boosting RelB for selective expression of NF-κB target genes, might reinforce cell survival in cFLIP-deleted astroglia.


Subject(s)
Astrocytes , CASP8 and FADD-Like Apoptosis Regulating Protein , Glaucoma , Neuroinflammatory Diseases , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Mice , Astrocytes/metabolism , Astrocytes/pathology , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/genetics , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice, Transgenic , Disease Models, Animal , Cytokines/metabolism , Retina/metabolism , Retina/pathology , Mice, Inbred C57BL , Optic Nerve/pathology , Optic Nerve/metabolism , Glial Fibrillary Acidic Protein/metabolism
3.
Int J Mol Sci ; 25(9)2024 May 06.
Article in English | MEDLINE | ID: mdl-38732259

ABSTRACT

Neuroinflammation, a hallmark of various central nervous system disorders, is often associated with oxidative stress and neuronal or oligodendrocyte cell death. It is therefore very interesting to target neuroinflammation pharmacologically. One therapeutic option is the use of nutraceuticals, particularly apigenin. Apigenin is present in plants: vegetables (parsley, celery, onions), fruits (oranges), herbs (chamomile, thyme, oregano, basil), and some beverages (tea, beer, and wine). This review explores the potential of apigenin as an anti-inflammatory agent across diverse neurological conditions (multiple sclerosis, Parkinson's disease, Alzheimer's disease), cancer, cardiovascular diseases, cognitive and memory disorders, and toxicity related to trace metals and other chemicals. Drawing upon major studies, we summarize apigenin's multifaceted effects and underlying mechanisms in neuroinflammation. Our review underscores apigenin's therapeutic promise and calls for further investigation into its clinical applications.


Subject(s)
Anti-Inflammatory Agents , Apigenin , Neuroinflammatory Diseases , Apigenin/pharmacology , Apigenin/therapeutic use , Humans , Animals , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Oxidative Stress/drug effects , Inflammation/drug therapy , Inflammation/metabolism
4.
PLoS One ; 19(5): e0303150, 2024.
Article in English | MEDLINE | ID: mdl-38728304

ABSTRACT

The Ang-(1-7)/MasR axis is critically involved in treating several diseases; For example, Ang-(1-7) improves inflammatory response and neurological function after traumatic brain injury and inhibits post-inflammatory hypothermia. However, its function in traumatic brain injury (TBI) combined with seawater immersion hypothermia remains unclear. Here, we used a mice model of hypothermic TBI and a BV2 cell model of hypothermic inflammation to investigate whether the Ang-(1-7)/MasR axis is involved in ameliorating hypothermic TBI. Quantitative reverse transcription PCR, western blotting assay, and immunofluorescence assay were performed to confirm microglia polarization and cytokine regulation. Hematoxylin-eosin staining, Nissl staining, and immunohistochemical assay were conducted to assess the extent of hypothermic TBI-induced damage and the ameliorative effect of Ang-(1-7) in mice. An open field experiment and neurological function scoring with two approaches were used to assess the degree of recovery and prognosis in mice. After hypothermic TBI establishment in BV2 cells, the Ang-(1-7)/MasR axis induced phenotypic transformation of microglia from M1 to M2, inhibited IL-6 and IL-1ß release, and upregulated IL-4 and IL-10 levels. After hypothermic TBI development in mice, intraperitoneally administered Ang-(1-7) attenuated histological damage and promoted neurological recovery. These findings suggest that hypothermia exacerbates TBI-induced damage and that the Ang-(1-7)/MasR axis can ameliorate hypothermic TBI and directly affect prognosis.


