Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.106
Filtrer
1.
J Transl Med ; 22(1): 876, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39350202

RÉSUMÉ

BACKGROUND: Tobacco smoking is the leading cause of preventable death and disease worldwide, with over 8 million annual deaths attributed to cigarette smoking. This study investigates the impact of cigarette smoke and heated tobacco products (HTPs) on microglial function, focusing on toxicological profiles, inflammatory responses, and oxidative stress using ISO standard and clinically relevant conditions of exposure. METHODS: We assessed cell viability, reactive oxygen species (ROS) production, lipid peroxidation, mitochondrial function, unfolded protein response, and inflammation in human microglial cells (HMC3) exposed to cigarette smoke, HTP aerosol or nicotine. RESULTS: Our findings show that cigarette smoke significantly reduces microglial viability, increases ROS formation, induces lipid peroxidation, and reduces intracellular glutathione levels. Cigarette smoke also alters the expression of genes involved in mitochondrial dynamics and biogenesis, leading to mitochondrial dysfunction. Additionally, cigarette smoke impairs the unfolded protein response, activates the NF-κB pathway, and induces a pro-inflammatory state characterized by increased TNF and IL-18 expression. Furthermore, cigarette smoke causes DNA damage and decreases the expression of the aging marker Klotho ß. In contrast, HTP, exhibited a lesser degree of microglial toxicity, with reduced ROS production, lipid peroxidation, and mitochondrial dysfunction compared to conventional cigarettes. CONCLUSION: These results highlight the differential toxicological profile of cigarette smoke and HTP on microglial cells, suggesting a potential harm reduction strategy for neurodegenerative disease for smokers unwilling or unable to quit.


Sujet(s)
Survie cellulaire , Inflammation , Peroxydation lipidique , Microglie , Mitochondries , Stress oxydatif , Espèces réactives de l'oxygène , Fumée , Produits du tabac , Réponse aux protéines mal repliées , Stress oxydatif/effets des médicaments et des substances chimiques , Humains , Espèces réactives de l'oxygène/métabolisme , Inflammation/anatomopathologie , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Microglie/anatomopathologie , Produits du tabac/effets indésirables , Fumée/effets indésirables , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Lignée cellulaire , Température élevée , Facteur de transcription NF-kappa B/métabolisme , Nicotiana/effets indésirables , Altération de l'ADN
2.
Nagoya J Med Sci ; 86(3): 392-406, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39355370

RÉSUMÉ

Traumatic spinal cord injury is characterized by immediate and irreversible tissue loss at the lesion site and secondary tissue damage. Secondary injuries should, in principle, be preventable, although no effective treatment options currently exist for patients with acute spinal cord injury. Traumatized tissues release excessive amounts of adenosine triphosphate and activate the P2X purinoceptor 7/pannexin1 complex, which is associated with secondary injury. We investigated the neuroprotective effects of the blue dye Brilliant Blue FCF, a selective inhibitor of P2X purinoceptor 7/pannexin1 that is approved for use as a food coloring, by comparing it with Brilliant Blue G, a P2X7 purinoceptor antagonist, and carbenoxolone, which attenuates P2X purinoceptor 7/pannexin1 function, in a rat spinal cord injury model. Brilliant Blue FCF administered early after spinal cord injury reduced spinal cord anatomical damage and improved motor recovery without apparent toxicity. Brilliant Blue G had the highest effect on this neurological recovery, with Brilliant Blue FCF and carbenoxolone having comparable improvement. Furthermore, Brilliant Blue FCF administration reduced local astrocytic and microglial activation and neutrophil infiltration, and no differences in these histological effects were observed between compounds. Thus, Brilliant Blue FCF protects spinal cord neurons after spinal cord injury and suppresses local inflammatory responses as well as Brilliant Blue G and carbenoxolone.


Sujet(s)
Adénosine triphosphate , Carbénoxolone , Connexines , Protéines de tissu nerveux , Récupération fonctionnelle , Magenta I , Traumatismes de la moelle épinière , Traumatismes de la moelle épinière/traitement médicamenteux , Traumatismes de la moelle épinière/métabolisme , Animaux , Connexines/métabolisme , Connexines/antagonistes et inhibiteurs , Adénosine triphosphate/métabolisme , Carbénoxolone/pharmacologie , Carbénoxolone/usage thérapeutique , Magenta I/pharmacologie , Magenta I/usage thérapeutique , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/antagonistes et inhibiteurs , Récupération fonctionnelle/effets des médicaments et des substances chimiques , Rats , Antagonistes des récepteurs purinergiques P2X/pharmacologie , Antagonistes des récepteurs purinergiques P2X/usage thérapeutique , Rat Sprague-Dawley , Modèles animaux de maladie humaine , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Moelle spinale/effets des médicaments et des substances chimiques , Moelle spinale/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Récepteurs purinergiques P2X7/effets des médicaments et des substances chimiques , Femelle , Infiltration par les neutrophiles/effets des médicaments et des substances chimiques
3.
J Toxicol Sci ; 49(10): 435-446, 2024.
Article de Anglais | MEDLINE | ID: mdl-39358233

RÉSUMÉ

BACKGROUND: Neuroinflammation plays a critical role in various neurological disorders. Oxycodone has anti-inflammatory properties. The purpose of this work was to look into the effect of oxycodone in controlling lipopolysaccharide (LPS)-induced neuroinflammation in microglia. METHODS: LPS-induced HMC3 cells were subjected to oxycodone (2.5, 5, 10 and 20 µg/mL). The mRNA and protein expressions were examined by qRT-PCR and western blotting. TNF-α, IL-1ß, IL-6, and IL-8 levels were assessed by ELISA. MTT assay was adopted to measure cell viability. The interactions between CREB, miR-181c and PDCD4 were analyzed by dual-luciferase reporter assay, ChIP and/or RIP assays. RESULTS: Oxycodone treatment alleviated LPS-induced inflammation in HMC3 cells and increased p-CREB level, but reduced PDCD4 and iNOS levels in LPS-treated cells. Mechanistically, oxycodone mitigated LPS-induced neuroinflammation by upregulating miR-181c. In addition, CREB promoted miR-181c expression by directly binding to the MIR181C promoter, and miR-181c inhibited PDCD4 expression by directly binding to PDCD4 3'UTR. As expected, oxycodone alleviated LPS-induced neuroinflammation by regulating the CREB/miR-181c/PDCD4 axis. CONCLUSION: Oxycodone attenuated LPS-induced neuroinflammation in microglia by regulating the CREB/miR-181c/PDCD4 axis. These findings proved that oxycodone is a potential drug for treating neuroinflammation and elucidate the mechanisms involved.


