Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673879

ABSTRACT

Reactive astrocytes are key players in HIV-associated neurocognitive disorders (HAND), and different types of reactive astrocytes play opposing roles in the neuropathologic progression of HAND. A recent study by our group found that gp120 mediates A1 astrocytes (neurotoxicity), which secrete proinflammatory factors and promote HAND disease progression. Here, by comparing the expression of A2 astrocyte (neuroprotective) markers in the brains of gp120 tgm mice and gp120+/α7nAChR-/- mice, we found that inhibition of alpha 7 nicotinic acetylcholine receptor (α7nAChR) promotes A2 astrocyte generation. Notably, kynurenine acid (KYNA) is an antagonist of α7nAChR, and is able to promote the formation of A2 astrocytes, the secretion of neurotrophic factors, and the enhancement of glutamate uptake through blocking the activation of α7nAChR/NF-κB signaling. In addition, learning, memory and mood disorders were significantly improved in gp120 tgm mice by intraperitoneal injection of kynurenine (KYN) and probenecid (PROB). Meanwhile, the number of A2 astrocytes in the mouse brain was significantly increased and glutamate toxicity was reduced. Taken together, KYNA was able to promote A2 astrocyte production and neurotrophic factor secretion, reduce glutamate toxicity, and ameliorate gp120-induced neuropathological deficits. These findings contribute to our understanding of the role that reactive astrocytes play in the development of HAND pathology and provide new evidence for the treatment of HAND via the tryptophan pathway.


Subject(s)
Astrocytes , Glutamic Acid , Kynurenine , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Mice , Kynurenine/metabolism , Kynurenic Acid/metabolism , Kynurenic Acid/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp120/toxicity , Signal Transduction/drug effects , Mice, Knockout , Probenecid/pharmacology , Mice, Inbred C57BL , Male , Brain/metabolism , Brain/pathology , Brain/drug effects , NF-kappa B/metabolism
2.
J Neuroinflammation ; 20(1): 87, 2023 Mar 30.
Article in English | MEDLINE | ID: mdl-36997969

ABSTRACT

Despite extensive astrocyte activation in patients suffering from HIV-associated neurocognitive disorders (HAND), little is known about the contribution of astrocytes to HAND neuropathology. Here, we report that the robust activation of neurotoxic astrocytes (A1 astrocytes) in the CNS promotes neuron damage and cognitive deficits in HIV-1 gp120 transgenic mice. Notably, knockout of α7 nicotinic acetylcholine receptors (α7nAChR) blunted A1 astrocyte responses, ultimately facilitating neuronal and cognitive improvement in the gp120tg mice. Furthermore, we provide evidence that Kynurenic acid (KYNA), a tryptophan metabolite with α7nAChR inhibitory properties, attenuates gp120-induced A1 astrocyte formation through the blockade of α7nAChR/JAK2/STAT3 signaling activation. Meanwhile, compared with gp120tg mice, mice fed with tryptophan showed dramatic improvement in cognitive performance, which was related to the inhibition of A1 astrocyte responses. These initial and determinant findings mark a turning point in our understanding of the role of α7nAChR in gp120-mediated A1 astrocyte activation, opening up new opportunities to control neurotoxic astrocyte generation through KYNA and tryptophan administration.


Subject(s)
HIV Infections , Kynurenic Acid , Mice , Animals , Kynurenic Acid/pharmacology , Kynurenic Acid/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Astrocytes/metabolism , Tryptophan/metabolism , HIV/metabolism , Mice, Transgenic , Neurocognitive Disorders/metabolism , HIV Infections/complications , HIV Infections/metabolism
3.
Biomedicines ; 10(10)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36289622

ABSTRACT

Despite the availability of antibiotics over the last several decades, excessive antibiotic treatments for bacterial sepsis and meningitis (BSM) in children may result in several adverse outcomes. Hematogenous pathogens may directly induce permeability increases in human brain microvascular endothelial cells (HBMECs) and blood-brain barrier (BBB) dysfunctions. Our preliminary studies demonstrated that the alpha7 nicotinic acetylcholine receptor (α7nAChR) played an important role in the pathogenesis of BSM, accompanied by increasing cytokine-inducible SH2-containing protein (CISH) at the transcriptome level, but it has remained unclear how α7nAChR-CISH works mechanistically. The study aims to explore the underlying mechanism of α7nAChR and CISH during E. coli-induced BSM in vitro (HBMECs) and in vivo (α7nAChR-KO mouse). We found that in the stage of E. coli K1-induced BBB disruptions, α7nAChR functioned as the key regulator that affects the integrity of HBMECs by activating the JAK2-STAT5 signaling pathway, while CISH inhibited JAK2-STAT5 activation and exhibited protective effects against E. coli infection. Notably, we first validated that the expression of CISH could be regulated by α7nAChR in HBMECs. In addition, we determined the protective effects of MLA (methyllycaconitine citrate) and MEM (memantine hydrochloride) (functioning as α7nAChR antagonists) on infected HBMECs and suggested that the α7nAChR-CISH axis could explain the protective effects of the two small-molecule compounds on E. coli-induced HBMECs injuries and BBB disruptions. In conclusion, we dissected the α7nAChR/CISH/JAK2/STAT5 axis as critical for the pathogenesis of E. coli-induced brain microvascular leakage and BBB disruptions and provided novel evidence for the development of α7nAChR antagonists in the prevention of pediatric E. coli BSM.