Subject(s)
Angiotensin I , Brain Injuries, Traumatic , Microglia , Neuroinflammatory Diseases , Peptide Fragments , Animals , Microglia/metabolism , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Mice , Male , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism , Phenotype , Disease Models, Animal , Hypothermia, Induced , Cytokines/metabolism , Cell Line , Hypothermia/metabolism , Inflammation/pathology , Inflammation/metabolism
5.
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
6.
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
7.
J Neuroinflammation ; 21(1): 117, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715127

ABSTRACT

BACKGROUND: Despite the high prevalence of neuropathic pain, treating this neurological disease remains challenging, given the limited efficacy and numerous side effects associated with current therapies. The complexity in patient management is largely attributed to an incomplete understanding of the underlying pathological mechanisms. Central sensitization, that refers to the adaptation of the central nervous system to persistent inflammation and heightened excitatory transmission within pain pathways, stands as a significant contributor to persistent pain. Considering the role of the cystine/glutamate exchanger (also designated as system xc-) in modulating glutamate transmission and in supporting neuroinflammatory responses, we investigated the contribution of this exchanger in the development of neuropathic pain. METHODS: We examined the implication of system xc- by evaluating changes in the expression/activity of this exchanger in the dorsal spinal cord of mice after unilateral partial sciatic nerve ligation. In this surgical model of neuropathic pain, we also examined the consequence of the genetic suppression of system xc- (using mice lacking the system xc- specific subunit xCT) or its pharmacological manipulation (using the pharmacological inhibitor sulfasalazine) on the pain-associated behavioral responses. Finally, we assessed the glial activation and the inflammatory response in the spinal cord by measuring mRNA and protein levels of GFAP and selected M1 and M2 microglial markers. RESULTS: The sciatic nerve lesion was found to upregulate system xc- at the spinal level. The genetic deletion of xCT attenuated both the amplitude and the duration of the pain sensitization after nerve surgery, as evidenced by reduced responses to mechanical and thermal stimuli, and this was accompanied by reduced glial activation. Consistently, pharmacological inhibition of system xc- had an analgesic effect in lesioned mice. CONCLUSION: Together, these observations provide evidence for a role of system xc- in the biochemical processes underlying central sensitization. We propose that the reduced hypersensitivity observed in the transgenic mice lacking xCT or in sulfasalazine-treated mice is mediated by a reduced gliosis in the lumbar spinal cord and/or a shift in microglial M1/M2 polarization towards an anti-inflammatory phenotype in the absence of system xc-. These findings suggest that drugs targeting system xc- could contribute to prevent or reduce neuropathic pain.


Subject(s)
Amino Acid Transport System y+ , Mice, Inbred C57BL , Neuralgia , Neuroinflammatory Diseases , Spinal Cord , Animals , Mice , Neuralgia/metabolism , Neuroinflammatory Diseases/metabolism , Male , Spinal Cord/metabolism , Spinal Cord/pathology , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Disease Models, Animal , Mice, Knockout , Sulfasalazine/pharmacology , Sulfasalazine/therapeutic use , Hyperalgesia/metabolism , Hyperalgesia/etiology , Mice, Transgenic
8.
J Neuroinflammation ; 21(1): 115, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698374