Sujet(s)
Protéines régulatrices de l'apoptose , Protéine de liaison à l'élément de réponse à l'AMP cyclique , Lipopolysaccharides , microARN , Microglie , Maladies neuro-inflammatoires , Oxycodone , Protéines de liaison à l'ARN , microARN/génétique , microARN/métabolisme , Oxycodone/pharmacologie , Oxycodone/effets indésirables , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/génétique , Humains , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Maladies neuro-inflammatoires/induit chimiquement , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/génétique , Anti-inflammatoires/pharmacologie , Lignée cellulaire , Inflammation/induit chimiquement , Inflammation/génétique , Transduction du signal/effets des médicaments et des substances chimiques
4.
Neurosciences (Riyadh) ; 29(4): 276-283, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39379083

RÉSUMÉ

OBJECTIVES: To determine the effects of all-trans-retinoic acid (ATRA) on the post-stroke inflammatory response and elucidate the underlying molecular mechanisms. METHODS: This animal experiment was conducted at Central Laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China during 2020-2022. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 1.5 h, and treated with ATRA at 2 and 24 h after reperfusion. Neurological deficit scores on behavioral tests, and cerebral infarct volume, microglial polarization, and the expression levels of inflammatory cytokines and proteins associated with TLR4/NF-κB signaling were assessed. RESULTS: The ATRA administration reduced cerebral infarct volume and ameliorated neurological deficit scores in MCAO rats. Additionally, ATRA relieved cerebral edema and downregulated the secretion of proinflammatory cytokines after stroke. Finally, ATRA attenuated the polarization of the microglia toward the M1 phenotype and promoted the activation of the beneficial M2 phenotype; the underlying mechanism potentially involved the suppression of the TLR4/NF-κB signaling pathway. CONCLUSION: The ATRA treatment promoted functional recovery in an experimental model of ischemic stroke by attenuating neural inflammation. ATRA potentially modulated microglia-mediated neuroinflammation via the downregulation of the TLR4/NF-κB signaling pathway, which makes it a candidate treatment for post-stroke neuroinflammation.


Sujet(s)
Régulation négative , Infarctus du territoire de l'artère cérébrale moyenne , Facteur de transcription NF-kappa B , Neuroprotecteurs , Rat Sprague-Dawley , Transduction du signal , Récepteur de type Toll-4 , Trétinoïne , Animaux , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/effets des médicaments et des substances chimiques , Trétinoïne/pharmacologie , Trétinoïne/usage thérapeutique , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Mâle , Rats , Modèles animaux de maladie humaine , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme
5.
J Neuroinflammation ; 21(1): 251, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39369253

RÉSUMÉ

Neuroinflammation has been implicated in the pathogenesis of several neurologic and psychiatric disorders. Microglia are key drivers of neuroinflammation and, in response to different inflammatory stimuli, overexpress a proinflammatory signature of genes. Among these, Ch25h is a gene overexpressed in brain tissue from Alzheimer's disease as well as various mouse models of neuroinflammation. Ch25h encodes cholesterol 25-hydroxylase, an enzyme upregulated in activated microglia under conditions of neuroinflammation, that hydroxylates cholesterol to form 25-hydroxycholesterol (25HC). 25HC can be further metabolized to 7α,25-dihydroxycholesterol, which is a potent chemoattractant of leukocytes. We have previously shown that 25HC increases the production and secretion of the proinflammatory cytokine, IL-1ß, by primary mouse microglia treated with lipopolysaccharide (LPS). In the present study, wildtype (WT) and Ch25h-knockout (KO) mice were peripherally administered LPS to induce an inflammatory state in the brain. In LPS-treated WT mice, Ch25h expression and 25HC levels increased in the brain relative to vehicle-treated WT mice. Among LPS-treated WT mice, females produced significantly higher levels of 25HC and showed transcriptomic changes reflecting higher levels of cytokine production and leukocyte migration than WT male mice. However, females were similar to males among LPS-treated KO mice. Ch25h-deficiency coincided with decreased microglial activation in response to systemic LPS. Proinflammatory cytokine production and intra-parenchymal infiltration of leukocytes were significantly lower in KO compared to WT mice. Amounts of IL-1ß and IL-6 in the brain strongly correlated with 25HC levels. Our results suggest a proinflammatory role for 25HC in the brain following peripheral administration of LPS.


Sujet(s)
Encéphale , Cytokines , Modèles animaux de maladie humaine , Hydroxycholestérols , Leucocytes , Lipopolysaccharides , Souris de lignée C57BL , Souris knockout , Maladies neuro-inflammatoires , Animaux , Lipopolysaccharides/toxicité , Lipopolysaccharides/pharmacologie , Hydroxycholestérols/métabolisme , Hydroxycholestérols/pharmacologie , Souris , Cytokines/métabolisme , Mâle , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Femelle , Leucocytes/effets des médicaments et des substances chimiques , Leucocytes/métabolisme , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/induit chimiquement , Maladies neuro-inflammatoires/anatomopathologie , Steroid hydroxylases/métabolisme , Steroid hydroxylases/génétique , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Cellules cultivées
6.
J Neuroinflammation ; 21(1): 256, 2024 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-39390483