4.
Molecules ; 27(13)2022 Jun 29.
Article in English | MEDLINE | ID: mdl-35807435

ABSTRACT

Enterovirus 71 (EV71) is a dominant pathogenic agent that may cause severe central nervous system (CNS) diseases among infants and young children in the Asia-pacific. The inflammasome is closely implicated in EV71-induced CNS injuries through a series of signaling pathways. However, the activation pathway of NLRP3 inflammasome involved in EV71-mediated CNS injuries remains poorly defined. In the studies, EV71 infection, ERK1/2 phosphorylation, and activation of NLRP3 are abolished in glioblastoma cells with low vimentin expression by CRISPR/Cas9-mediated knockdown. PD098059, an inhibitor of p-ERK, remarkably blocks the vimentin-mediated ERK1/2 phosphorylation in EV71-infected cells. Nuclear translocation of NF-κB p65 is dependent on p-ERK in a time-dependent manner. Moreover, NLRP3 activation and caspase-1 production are limited in EV71-infected cells upon the caffeic acid phenethyl ester (CAPE) administration, an inhibitor of NF-κB, which contributes to the inflammasome regulation. In conclusion, these results suggest that EV71-mediated NLRP3 inflammasome could be activated via the VIM-ERK-NF-κB pathway, and the treatment of the dephosphorylation of ERK and NF-κB inhibitors is beneficial to host defense in EV71-infected CNS.


Subject(s)
Enterovirus , Glioblastoma , NF-kappa B , Vimentin , Child , Child, Preschool , Enterovirus/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/virology , Humans , Inflammasomes/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphorylation , Vimentin/genetics , Vimentin/metabolism
5.
Front Immunol ; 11: 551449, 2020.
Article in English | MEDLINE | ID: mdl-33123130

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic and relapsing intestinal inflammatory condition with no effective treatment. Probiotics have gained wide attention because of their outstanding advantages in intestinal health issues. In previous studies, a novel soluble protein, HM0539, which is derived from Lactobacillus rhamnosus GG (LGG), showed significant protective effects against murine colitis, but no clear precise mechanism for this effect was provided. In this study, we hypothesized that the protective function of HM0539 might be derived from its modulation of the TLR4/Myd88/NF-κB axis signaling pathway, which is a critical pathway widely involved in the modulation of inflammatory responses. To test this hypothesis, the underlying anti-inflammatory effects and associated mechanisms of HM0539 were determined both in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages and in dextran sulfate sodium (DSS)-induced murine colitis. Our results showed that HM0539 inhibited the expression of cyclooxygenase-2 (COX-2) and the expression inducible nitric oxide synthase (iNOS) by down-regulating the activation of their respective promoter, and as a result this inhibited the production of prostaglandin E2 (PGE2) and nitric oxide (NO). Meanwhile, we demonstrated that HM0539 could ultimately modulate the activation of distal NF-κB by reducing the activation of TLR4 and suppressing the transduction of MyD88. However, even though the overexpression of TLR4 or MyD88 obviously reversed the effect of HM0539 on LPS-induced inflammation, HM0539 still retained some anti-inflammatory activity. Consistent with the in vitro findings, we found that HM0539 inhibited to a great extent the production of inflammatory mediators associated with the suppression of the TLR4/Myd88/NF-κB axis activation in colon tissue. In conclusion, HM0539 was shown to be a promising anti-inflammatory agent, at least in part through its down-regulation of the TLR4-MyD88 axis as well as of the downstream MyD88-dependent activated NF-κB signaling, and hence might be considered as a potential therapeutic option for IBD.


Subject(s)
Lacticaseibacillus rhamnosus/immunology , Myeloid Differentiation Factor 88/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Mice , RAW 264.7 Cells
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(8): 1184-1191, 2020 Aug 30.
Article in Chinese | MEDLINE | ID: mdl-32895175

ABSTRACT

OBJECTIVE: To construct a HIV-1 gp120 transgenic mouse model (gp120+) with α7 nicotinic acetylcholine receptor (α7nAChR) gene knockout. METHODS: The α7nAChR gene knockout mice (α7R-/-) were crossed with HIV-1gp120 transgenic mice (gp120+) to generate F1 generation mice. We selected the F1 mice with the genotype of α7R+/-/gp120+ to mate to obtain the F2 mice. The genotypes of the F3 mice were identified by PCR, and the protein expressions in the double transgenic animal model was analyzed by immunohistochemistry. BV2 cells were treated with gp120 protein and α7nAChR inhibitor, and the expressions of IL-1ß and TNF-α were detected using ELISA. RESULTS: The results of PCR showed the bands of the expected size in F3 mice. Two F3 mice with successful double gene editing (α7R-/-/gp120+) were obtained, and immunohistochemistry showed that the brain tissue of the mice did not express α7 nAChR but with high gp120 protein expression. In the in vitro cell experiment, treatment with gp120 promoted the secretion of IL-1ß and TNF-α in BV2 cells, while inhibition of α7nAChR significantly decreased the expression of IL-1ß and TNF-α (P < 0.001). CONCLUSIONS: By mating gp120 Tg mice with α7R-/- mice, we obtained gp120 transgenic mice with α7nAChR gene deletion, which serve as a new animal model for exploring the role of α7nAChR in gp120-induced neurotoxicity.


Subject(s)
alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Disease Models, Animal , Glycoproteins , Mice , Mice, Knockout , Mice, Transgenic , Tumor Necrosis Factor-alpha
SELECTION OF CITATIONS
SEARCH DETAIL
...