ABSTRACT

BACKGROUND: Macrophages play a pivotal role in the regulation of Japanese encephalitis (JE), a severe neuroinflammation in the central nervous system (CNS) following infection with JE virus (JEV). Macrophages are known for their heterogeneity, polarizing into M1 or M2 phenotypes in the context of various immunopathological diseases. A comprehensive understanding of macrophage polarization and its relevance to JE progression holds significant promise for advancing JE control and therapeutic strategies. METHODS: To elucidate the role of NADPH oxidase-derived reactive oxygen species (ROS) in JE progression, we assessed viral load, M1 macrophage accumulation, and cytokine production in WT and NADPH oxidase 2 (NOX2)-deficient mice using murine JE model. Additionally, we employed bone marrow (BM) cell-derived macrophages to delineate ROS-mediated regulation of macrophage polarization by ROS following JEV infection. RESULTS: NOX2-deficient mice exhibited increased resistance to JE progression rather than heightened susceptibility, driven by the regulation of macrophage polarization. These mice displayed reduced viral loads in peripheral lymphoid tissues and the CNS, along with diminished infiltration of inflammatory cells into the CNS, thereby resulting in attenuated neuroinflammation. Additionally, NOX2-deficient mice exhibited enhanced JEV-specific Th1 CD4 + and CD8 + T cell responses and increased accumulation of M1 macrophages producing IL-12p40 and iNOS in peripheral lymphoid and inflamed extraneural tissues. Mechanistic investigations revealed that NOX2-deficient macrophages displayed a more pronounced differentiation into M1 phenotypes in response to JEV infection, thereby leading to the suppression of viral replication. Importantly, the administration of H2O2 generated by NOX2 was shown to inhibit M1 macrophage polarization. Finally, oral administration of the ROS scavenger, butylated hydroxyanisole (BHA), bolstered resistance to JE progression and reduced viral loads in both extraneural tissues and the CNS, along with facilitated accumulation of M1 macrophages. CONCLUSION: In light of our results, it is suggested that ROS generated by NOX2 play a role in undermining the control of JEV replication within peripheral extraneural tissues, primarily by suppressing M1 macrophage polarization. Subsequently, this leads to an augmentation in the viral load invading the CNS, thereby facilitating JE progression. Hence, our findings ultimately underscore the significance of ROS-mediated macrophage polarization in the context of JE progression initiated JEV infection.


Subject(s)
Macrophages , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , Animals , Mice , Macrophages/metabolism , Macrophages/immunology , Macrophages/virology , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Encephalitis, Japanese/immunology , Reactive Oxygen Species/metabolism , Encephalitis Virus, Japanese , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/virology , Cell Polarity/drug effects , Cell Polarity/physiology
9.
J Neuroinflammation ; 21(1): 116, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702778

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH), a severe subtype of stroke, is characterized by notably high mortality and morbidity, largely due to the lack of effective therapeutic options. Although the neuroprotective potential of PPARg and Nrf2 has been recognized, investigative efforts into oroxin A (OA), remain limited in preclinical studies. METHODS: SAH was modeled in vivo through filament perforation in male C57BL/6 mice and in vitro by exposing HT22 cells to hemin to induce neuronal damage. Following the administration of OA, a series of methods were employed to assess neurological behaviors, brain water content, neuronal damage, cell ferroptosis, and the extent of neuroinflammation. RESULTS: The findings indicated that OA treatment markedly improved survival rates, enhanced neurological functions, mitigated neuronal death and brain edema, and attenuated the inflammatory response. These effects of OA were linked to the suppression of microglial activation. Moreover, OA administration was found to diminish ferroptosis in neuronal cells, a critical factor in early brain injury (EBI) following SAH. Further mechanistic investigations uncovered that OA facilitated the translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus, thereby activating the Nrf2/GPX4 pathway. Importantly, OA also upregulated the expression of FSP1, suggesting a significant and parallel protective effect against ferroptosis in EBI following SAH in synergy with GPX4. CONCLUSION: In summary, this research indicated that the PPARg activator OA augmented the neurological results in rodent models and diminished neuronal death. This neuroprotection was achieved primarily by suppressing neuronal ferroptosis. The underlying mechanism was associated with the alleviation of cellular death through the Nrf2/GPX4 and FSP1/CoQ10 pathways.