RÉSUMÉ

BACKGROUND: Adverse events in early life can have impact lasting into adulthood. We investigated the long-term effects of systemic inflammation during postnatal development on adult microglial responses to lipopolysaccharide (LPS) in two CNS regions (cortex, cervical spinal cord) in male and female rats. METHODS: Inflammation was induced in Sprague-Dawley rats by LPS (1 mg/kg) administered intraperitoneally during postnatal development at P7, P12 or P18. As adults (12 weeks of age), the rats received a second LPS dose (1 mg/kg). Control rats received saline. Microglia were isolated 3 h post-LPS followed by gene expression analysis via qRT-PCR for pro-inflammatory (IL-6, iNOS, Ptgs2, C/EBPb, CD14, CXCL10), anti-inflammatory (CD68, Arg-1), and homeostatic genes (P2Y12, Tmemm119). CSF-1 and CX3CL1 mRNAs were analyzed in microglia-free homogenates. RESULTS: Basal gene expression in adult microglia was largely unaffected by postnatal inflammation. Adult cortical microglial pro-inflammatory gene responses to LPS were either unchanged or attenuated in rats exposed to LPS during postnatal development. Ptgs2, C/EBPb, CXCL10 and Arg-1 were the most affected genes, with expression significantly downregulated vs. rats without postnatal LPS. Spinal microglia were affected most by LPS at P18, with mixed and sometimes opposing effects on proinflammatory genes in males vs. females. Overall, male cortical vs. spinal microglia were more affected by postnatal LPS. Females were affected in both cortex and spinal cord, but the effect was dependent on timing of postnatal LPS. Overall, inflammatory challenge at P18 had greater effect on adult microglia vs. challenge at P12 or P7. CONCLUSIONS: Long-lasting effects of postnatal inflammation on adult microglia depend on postnatal timing, CNS region and sex.


Sujet(s)
Animaux nouveau-nés , Inflammation , Lipopolysaccharides , Microglie , Rat Sprague-Dawley , Caractères sexuels , Animaux , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Femelle , Rats , Mâle , Inflammation/induit chimiquement , Inflammation/métabolisme , Inflammation/anatomopathologie , Facteurs âges , Moelle spinale/métabolisme , Moelle spinale/anatomopathologie , Moelle spinale/effets des médicaments et des substances chimiques
7.
FASEB J ; 38(19): e70094, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39373933

RÉSUMÉ

Berberine and palmatine are isoquinoline quaternary alkaloids derived from Chinese medicinal herbs. These alkaloids have shown promising synergy in inhibiting acetylcholinesterase (AChE), indicating their potential in treating Alzheimer's disease (AD). Besides, the anti-inflammatory effects of berberine and palmatine have been widely reported, although the underlying mechanism remains unclear. Here, we found that berberine and palmatine could induce calcium ion (Ca2+) influx via activating α7 nicotinic acetylcholine receptor (α7 nAChR) in cultured microglial cells, possibly serving as its allosteric potential ligands. Furthermore, we examined the synergistic anti-inflammatory effects of berberine and palmatine in the LPS-induced microglia, that significantly suppressed the production of TNF-α and iNOS. Notably, this suppression was reversed by co-treatment with a selective antagonist of α7 nAChR. Moreover, the alkaloid-induced microglial phagocytosis was shown to be mediated by the induction of Ca2+ influx through α7 nAChR and subsequent CaMKII-Rac1-dependent pathway. Additionally, the combination of berberine and palmatine, at low concentration, protected against the LPS-induced endoplasmic reticulum stress and mitochondrial dysfunction in microglia. These findings indicate the potential of berberine and palmatine, either individually or in combination, in contributing to anti-AD drug development, which provide valuable insights into the mechanisms by which natural products, such as plant alkaloids, exert their anti-AD effects.


Sujet(s)
Alcaloïdes de type berbérine , Berbérine , Inflammation , Microglie , Phagocytose , Récepteur nicotinique de l'acétylcholine alpha7 , Berbérine/pharmacologie , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Alcaloïdes de type berbérine/pharmacologie , Animaux , Récepteur nicotinique de l'acétylcholine alpha7/métabolisme , Phagocytose/effets des médicaments et des substances chimiques , Souris , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Régulation allostérique/effets des médicaments et des substances chimiques , Lipopolysaccharides/pharmacologie , Synergie des médicaments , Ligands , Calcium/métabolisme , Anti-inflammatoires/pharmacologie
8.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-39337650

RÉSUMÉ

Anxiety disorder is a universal disease related to neuro-inflammation. Solanesol has shown positive effects because of its anti-inflammatory, anti-tumor, and anti-ulcer properties. This study focused on determining whether solanesol could ameliorate anxiety-like behaviors in a mouse model of neuro-inflammation and identify its working targets. Complete Freund's adjuvant (CFA)-induced mice that were intra-peritoneally administered with solanesol (50 mg/kg) for 1 week showed a statistically significant reduction in anxiety-like behaviors, as measured by open field and elevated plus-maze tests. Western blot analysis revealed that CFA-induced upregulation of the levels of pro-inflammatory cytokines interleukin (IL)-1ß and tumor necrosis factor α (TNF-α), which played crucial roles in regulating anxiety, returned to normal in the anterior cingulate cortex (ACC) after solanesol treatment. The level of T cell-restricted intracellular antigen-1 (TIA1), a key component of stress granules, also decreased in the ACC. Moreover, immunofluorescence results indicated that solanesol suppressed CFA-induced microglial and astrocytic activation in the ACC. CFA was injected in the hind paws of TIA1Nestin conditional knockout (cKO) mice to confirm whether TIA1 is a potential modulatory molecule that influences pro-inflammatory cytokines and anxiety-like behaviors. Anxiety-like behaviors could not be observed in cKO mice after CFA injection with IL-1ß and TNF-α levels not remarkedly increasing. Our findings suggest that solanesol inhibits neuro-inflammation by decreasing the TIA1 level to reduce IL-1ß and TNF-α expression, meanwhile inhibiting microglial and astrocytic activation in the ACC and ultimately ameliorating anxiety-like behaviors in mice.


Sujet(s)
Anxiété , Modèles animaux de maladie humaine , Adjuvant Freund , Gyrus du cingulum , Animaux , Souris , Anxiété/traitement médicamenteux , Mâle , Gyrus du cingulum/effets des médicaments et des substances chimiques , Gyrus du cingulum/métabolisme , Interleukine-1 bêta/métabolisme , Souris de lignée C57BL , Comportement animal/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme
9.
eNeuro ; 11(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39293937

RÉSUMÉ

Retinitis pigmentosa (RP) is a family of genetically heterogeneous diseases still without a cure. Despite the causative genetic mutation typically not expressed in cone photoreceptors, these cells inevitably degenerate following the primary death of rods, causing blindness. The reasons for the "bystander" degeneration of cones are presently unknown but decrement of survival factors, oxidative stress, and inflammation all play a role. Targeting these generalized biological processes represents a strategy to develop mutation-agnostic therapies for saving vision in large populations of RP individuals. A classical method to support neuronal survival is by employing neurotrophic factors, such as NGF. This study uses painless human NGF (hNGFp), a TrkA receptor-biased variant of the native molecule with lower affinity for nociceptors and limited activity as a pain inducer; the molecule has identical neurotrophic power of the native form but a reduced affinity for the p75NTR receptors, known to trigger apoptosis. hNGFp has a recognized activity on brain microglial cells, which are induced to a phenotype switch from a highly activated to a more homeostatic configuration. hNGFp was administered to RP-like mice in vivo with the aim of decreasing retinal inflammation and also providing retinal neuroprotection. However, the ability of this treatment to counteract the bystander degeneration of cones remained limited.


Sujet(s)
Facteur de croissance nerveuse , Rétinite pigmentaire , Rétinite pigmentaire/métabolisme , Rétinite pigmentaire/génétique , Animaux , Facteur de croissance nerveuse/administration et posologie , Facteur de croissance nerveuse/métabolisme , Humains , Rétine/métabolisme , Souris de lignée C57BL , Souris , Modèles animaux de maladie humaine , Récepteur trkA/métabolisme , Mâle , Femelle , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques
10.
Acta Neuropathol Commun ; 12(1): 150, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39300576

RÉSUMÉ

BACKGROUND: Glaucoma is a leading cause of blindness, affecting retinal ganglion cells (RGCs) and their axons. By 2040, it is likely to affect 110 million people. Neuroinflammation, specifically through the release of proinflammatory cytokines by M1 microglial cells, plays a crucial role in glaucoma progression. Indeed, in post-mortem human studies, pre-clinical models, and ex-vivo models, RGC degeneration has been consistently shown to be linked to inflammation in response to cell death and tissue damage. Recently, Rho kinase inhibitors (ROCKis) have emerged as potential therapies for neuroinflammatory and neurodegenerative diseases. This study aimed to investigate the potential effects of three ROCKis (Y-27632, Y-33075, and H-1152) on retinal ganglion cell (RGC) loss and retinal neuroinflammation using an ex-vivo retinal explant model. METHODS: Rat retinal explants underwent optic nerve axotomy and were treated with Y-27632, Y-33075, or H-1152. The neuroprotective effects on RGCs were evaluated using immunofluorescence and Brn3a-specific markers. Reactive glia and microglial activation were studied by GFAP, CD68, and Iba1 staining. Flow cytometry was used to quantify day ex-vivo 4 (DEV 4) microglial proliferation and M1 activation by measuring the number of CD11b+, CD68+, and CD11b+/CD68+ cells after treatment with control solvent or Y-33075. The modulation of gene expression was measured by RNA-seq analysis on control and Y-33075-treated explants and glial and pro-inflammatory cytokine gene expression was validated by RT-qPCR. RESULTS: Y-27632 and H-1152 did not significantly protect RGCs. By contrast, at DEV 4, 50 µM Y-33075 significantly increased RGC survival. Immunohistology showed a reduced number of Iba1+/CD68+ cells and limited astrogliosis with Y-33075 treatment. Flow cytometry confirmed lower CD11b+, CD68+, and CD11b+/CD68+ cell numbers in the Y-33075 group. RNA-seq showed Y-33075 inhibited the expression of M1 microglial markers (Tnfα, Il-1ß, Nos2) and glial markers (Gfap, Itgam, Cd68) and to reduce apoptosis, ferroptosis, inflammasome formation, complement activation, TLR pathway activation, and P2rx7 and Gpr84 gene expression. Conversely, Y-33075 upregulated RGC-specific markers, neurofilament formation, and neurotransmitter regulator expression, consistent with its neuroprotective effects. CONCLUSION: Y-33075 demonstrates marked neuroprotective and anti-inflammatory effects, surpassing the other tested ROCKis (Y-27632 and H-1152) in preventing RGC death and reducing microglial inflammatory responses. These findings highlight its potential as a therapeutic option for glaucoma.


Sujet(s)
Neuroprotecteurs , Pyridines , Cellules ganglionnaires rétiniennes , rho-Associated Kinases , Animaux , Pyridines/pharmacologie , rho-Associated Kinases/métabolisme , rho-Associated Kinases/antagonistes et inhibiteurs , Neuroprotecteurs/pharmacologie , Rats , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Cellules ganglionnaires rétiniennes/anatomopathologie , Cellules ganglionnaires rétiniennes/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Rétine/effets des médicaments et des substances chimiques , Rétine/anatomopathologie , Rétine/métabolisme , Amides/pharmacologie , 5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine/analogues et dérivés , 5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine/pharmacologie , Rat Sprague-Dawley , Neuroprotection/effets des médicaments et des substances chimiques , Neuroprotection/physiologie , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Microglie/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Mâle , Lésions traumatiques du nerf optique/traitement médicamenteux , Lésions traumatiques du nerf optique/anatomopathologie , Lésions traumatiques du nerf optique/métabolisme , Isoquinoléines , Sulfonamides
11.
Virol J ; 21(1): 219, 2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-39285274

RÉSUMÉ

BACKGROUND: To investigate the mechanism of Golgi matrix protein 130(GM130) regulating the antiviral immune response of TLR3 after herpes simplex virus type 1(HSV-1) infection of microglia cells. We explored the regulatory effects of berberine on the immune response mediated by GM130 and TLR3. METHODS: An in vitro model of HSV-1 infection was established by infecting BV2 cells with HSV-1. RESULTS: Compared to the uninfected group, the Golgi apparatus (GA) fragmentation and GM130 decreased after HSV-1 infection; TLR3 increased at 6 h and began to decrease at 12 h after HSV-1 infection; the secretion of interferon-beta(IFN-ß), tumour necrosis factor alpha(TNF-α), and interleukin-6(IL-6) increased after infection. Knockdown of GM130 aggravated fragmentation of the GA and caused TLR3 to further decrease, and the virus titer also increased significantly. GM130 knockdown inhibits the increase in TLR3 and inflammatory factors induced by TLR3 agonists and increases the viral titer. Overexpression of GM130 alleviated fragmentation of the GA induced by HSV-1, partially restored the levels of TLR3, and reduced viral titers. GM130 overexpression reversed the reduction in TLR3 and inflammatory cytokine levels induced by TLR3 inhibitors. Therefore, the decrease in GM130 levels caused by HSV-1 infection leads to increased viral replication by inhibiting TLR3-mediated innate immunity. Berberine can protect the GA and reverse the downregulation of GM130, as well as the downregulation of TLR3 and its downstream factors after HSV-1 infection, reducing the virus titer. CONCLUSIONS: In microglia, one mechanism of HSV-1 immune escape is disruption of the GM130/TLR3 pathway. Berberine protects the GA and enhances TLR3-mediated antiviral immune responses.