Subject(s)
Ferroptosis , Mice, Inbred C57BL , Neuroinflammatory Diseases , Subarachnoid Hemorrhage , Animals , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/complications , Ferroptosis/drug effects , Ferroptosis/physiology , Mice , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurons/metabolism , Neurons/drug effects , Neurons/pathology
10.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791125

ABSTRACT

The brain is the central organ of adaptation to stress because it perceives and determines threats that induce behavioral, physiological, and molecular responses. In humans, chronic stress manifests as an enduring consistent feeling of pressure and being overwhelmed for an extended duration. This can result in a persistent proinflammatory response in the peripheral and central nervous system (CNS), resulting in cellular, physiological, and behavioral effects. Compounding stressors may increase the risk of chronic-stress-induced inflammation, which can yield serious health consequences, including mental health disorders. This review summarizes the current knowledge surrounding the neuroinflammatory response in rodent models of chronic stress-a relationship that is continually being defined. Many studies investigating the effects of chronic stress on neuroinflammation in rodent models have identified significant changes in inflammatory modulators, including nuclear factor-κB (NF-κB) and toll-like receptors (TLRs), and cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1ß, and IL-6. This suggests that these are key inflammatory factors in the chronic stress response, which may contribute to the establishment of anxiety and depression-like symptoms. The behavioral and neurological effects of modulating inflammatory factors through gene knockdown (KD) and knockout (KO), and conventional and alternative medicine approaches, are discussed.


Subject(s)
Disease Models, Animal , Neuroinflammatory Diseases , Stress, Psychological , Animals , Humans , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Stress, Psychological/metabolism , Rodentia , Chronic Disease , Cytokines/metabolism , NF-kappa B/metabolism , Inflammation/metabolism
11.
Int J Mol Sci ; 25(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38791588

ABSTRACT

Several clinical studies reported that the elevated expression of Chitinase-3-like 1 (CHI3L1) was observed in patients suffering from a wide range of diseases: cancer, metabolic, and neurological diseases. However, the role of CHI3L1 in AD is still unclear. Our previous study demonstrated that 2-({3-[2-(1-Cyclohexen-1-yl)ethyl]-6,7-dimethoxy-4-oxo-3,4-dihydro-2-quinazolinyl}culfanyl)-N-(4-ethylphenyl)butanamide, a CHI3L1 inhibiting compound, alleviates memory and cognitive impairment and inhibits neuroinflammation in AD mouse models. In this study, we studied the detailed correlation of CHI3L1 and AD using serum from AD patients and using CHI3L1 knockout (KO) mice with Aß infusion (300 pmol/day, 14 days). Serum levels of CHI3L1 were significantly elevated in patients with AD compared to normal subjects, and receiver operating characteristic (ROC) analysis data based on serum analysis suggested that CHI3L1 could be a significant diagnostic reference for AD. To reveal the role of CHI3L1 in AD, we investigated the CHI3L1 deficiency effect on memory impairment in Aß-infused mice and microglial BV-2 cells. In CHI3L1 KO mice, Aß infusion resulted in lower levels of memory dysfunction and neuroinflammation compared to that of WT mice. CHI3L1 deficiency selectively inhibited phosphorylation of ERK and IκB as well as inhibition of neuroinflammation-related factors in vivo and in vitro. On the other hand, treatment with recombinant CHI3L1 increased neuroinflammation-related factors and promoted phosphorylation of IκB except for ERK in vitro. Web-based gene network analysis and our results showed that CHI3L1 is closely correlated with PTX3. Moreover, in AD patients, we found that serum levels of PTX3 were correlated with serum levels of CHI3L1 by Spearman correlation analysis. These results suggest that CHI3L1 deficiency could inhibit AD development by blocking the ERK-dependent PTX3 pathway.


Subject(s)
Amyloid beta-Peptides , Chitinase-3-Like Protein 1 , Cognitive Dysfunction , MAP Kinase Signaling System , Mice, Knockout , Neuroinflammatory Diseases , Animals , Chitinase-3-Like Protein 1/genetics , Chitinase-3-Like Protein 1/metabolism , Mice , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/genetics , Amyloid beta-Peptides/metabolism , Humans , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Male , MAP Kinase Signaling System/drug effects , C-Reactive Protein/metabolism , Female , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/drug therapy , Down-Regulation , Disease Models, Animal , Aged , Mice, Inbred C57BL
12.
ACS Nano ; 18(21): 13836-13848, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38753820