Sujet(s)
Régulation négative , Herpèsvirus humain de type 1 , Immunité innée , Microglie , Récepteur de type Toll-3 , Herpèsvirus humain de type 1/immunologie , Herpèsvirus humain de type 1/effets des médicaments et des substances chimiques , Herpèsvirus humain de type 1/physiologie , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Microglie/virologie , Microglie/immunologie , Microglie/effets des médicaments et des substances chimiques , Animaux , Souris , Lignée cellulaire , Échappement immunitaire , Berbérine/pharmacologie , Cytokines/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Herpès/immunologie , Herpès/virologie
12.
Cell Mol Neurobiol ; 44(1): 60, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39287687

RÉSUMÉ

Microglia are macrophage cells residing in the brain, where they exert a key role in neuronal protection. Through the gut-brain axis, metabolites produced by gut commensal microbes can influence brain functions, including microglial activity. The nuclear factor erythroid 2-related factor 2 (NRF2) is a key regulator of the oxidative stress response in microglia, controlling the expression of cytoprotective genes. Lactobacilli-derived cell-free supernatants (CFSs) are postbiotics that have shown antioxidant and immunomodulatory effects in several in vitro and in vivo studies. This study aimed to explore the effects of lactobacilli CFSs on modulating microglial responses against oxidative stress and inflammation. HMC3 microglia were exposed to lipopolysaccaride (LPS), as an inflammatory trigger, before and after administration of CFSs from three human gut probiotic species. The NRF2 nuclear protein activation and the expression of NRF2-controlled antioxidant genes were investigated by immunoassay and quantitative RT-PCR, respectively. Furthermore, the level of pro- and anti-inflammatory cytokines was evaluated by immunoassay. All CFSs induced a significant increase of NRF2 nuclear activity in basal conditions and upon inflammation. The transcription of antioxidant genes, namely heme oxygenase 1, superoxide dismutase (SOD), glutathione-S transferase, glutathione peroxidase, and catalase also increased, especially after inflammatory stimulus. Besides, higher SOD1 activity was detected relative to inflamed microglia. In addition, CFSs pre-treatment of microglia attenuated pro-inflammatory TNF-α levels while increasing anti-inflammatory IL-10 levels. These findings confirmed that gut microorganisms' metabolites can play a relevant role in adjuvating the microglia cellular response against neuroinflammation and oxidative stress, which are known to cause neurodegenerative diseases.


Sujet(s)
Inflammation , Lactobacillus , Microglie , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Transduction du signal , Superoxide dismutase-1 , Humains , Facteur-2 apparenté à NF-E2/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Stress oxydatif/physiologie , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Superoxide dismutase-1/métabolisme , Lipopolysaccharides/pharmacologie , Cytokines/métabolisme , Antioxydants/métabolisme , Antioxydants/pharmacologie , Lignée cellulaire
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1589-1598, 2024 Aug 20.
Article de Chinois | MEDLINE | ID: mdl-39276055

RÉSUMÉ

OBJECTIVE: To investigate the mechanism underlying the neuroprotective effect of linarin (LIN) against microglia activation-mediated inflammation and neuronal apoptosis following spinal cord injury (SCI). METHODS: Fifty C57BL/6J mice (8- 10 weeks old) were randomized to receive sham operation, SCI and linarin treatment at 12.5, 25, and 50 mg/kg following SCI (n=10). Locomotor function recovery of the SCI mice was assessed using the Basso Mouse Scale, inclined plane test, and footprint analysis, and spinal cord tissue damage and myelination were evaluated using HE and LFB staining. Nissl staining, immunofluorescence assay and Western blotting were used to observe surviving anterior horn motor neurons in injured spinal cord tissue. In cultured BV2 cells, the effects of linarin against lipopolysaccharide (LPS)­induced microglia activation, inflammatory factor release and signaling pathway changes were assessed with immunofluorescence staining, Western blotting, RT-qPCR, and ELISA. In a BV2 and HT22 cell co-culture system, Western blotting was performed to examine the effect of linarin against HT22 cell apoptosis mediated by LPS-induced microglia activation. RESULTS: Linarin treatment significantly improved locomotor function (P < 0.05), reduced spinal cord damage area, increased spinal cord myelination, and increased the number of motor neurons in the anterior horn of the SCI mice (P < 0.05). In both SCI mice and cultured BV2 cells, linarin effectively inhibited glial cell activation and suppressed the release of iNOS, COX-2, TNF-α, IL-6, and IL-1ß, resulting also in reduced neuronal apoptosis in SCI mice (P < 0.05). Western blotting suggested that linarin-induced microglial activation inhibition was mediated by inhibition of the TLR4/NF- κB signaling pathway. In the cell co-culture experiments, linarin treatment significantly decreased inflammation-mediated apoptosis of HT22 cells (P < 0.05). CONCLUSION: The neuroprotective effect of linarin is medicated by inhibition of microglia activation via suppressing the TLR4/NF­κB signaling pathway, which mitigates neural inflammation and reduce neuronal apoptosis to enhance motor function of the SCI mice.


Sujet(s)
Apoptose , Souris de lignée C57BL , Microglie , Facteur de transcription NF-kappa B , Transduction du signal , Traumatismes de la moelle épinière , Récepteur de type Toll-4 , Animaux , Souris , Traumatismes de la moelle épinière/traitement médicamenteux , Traumatismes de la moelle épinière/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Récepteur de type Toll-4/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neuroprotecteurs/pharmacologie , Coumarines/pharmacologie , Inflammation/métabolisme , Lipopolysaccharides , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/étiologie , Hétérosides
14.
Sci Rep ; 14(1): 21410, 2024 09 13.
Article de Anglais | MEDLINE | ID: mdl-39271781

RÉSUMÉ

Acute ischemic stroke (AIS) is a major global health concern due to its high mortality and disability rates. Hemorrhagic transformation, a common complication of AIS, leads to poor prognosis yet lacks effective treatments. Preclinical studies indicate that hyperbaric oxygen (HBO) treatment within 12 h of AIS onset alleviates ischemia/reperfusion injuries, including hemorrhagic transformation. However, clinical trials have yielded conflicting results, suggesting some underlying mechanisms remain unclear. In this study, we confirmed that HBO treatments beginning within 1 h post reperfusion significantly alleviated the haemorrhage and neurological deficits in hyperglycemic transient middle cerebral arterial occlusion (tMCAO) mice, partly due to the inhibition of the NLRP3 inflammasome-mediated pro-inflammatory response in microglia. Notably, reactive oxygen species (ROS) mediate the anti-inflammatory and protective effect of early HBO treatment, as edaravone and N-Acetyl-L-Cysteine (NAC), two commonly used antioxidants, reversed the suppressive effect of HBO treatment on NLRP3 inflammasome-mediated inflammation in microglia. Furthermore, NAC countered the protective effect of early HBO treatment in tMCAO mice with hyperglycemia. These findings support that early HBO treatment is a promising intervention for AIS, however, caution is warranted when combining antioxidants with HBO treatment. Further assessments are needed to clarify the role of antioxidants in HBO therapy for AIS.


Sujet(s)
Oxygénation hyperbare , Hyperglycémie , Microglie , Espèces réactives de l'oxygène , Animaux , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Oxygénation hyperbare/méthodes , Souris , Espèces réactives de l'oxygène/métabolisme , Hyperglycémie/métabolisme , Hyperglycémie/complications , Mâle , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Inflammasomes/métabolisme , Modèles animaux de maladie humaine , Accident vasculaire cérébral/thérapie , Accident vasculaire cérébral/métabolisme , Antioxydants/pharmacologie , Souris de lignée C57BL , Infarctus du territoire de l'artère cérébrale moyenne/thérapie , Édaravone/pharmacologie , Lésion d'ischémie-reperfusion/métabolisme
15.
J Neuroinflammation ; 21(1): 222, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39272155

RÉSUMÉ

Gulf War Illness (GWI) is a chronic multisymptom disorder that affects approximately 25-32% of Gulf War veterans and is characterized by a number of symptoms such as cognitive impairment, psychiatric disturbances, chronic fatigue and gastrointestinal distress, among others. While the exact etiology of GWI is unknown, it is believed to have been caused by toxic exposures encountered during deployment in combination with other factors such as stress. In the present study we sought to evaluate the hypothesis that exposure to the toxin permethrin could prime neuroinflammatory stress response and elicit psychiatric symptoms associated with GWI. Specifically, we developed a mouse model of GWI, to evaluate the effects of chronic permethrin exposure followed by unpredictable stress. We found that subjecting mice to 14 days of chronic permethrin exposure followed by 7 days of unpredictable stress resulted in the development of depression-like behavior. This behavioral change coincided with distinct alterations in the microglia phenotype, indicating microglial activation in the hippocampus. We revealed that blocking microglial activation through Gi inhibitory DREADD receptors in microglia effectively prevented the behavioral change associated with permethrin and stress exposure. To elucidate the transcriptional networks impacted within distinct microglia populations linked to depression-like behavior in mice exposed to both permethrin and stress, we conducted a single-cell RNA sequencing analysis using 21,566 single nuclei collected from the hippocampus of mice. For bioinformatics, UniCell Deconvolve was a pre-trained, interpretable, deep learning model used to deconvolve cell type fractions and predict cell identity across spatial datasets. Our bioinformatics analysis identified significant alterations in permethrin exposure followed by stress-associated microglia population, notably pathways related to neuronal development, neuronal communication, and neuronal morphogenesis, all of which are associated with neural synaptic plasticity. Additionally, we observed permethrin exposure followed by stress-mediated changes in signal transduction, including modulation of chemical synaptic transmission, regulation of neurotransmitter receptors, and regulation of postsynaptic neurotransmitter receptor activity, a known contributor to the pathophysiology of depression in a subset of the hippocampal pyramidal neurons in CA3 subregions. Our findings tentatively suggest that permethrin may prime microglia towards a state of inflammatory activation that can be triggered by psychological stressors, resulting in depression-like behavior and alterations of neural plasticity. These findings underscore the significance of synergistic interactions between multi-causal factors associated with GWI.


Sujet(s)
Dépression , Modèles animaux de maladie humaine , Microglie , Maladies neuro-inflammatoires , Perméthrine , Syndrome de la guerre du Golfe , Animaux , Perméthrine/toxicité , Souris , Syndrome de la guerre du Golfe/induit chimiquement , Syndrome de la guerre du Golfe/anatomopathologie , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Dépression/induit chimiquement , Dépression/étiologie , Maladies neuro-inflammatoires/induit chimiquement , Maladies neuro-inflammatoires/métabolisme , Mâle , Souris de lignée C57BL , Stress psychologique
16.
Cytokine ; 183: 156737, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39217915