ABSTRACT

Advanced in vivo imaging techniques have facilitated the comprehensive visual exploration of animal biological processes, leading to groundbreaking discoveries such as the glymphatic system. However, current limitations of macroscopic imaging techniques impede the precise investigation of physiological parameters regulating this specialized lymphatic transport system. While NIR-II fluorescence imaging has demonstrated advantages in peripheral lymphatic imaging, there are few reports regarding its utilization in the glymphatic system. To address this, a noninvasive transcranial macroscopic NIR-II fluorescence imaging model is developed using a cyanine dye-protein coupled nanoprobe. NIR-II imaging with high temporal and spatial resolution reveals that hypothermia can increase the glymphatic influx by reducing the flow rate of cerebrospinal fluid. In addition, respiratory rate, respiratory amplitude, and heart rate all play a role in regulating the glymphatic influx. Thus, targeting the glymphatic influx may alter the trajectory of immune inflammation following brain injury, providing therapeutic prospects for treating brain injury with mild hypothermia.


Subject(s)
Brain Injuries , Glymphatic System , Animals , Glymphatic System/diagnostic imaging , Glymphatic System/metabolism , Brain Injuries/metabolism , Brain Injuries/diagnostic imaging , Brain Injuries/therapy , Mice , Optical Imaging , Hypothermia/metabolism , Neuroinflammatory Diseases/diagnostic imaging , Neuroinflammatory Diseases/metabolism , Infrared Rays , Fluorescent Dyes/chemistry , Male , Hypothermia, Induced , Mice, Inbred C57BL , Carbocyanines/chemistry
13.
Int J Mol Sci ; 25(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38791415

ABSTRACT

In recent years, there has been a growing interest in the concept of the "gut-brain axis". In addition to well-studied diseases associated with an imbalance in gut microbiota, such as cancer, chronic inflammation, and cardiovascular diseases, research is now exploring the potential role of gut microbial dysbiosis in the onset and development of brain-related diseases. When the function of the intestinal barrier is altered by dysbiosis, the aberrant immune system response interacts with the nervous system, leading to a state of "neuroinflammation". The gut microbiota-brain axis is mediated by inflammatory and immunological mechanisms, neurotransmitters, and neuroendocrine pathways. This narrative review aims to illustrate the molecular basis of neuroinflammation and elaborate on the concept of the gut-brain axis by virtue of analyzing the various metabolites produced by the gut microbiome and how they might impact the nervous system. Additionally, the current review will highlight how sex influences these molecular mechanisms. In fact, sex hormones impact the brain-gut microbiota axis at different levels, such as the central nervous system, the enteric nervous one, and enteroendocrine cells. A deeper understanding of the gut-brain axis in human health and disease is crucial to guide diagnoses, treatments, and preventive interventions.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Neuroinflammatory Diseases , Sex Characteristics , Humans , Brain-Gut Axis/physiology , Neuroinflammatory Diseases/metabolism , Animals , Dysbiosis , Gonadal Steroid Hormones/metabolism , Brain/metabolism , Female , Male , Inflammation/metabolism
14.
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
15.
Exp Neurol ; 377: 114806, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38701941

ABSTRACT

Endoplasmic reticulum (ER) stress and neuroinflammation play an important role in secondary brain damage after traumatic brain injury (TBI). Due to the complex brain cytoarchitecture, multiple cell types are affected by TBI. However, cell type-specific and sex-specific responses to ER stress and neuroinflammation remain unclear. Here we investigated differential regulation of ER stress and neuroinflammatory pathways in neurons and microglia during the acute phase post-injury in a mouse model of impact acceleration TBI in both males and females. We found that TBI resulted in significant weight loss only in males, and sensorimotor impairment and depressive-like behaviors in both males and females at the acute phase post-injury. By concurrently isolating neurons and microglia from the same brain sample of the same animal, we were able to evaluate the simultaneous responses in neurons and microglia towards ER stress and neuroinflammation in both males and females. We discovered that the ER stress and anti-inflammatory responses were significantly stronger in microglia, especially in female microglia, compared with the male and female neurons. Whereas the degree of phosphorylated-tau (pTau) accumulation was significantly higher in neurons, compared with the microglia. In conclusion, TBI resulted in behavioral deficits and cell type-specific and sex-specific responses to ER stress and neuroinflammation, and abnormal protein accumulation at the acute phase after TBI in immature mice.


Subject(s)
Brain Injuries, Traumatic , Endoplasmic Reticulum Stress , Mice, Inbred C57BL , Microglia , Neuroinflammatory Diseases , Neurons , Sex Characteristics , Animals , Female , Mice , Male , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/psychology , Endoplasmic Reticulum Stress/physiology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology
16.
Cells ; 13(10)2024 May 20.
Article in English | MEDLINE | ID: mdl-38786099

ABSTRACT

Mucopolysaccharidosis III type C (MPS IIIC) is an untreatable neuropathic lysosomal storage disease caused by a genetic deficiency of the lysosomal N-acetyltransferase, HGSNAT, catalyzing a transmembrane acetylation of heparan sulfate. HGSNAT is a transmembrane enzyme incapable of free diffusion between the cells or their cross-correction, which limits development of therapies based on enzyme replacement and gene correction. Since our previous work identified neuroinflammation as a hallmark of the CNS pathology in MPS IIIC, we tested whether it can be corrected by replacement of activated brain microglia with neuroprotective macrophages/microglia derived from a heterologous HSPC transplant. Eight-week-old MPS IIIC (HgsnatP304L) mice were transplanted with HSPC from congenic wild type mice after myeloablation with Busulfan and studied using behavior test battery, starting from the age of 6 months. At the age of ~8 months, mice were sacrificed to study pathological changes in the brain, heparan sulfate storage, and other biomarkers of the disease. We found that the treatment corrected several behavior deficits including hyperactivity and reduction in socialization, but not memory decline. It also improved several features of CNS pathology such as microastroglyosis, expression of pro-inflammatory cytokine IL-1ß, and accumulation of misfolded amyloid aggregates in cortical neurons. At the periphery, the treatment delayed development of terminal urinary retention, potentially increasing longevity, and reduced blood levels of heparan sulfate. However, we did not observe correction of lysosomal storage phenotype in neurons and heparan sulfate brain levels. Together, our results demonstrate that neuroinflammation in a neurological lysosomal storage disease, caused by defects in a transmembrane enzyme, can be effectively ameliorated by replacement of microglia bearing the genetic defect with cells from a normal healthy donor. They also suggest that heterologous HSPC transplant, if used together with other methods, such as chaperone therapy or substrate reduction therapy, may constitute an effective combination therapy for MPS IIIC and other disorders with a similar etiology.


Subject(s)
Disease Models, Animal , Mucopolysaccharidosis III , Neuroinflammatory Diseases , Animals , Mucopolysaccharidosis III/pathology , Mucopolysaccharidosis III/therapy , Mucopolysaccharidosis III/genetics , Mice , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Lysosomes/metabolism , Microglia/pathology , Microglia/metabolism , Mice, Inbred C57BL , Brain/pathology , Brain/metabolism , Heparitin Sulfate/metabolism , Inflammation/pathology
17.
J Med Chem ; 67(10): 8383-8395, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38695469

ABSTRACT

Interleukin receptor associated kinase 4 (IRAK4) plays an important role in innate immune signaling through Toll-like and interleukin-1 receptors and represents an attractive target for the treatment of inflammatory diseases and cancer. We previously reported the development of a potent, selective, and brain-penetrant imidazopyrimidine series of IRAK4 inhibitors. However, lead molecule BIO-7488 (1) suffered from low solubility which led to variable PK, compound accumulation, and poor in vivo tolerability. Herein, we describe the discovery of a series of pyridone analogs with improved solubility which are highly potent, selective and demonstrate desirable PK profiles including good oral bioavailability and excellent brain penetration. BIO-8169 (2) reduced the in vivo production of pro-inflammatory cytokines, was well tolerated in safety studies in rodents and dog at margins well above the predicted efficacious exposure and showed promising results in a mouse model for multiple sclerosis.


Subject(s)
Brain , Interleukin-1 Receptor-Associated Kinases , Protein Kinase Inhibitors , Animals , Dogs , Male , Mice , Rats , Brain/metabolism , Brain/drug effects , Drug Discovery , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/therapeutic use , Structure-Activity Relationship
18.
Medicine (Baltimore) ; 103(18): e38086, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701247

ABSTRACT

BACKGROUND: Dementia is a major public health challenge for aging societies worldwide. Neuroinflammation is thought to be a key factor in dementia development. The aim of this study was to comprehensively assess translocator protein (TSPO) expression by positron emission tomography (PET) imaging to reveal the characteristics of neuroinflammation in dementia. METHODS: We used a meta-analysis to retrieve literature on TSPO expression in dementia using PET imaging technology, including but not limited to the quality of the study design, sample size, and the type of TSPO ligand used in the study. For the included studies, we extracted key data, including TSPO expression levels, clinical characteristics of the study participants, and specific information on brain regions. Meta-analysis was performed using R software to assess the relationship between TSPO expression and dementia. RESULTS: After screening, 12 studies that met the criteria were included. The results of the meta-analysis showed that the expression level of TSPO was significantly elevated in patients with dementia, especially in the hippocampal region. The OR in the hippocampus was 1.50 with a 95% CI of 1.09 to 1.25, indicating a significant increase in the expression of TSPO in this region compared to controls. Elevated levels of inflammation in the prefrontal lobe and cingulate gyrus are associated with cognitive impairment in patients. This was despite an OR of 1.00 in the anterior cingulate gyrus, indicating that TSPO expression in this region did not correlate significantly with the findings. The overall heterogeneity test showed I² = 51%, indicating moderate heterogeneity. CONCLUSION: This study summarizes the existing literature on TSPO expression in specific regions of the brain in patients with dementia, and also provides some preliminary evidence on the possible association between neuroinflammation and dementia. However, the heterogeneity of results and limitations of the study suggest that we need to interpret these findings with caution. Future studies need to adopt a more rigorous and consistent methodological design to more accurately assess the role of neuroinflammation in dementia, thereby providing a more reliable evidence base for understanding pathological mechanisms and developing potential therapeutic strategies.


Subject(s)
Dementia , Neuroinflammatory Diseases , Positron-Emission Tomography , Receptors, GABA , Humans , Positron-Emission Tomography/methods , Dementia/diagnostic imaging , Dementia/metabolism , Receptors, GABA/metabolism , Neuroinflammatory Diseases/diagnostic imaging , Neuroinflammatory Diseases/metabolism , Brain/diagnostic imaging , Brain/metabolism
19.
Cell Mol Biol Lett ; 29(1): 75, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755530

ABSTRACT

BACKGROUND: Mechanical spinal cord injury (SCI) is a deteriorative neurological disorder, causing secondary neuroinflammation and neuropathy. ADAM8 is thought to be an extracellular metalloproteinase, which regulates proteolysis and cell adherence, but whether its intracellular region is involved in regulating neuroinflammation in microglia after SCI is unclear. METHODS: Using animal tissue RNA-Seq and clinical blood sample examinations, we found that a specific up-regulation of ADAM8 in microglia was associated with inflammation after SCI. In vitro, microglia stimulated by HMGB1, the tail region of ADAM8, promoted microglial inflammation, migration and proliferation by directly interacting with ERKs and Fra-1 to promote activation, then further activated Map3k4/JNKs/p38. Using SCI mice, we used BK-1361, a specific inhibitor of ADAM8, to treat these mice. RESULTS: The results showed that administration of BK-1361 attenuated the level of neuroinflammation and reduced microglial activation and recruitment by inhibiting the ADAM8/Fra-1 axis. Furthermore, treatment with BK-1361 alleviated glial scar formation, and also preserved myelin and axonal structures. The locomotor recovery of SCI mice treated with BK-1361 was therefore better than those without treatment. CONCLUSIONS: Taken together, the results showed that ADAM8 was a critical molecule, which positively regulated neuroinflammatory development and secondary pathogenesis by promoting microglial activation and migration. Mechanically, ADAM8 formed a complex with ERK and Fra-1 to further activate the Map3k4/JNK/p38 axis in microglia. Inhibition of ADAM8 by treatment with BK-1361 decreased the levels of neuroinflammation, glial formation, and neurohistological loss, leading to favorable improvement in locomotor functional recovery in SCI mice.


Subject(s)
ADAM Proteins , Membrane Proteins , Microglia , Neuroinflammatory Diseases , Proto-Oncogene Proteins c-fos , Spinal Cord Injuries , Animals , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/drug therapy , Mice , Microglia/metabolism , Microglia/drug effects , ADAM Proteins/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , MAP Kinase Signaling System/drug effects , Inflammation/pathology , Inflammation/drug therapy , Cell Movement/drug effects , Humans , Antigens, CD
20.
J Neuroinflammation ; 21(1): 137, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802820

ABSTRACT

Hyperglycemia has been shown to modulate the immune response of peripheral immune cells and organs, but the impact of hyperglycemia on neuroinflammation within the brain remains elusive. In the present study, we provide evidences that streptozotocin (STZ)-induced hyperglycemic condition in mice drives a phenotypic switch of brain astrocytes to a proinflammatory state, and increases brain vulnerability to mild peripheral inflammation. In particular, we found that hyperglycemia led to a significant increase in the astrocyte proliferation as determined by flow cytometric and immunohistochemical analyses of mouse brain. The increased astrocyte proliferation by hyperglycemia was reduced by Glut1 inhibitor BAY-876. Transcriptomic analysis of isolated astrocytes from Aldh1l1CreERT2;tdTomato mice revealed that peripheral STZ injection induced astrocyte reprogramming into proliferative, and proinflammatory phenotype. Additionally, STZ-induced hyperglycemic condition significantly enhanced the infiltration of circulating myeloid cells into the brain and the disruption of blood-brain barrier in response to mild lipopolysaccharide (LPS) administration. Systemic hyperglycemia did not alter the intensity and sensitivity of peripheral inflammation in mice to LPS challenge, but increased the inflammatory potential of brain microglia. In line with findings from mouse experiments, a high-glucose environment intensified the LPS-triggered production of proinflammatory molecules in primary astrocyte cultures. Furthermore, hyperglycemic mice exhibited a significant impairment in cognitive function after mild LPS administration compared to normoglycemic mice as determined by novel object recognition and Y-maze tasks. Taken together, these results demonstrate that hyperglycemia directly induces astrocyte reprogramming towards a proliferative and proinflammatory phenotype, which potentiates mild LPS-triggered inflammation within brain parenchymal regions.


Subject(s)
Astrocytes , Brain , Hyperglycemia , Lipopolysaccharides , Mice, Inbred C57BL , Neuroinflammatory Diseases , Animals , Hyperglycemia/chemically induced , Hyperglycemia/pathology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Mice , Lipopolysaccharides/toxicity , Lipopolysaccharides/pharmacology , Brain/pathology , Brain/metabolism , Brain/drug effects , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/chemically induced , Male , Cellular Reprogramming/drug effects , Cellular Reprogramming/physiology , Mice, Transgenic , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...