RÉSUMÉ

BACKGROUND: Opioid activation of the microglia or macrophage Toll-like receptor 4 (TLR4) and associated inflammatory cytokine release are implicated in opioid-induced hyperalgesia and tolerance. The cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS-STING) signaling pathway, activated by double-stranded DNA including mitochondrial DNA (mtDNA), has emerged as another key mediator of inflammatory responses. This study tested the hypothesis that morphine induces immune inflammatory responses in microglia and macrophages involving TLR4 and cGAS-STING pathway. METHODS: BV2 microglia and Raw 264.7 (Raw) macrophage cells were exposed to morphine with and without a STING inhibitor (C176) for 6 h or TLR 4 inhibitor (TAK242) for 24 h. Western blotting and RT-qPCR analyses assessed TLR4, cGAS, STING, nuclear factor-kappa B (NF-κB), and pro-inflammatory cytokine expression. Morphine-induced mitochondria dysfunction was quantified by reactive oxygen species (ROS) release using MitoSOX, mtDNA release by immunofluorescence, and RT-qPCR. Polarization of BV2 and Raw cells was assessed by inducible nitric oxide (iNOS) and CD86 expression. The role of mtDNA on morphine-related inflammation was investigated by mtDNA depletion of the cells with ethidium bromide (EtBr) or cell transfection of mtDNA extracted from morphine-treated cells. RESULTS: Morphine significantly increased the expression of TLR4, cGAS, STING, p65 NF-κB, and cytokines (IL-6 and TNF-α) in BV2 and Raw cells. Morphine-induced mitochondrial dysfunction by increased ROS and mtDNA release; the increased iNOS and CD86 evidenced inflammatory M1-like phenotype polarization. TLR4 and STING inhibitors reduced morphine-induced cytokine release in both cell types. The transfection of mtDNA activated inflammatory signaling proteins, cytokine release, and polarization. Conversely, mtDNA depletion led to the reversal of these effects. CONCLUSION: Morphine activates the cGAS-STING pathway in macrophage cell types. Inhibition of the STING pathway can be an additional method to overcome immune cell inflammation-related morphine tolerance and opioid-induced hyperalgesia.


Sujet(s)
Inflammation , Macrophages , Protéines membranaires , Morphine , Nucleotidyltransferases , Transduction du signal , Récepteur de type Toll-4 , Récepteur de type Toll-4/métabolisme , Animaux , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Morphine/pharmacologie , Nucleotidyltransferases/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Inflammation/métabolisme , Cellules RAW 264.7 , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , ADN mitochondrial/métabolisme , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Espèces réactives de l'oxygène/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire
17.
CNS Neurosci Ther ; 30(9): e70015, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39218796

RÉSUMÉ

INTRODUCTION: Neuroinflammation and microglial activation-related dendritic injury contribute to the pathogenesis of Autism Spectrum Disorder (ASD). Previous studies show that Progranulin (PGRN) is a growth factor associated with inflammation and synaptic development, but the role of PGRN in autism and the mechanisms underlying changes in PGRN expression remain unclear. AIMS: To investigate the impact of PGRN in autism, we stereotactically injected recombinant PGRN into the hippocampus of ASD model rats. Additionally, we explored the possibility that sortilin may be the factor behind the alterations in PGRN by utilizing SORT1 knockdown. Ultimately, we aimed to identify potential targets for the treatment of autism. RESULTS: PGRN could alleviate inflammatory responses, protect neuronal dendritic spines, and ameliorate autism-like behaviors. Meanwhile, elevated expression of sortilin and decreased levels of PGRN were observed in both ASD patients and rats. Enhanced sortilin levels facilitated PGRN internalization into lysosomes. Notably, suppressing SORT1 expression amplified PGRN levels, lessened microglial activation, and mitigated inflammation, thereby alleviating autism-like behaviors. CONCLUSION: Collectively, our findings highlight elevated sortilin levels in ASD rat brains, exacerbating dendrite impairment by affecting PGRN expression. PGRN supplementation and SORT1 knockdown hold potential as therapeutic strategies for ASD.


Sujet(s)
Protéines adaptatrices du transport vésiculaire , Trouble autistique , Progranulines , Acide valproïque , Animaux , Femelle , Humains , Mâle , Rats , Protéines adaptatrices du transport vésiculaire/métabolisme , Protéines adaptatrices du transport vésiculaire/génétique , Trouble autistique/métabolisme , Trouble autistique/induit chimiquement , Trouble autistique/traitement médicamenteux , Épines dendritiques/effets des médicaments et des substances chimiques , Épines dendritiques/anatomopathologie , Épines dendritiques/métabolisme , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Progranulines/génétique , Rat Sprague-Dawley , Acide valproïque/pharmacologie
18.
CNS Neurosci Ther ; 30(9): e70028, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39218784

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Spinal cord injury (SCI) results in significant neurological deficits, and microglia play the critical role in regulating the immune microenvironment and neurological recovery. Protein lactylation has been found to modulate the function of immune cells. Therefore, this study aimed to elucidate the effects of glycolysis-derived lactate on microglial function and its potential neuroprotective mechanisms via lactylation after SCI. METHODS: Single-cell RNA sequencing (scRNA-seq) data were obtained from figshare to analyze cellular and molecular alterations within the spinal cord post-SCI, further focusing on the expression of microglia-related genes for cell sub-clustering, trajectory analysis, and glycolysis function analysis. We also evaluated the expression of lactylation-related genes in microglia between day 7 after SCI and sham group. Additionally, we established the mice SCI model and performed the bulk RNA sequencing in a time-dependent manner. The expression of glycolysis- and lactylation-related genes was evaluated, as well as the immune infiltration analysis based on the lactylation-related genes. Then, we investigated the bio-effects of lactate on the inflammation and polarization phenotype of microglia. Finally, adult male C57BL/6 mice were subjected to exercise first to increase lactate level, before SCI surgery, aiming to evaluate the protective effects of lactate-mediated lactylation of microglia-related proteins on SCI. RESULTS: scRNA-seq identified a subcluster of microglia, recombinant chemokine C-X3-C-motif receptor 1+ (CX3CR1+) microglia, which is featured by M1-like phenotype and increased after SCI. KEGG analysis revealed the dysfunctional glycolysis in microglia after SCI surgery, and AUCell analysis suggested that the decreased glycolysis an increased oxidative phosphorylation in CX3CR1+ microglia. Differential gene analysis suggested that several lactylation-related genes (Fabp5, Lgals1, Vim, and Nefl) were downregulated in CX3CR1+ microglia at day 7 after SCI, further validated by the results from bulk RNA sequencing. Immunofluorescence staining indicated the expression of lactate dehydrogenase A (LDHA) in CX3CR1+ microglia also decreased at day 7 after SCI. Cellular experiments demonstrated that the administration of lactate could increase the lactylation level and inhibit the pro-inflammatory phenotype in microglia. Functionally, exercise-mediated lactate production resulted in improved locomotor recovery and decreased inflammatory markers in SCI mice compared to SCI alone. CONCLUSIONS: In the subacute phase of SCI, metabolic remodeling in microglia may be key therapeutic targets to promote nerve regeneration, and lactate contributed to neuroprotection after SCI by influencing microglial lactylation and inflammatory phenotype, which offered a novel approach for therapeutic intervention.


Sujet(s)
Acide lactique , Souris de lignée C57BL , Microglie , Analyse de séquence d'ARN , Analyse sur cellule unique , Traumatismes de la moelle épinière , Animaux , Traumatismes de la moelle épinière/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Souris , Mâle , Acide lactique/métabolisme , Analyse de séquence d'ARN/méthodes , Neuroprotecteurs/pharmacologie , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/physiologie
19.
J Neuroinflammation ; 21(1): 215, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39218898

RÉSUMÉ

BACKGROUND: Alzheimer's disease (AD) features progressive neurodegeneration and microglial activation that results in dementia and cognitive decline. The release of soluble amyloid (Aß) oligomers into the extracellular space is an early feature of AD pathology. This can promote excitotoxicity and microglial activation. Microglia can adopt several activation states with various functional outcomes. Protective microglial activation states have been identified in response to Aß plaque pathology in vivo. However, the role of microglia and immune mediators in neurotoxicity induced by soluble Aß oligomers is unclear. Further, there remains a need to identify druggable molecular targets that promote protective microglial states to slow or prevent the progression of AD. METHODS: Hippocampal entorhinal brain slice culture (HEBSC) was employed to study mechanisms of Aß1-42 oligomer-induced neurotoxicity as well as the role of microglia. The roles of glutamate hyperexcitation and immune signaling in Aß-induced neurotoxicity were assessed using MK801 and neutralizing antibodies to the TNF-related apoptosis-inducing ligand (TRAIL) respectively. Microglial activation state was manipulated using Gi-hM4di designer receptor exclusively activated by designer drugs (DREADDs), microglial depletion with the colony-stimulating factor 1 receptor (CSF1R) antagonist PLX3397, and microglial repopulation (PLX3397 withdrawal). Proteomic changes were assessed by LC-MS/MS in microglia isolated from control, repopulated, or Aß-treated HEBSCs. RESULTS: Neurotoxicity induced by soluble Aß1-42 oligomers involves glutamatergic hyperexcitation caused by the proinflammatory mediator and death receptor ligand TRAIL. Microglia were found to have the ability to both promote and restrain Aß-induced toxicity. Induction of microglial Gi-signaling with hM4di to prevent pro-inflammatory activation blunted Aß neurotoxicity, while microglial depletion with CSF1R antagonism worsened neurotoxicity caused by Aß as well as TRAIL. HEBSCs with repopulated microglia, however, showed a near complete resistance to Aß-induced neurotoxicity. Comparison of microglial proteomes revealed that repopulated microglia have a baseline anti-inflammatory and trophic phenotype with a predicted pathway activation that is nearly opposite that of Aß-exposed microglia. mTORC2 and IRF7 were identified as potential targets for intervention. CONCLUSION: Microglia are key mediators of both protection and neurodegeneration in response to Aß. Polarizing microglia toward a protective state could be used as a preventative strategy against Aß-induced neurotoxicity.


Sujet(s)
Peptides bêta-amyloïdes , Microglie , Fragments peptidiques , Ligand TRAIL , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Peptides bêta-amyloïdes/toxicité , Peptides bêta-amyloïdes/métabolisme , Animaux , Fragments peptidiques/toxicité , Fragments peptidiques/métabolisme , Ligand TRAIL/métabolisme , Ligand TRAIL/toxicité , Souris , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cortex entorhinal/métabolisme , Cortex entorhinal/effets des médicaments et des substances chimiques , Cortex entorhinal/anatomopathologie , Techniques de culture d'organes
20.
Drug Dev Res ; 85(6): e22254, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39234934

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease. Syncytin-1 (Syn), an envelope glycoprotein encoded by the env gene of the human endogenous retrovirus-W family, has been resorted to be highly expressed in biopsies from the muscles from ALS patients; however, the specific regulatory role of Syn during ALS progression remains uncovered. In this study, C57BL/6 mice were injected with adeno-associated virus-overexpressing Syn, with or without Fasudil administration. The Syn expression was assessed by quantitative real-time polymerase chain reaction and immunohistochemistry analysis. The histological change of anterior tibial muscles was determined by hematoxylin-eosin staining. Qualitative ultrastructural analysis of electron micrographs obtained from lumbar spinal cords was carried out. Serum inflammatory cytokines were assessed by enzyme linked immunosorbent assay (ELISA) assay and motor function was recorded using Basso, Beattie, and Bresnahan (BBB) scoring, climbing test and treadmill running test. Immunofluorescence and western blot assays were conducted to examine microglial- and motor neurons-related proteins. Syn overexpression significantly caused systemic inflammatory response, muscle tissue lesions, and motor dysfunction in mice. Meanwhile, Syn overexpression promoted the impairment of motor neuron, evidenced by the damaged structure of the neurons and reduced expression of microtubule-associated protein 2, HB9, neuronal nuclei and neuron-specific enolase in Syn-induced mice. In addition, Syn overexpression greatly promoted the expression of CD16/CD32 and inducible nitric oxide synthase (M1 phenotype markers), and reduced the expression of CD206 and arginase 1 (M2 phenotype markers). Importantly, the above changes caused by Syn overexpression were partly abolished by Fasudil administration. This study provides evidence that Syn-activated microglia plays a pivotal role during the progression of ALS.


Sujet(s)
5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine , Souris de lignée C57BL , Microglie , Motoneurones , Animaux , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Motoneurones/effets des médicaments et des substances chimiques , Motoneurones/métabolisme , Souris , 5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine/analogues et dérivés , 5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine/pharmacologie , Produits du gène env , Sclérose latérale amyotrophique/traitement médicamenteux , Sclérose latérale amyotrophique/métabolisme , Protéines de la grossesse/métabolisme , Mâle , Cytokines/métabolisme , Modèles animaux de maladie humaine , Activité motrice/effets des médicaments et des substances chimiques , Moelle spinale/métabolisme , Moelle spinale/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE