Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Immunity ; 56(12): 2773-2789.e8, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37992711

ABSTRACT

Although the gut microbiota can influence central nervous system (CNS) autoimmune diseases, the contribution of the intestinal epithelium to CNS autoimmunity is less clear. Here, we showed that intestinal epithelial dopamine D2 receptors (IEC DRD2) promoted sex-specific disease progression in an animal model of multiple sclerosis. Female mice lacking Drd2 selectively in intestinal epithelial cells showed a blunted inflammatory response in the CNS and reduced disease progression. In contrast, overexpression or activation of IEC DRD2 by phenylethylamine administration exacerbated disease severity. This was accompanied by altered lysozyme expression and gut microbiota composition, including reduced abundance of Lactobacillus species. Furthermore, treatment with N2-acetyl-L-lysine, a metabolite derived from Lactobacillus, suppressed microglial activation and neurodegeneration. Taken together, our study indicates that IEC DRD2 hyperactivity impacts gut microbial abundances and increases susceptibility to CNS autoimmune diseases in a female-biased manner, opening up future avenues for sex-specific interventions of CNS autoimmune diseases.


Subject(s)
Autoimmune Diseases of the Nervous System , Multiple Sclerosis , Male , Female , Mice , Animals , Multiple Sclerosis/metabolism , Disease Models, Animal , Signal Transduction , Disease Progression , Receptors, Dopamine
2.
J Neuroinflammation ; 20(1): 203, 2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37674228

ABSTRACT

Astrocytes contribute to chronic neuroinflammation in a variety of neurodegenerative diseases, including Parkinson's disease (PD), the most common movement disorder. However, the precise role of astrocytes in neuroinflammation remains incompletely understood. Herein, we show that regulator of G-protein signaling 5 (RGS5) promotes neurodegenerative process through augmenting astrocytic tumor necrosis factor receptor (TNFR) signaling. We found that selective ablation of Rgs5 in astrocytes caused an inhibition in the production of cytokines resulting in mitigated neuroinflammatory response and neuronal survival in animal models of PD, whereas overexpression of Rgs5 had the opposite effects. Mechanistically, RGS5 switched astrocytes from neuroprotective to pro-inflammatory property via binding to the receptor TNFR2. RGS5 also augmented TNFR signaling-mediated pro-inflammatory response by interacting with the receptor TNFR1. Moreover, interrupting RGS5/TNFR interaction by either RGS5 aa 1-108 or small molecular compounds feshurin and butein, suppressed astrocytic cytokine production. We showed that the transcription of astrocytic RGS5 was controlled by transcription factor early B cell factor 1 whose expression was reciprocally influenced by RGS5-modulated TNF signaling. Thus, our study indicates that beyond its traditional role in G-protein coupled receptor signaling, astrocytic RGS5 is a key modulator of TNF signaling circuit with resultant activation of astrocytes thereby contributing to chronic neuroinflammation. Blockade of the astrocytic RGS5/TNFR interaction is a potential therapeutic strategy for neuroinflammation-associated neurodegenerative diseases.


Subject(s)
Neuroinflammatory Diseases , RGS Proteins , Animals , Astrocytes , Signal Transduction , RGS Proteins/genetics , Inflammation
4.
Proc Natl Acad Sci U S A ; 120(8): e2210643120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36795751

ABSTRACT

Microglia play a critical role in the pathogenic process of neurodegenerative diseases, such as Parkinson's disease (PD) and Alzheimer's disease (AD). Upon pathological stimulation, microglia are converted from a surveillant to an overactivated phenotype. However, the molecular characters of proliferating microglia and their contributions to the pathogenesis of neurodegeneration remain unclear. Here, we identify chondroitin sulfate proteoglycan 4 (Cspg4, also known as neural/glial antigen 2)-expressing microglia as a specific subset of microglia with proliferative capability during neurodegeneration. We found that the percentage of Cspg4+ microglia was increased in mouse models of PD. The transcriptomic analysis of Cspg4+ microglia revealed that the subcluster Cspg4high microglia displayed a unique transcriptomic signature, which was characterized by the enrichment of orthologous cell cycle genes and a lower expression of genes responsible for neuroinflammation and phagocytosis. Their gene signatures were also distinct from that of known disease-associated microglia. The proliferation of quiescent Cspg4high microglia was evoked by pathological α-synuclein. Following the transplantation in the adult brain with the depletion of endogenous microglia, Cspg4high microglia grafts showed higher survival rates than their Cspg4- counterparts. Consistently, Cspg4high microglia were detected in the brain of AD patients and displayed the expansion in animal models of AD. These findings suggest that Cspg4high microglia are one of the origins of microgliosis during neurodegeneration and may open up a avenue for the treatment of neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Parkinson Disease , Mice , Animals , Microglia/metabolism , Parkinson Disease/metabolism , Alzheimer Disease/metabolism , Neurodegenerative Diseases/metabolism , Phagocytosis
5.
J Exp Med ; 219(9)2022 09 05.
Article in English | MEDLINE | ID: mdl-35877595

ABSTRACT

Astrocyte activation is associated with progressive inflammatory demyelination in multiple sclerosis (MS). The molecular mechanisms underlying astrocyte activation remain incompletely understood. Recent studies have suggested that classical neurotransmitter receptors are implicated in the modulation of brain innate immunity. We investigated the role of dopamine signaling in the process of astrocyte activation. Here, we show the upregulation of dopamine D2 receptor (DRD2) in reactive astrocytes in MS brain and noncanonical role of astrocytic DRD2 in MS pathogenesis. Mice deficient in astrocytic Drd2 exhibit a remarkable suppression of reactive astrocytes and amelioration of experimental autoimmune encephalomyelitis (EAE). Mechanistically, DRD2 regulates the expression of 6-pyruvoyl-tetrahydropterin synthase, which modulates NF-κB activity through protein kinase C-δ. Pharmacological blockade of astrocytic DRD2 with a DRD2 antagonist dehydrocorybulbine remarkably inhibits the inflammatory response in mice lacking neuronal Drd2. Together, our findings reveal previously an uncharted role for DRD2 in astrocyte activation during EAE-associated CNS inflammation. Its therapeutic inhibition may provide a potent lever to alleviate autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Astrocytes/metabolism , Disease Models, Animal , Inflammation/pathology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Receptors, Dopamine D2/metabolism
6.
Neurosci Res ; 180: 72-82, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35257836

ABSTRACT

Parkinson's disease (PD) is an age-related neurodegenerative disease, mainly characterized by the loss of dopaminergic (DA) neurons in the substantia nigra. Several non-motor symptoms, including those associated with gastrointestinal dysfunction, precede the classical motor symptoms in PD. However, the mechanisms underlying gastrointestinal dysfunction in the prodromal phase of PD remain elusive. Here, we investigated the contribution of the central DA system to cell proliferation in the colonic epithelium. Degeneration of nigrostriatal DA pathway induced by striatal 6-hydroxydopamine (6-OHDA) injection resulted in a marked reduction in cell proliferation in the colonic epithelium as assessed by Ki-67 and bromodeoxyuridine labeling assays. RNA-sequencing analysis confirmed the suppression of cell cycle-related gene expression in the colonic epithelium of 6-OHDA-lesioned mice. Mesencephalic DA neuron degeneration also caused the gut microbiota dysbiosis. Moreover, 6-OHDA-lesioned mice showed profoundly increased vulnerability to dextran sulfate sodium-induced colitis. Together, our study uncovers a crucial role for the integrity of nigral DA neurons in the maintenance of colonic epithelial cell homeostasis. Our data also provide a new strategy for protecting intestinal homeostasis in PD.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Animals , Cell Proliferation , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Epithelium/metabolism , Mice , Neurodegenerative Diseases/metabolism , Oxidopamine , Substantia Nigra/metabolism
7.
Cell Rep ; 35(7): 109127, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34010636

ABSTRACT

The hippocampus is one of two niches in the mammalian brain with persistent neurogenesis into adulthood. The neurogenic capacity of hippocampal neural stem cells (NSCs) declines with age, but the molecular mechanisms of this process remain unknown. In this study, we find that fibroblast growth factor 13 (FGF13) is essential for the post-natal neurogenesis in mouse hippocampus, and FGF13 deficiency impairs learning and memory. In particular, we find that FGF13A, the nuclear isoform of FGF13, is involved in the maintenance of NSCs and the suppression of neuronal differentiation during post-natal hippocampal development. Furthermore, we find that FGF13A interacts with ARID1B, a unit of Brahma-associated factor chromatin remodeling complex, and suppresses the expression of neuron differentiation-associated genes through chromatin modification. Our results suggest that FGF13A is an important regulator for maintaining the self-renewal and neurogenic capacity of NSCs in post-natal hippocampus, revealing an epigenomic regulatory function of FGFs in neurogenesis.


Subject(s)
Epigenomics/methods , Hippocampus/metabolism , Neurogenesis/genetics , Protein Isoforms/metabolism , Animals , Cell Differentiation , Cell Proliferation , Humans , Mice
8.
BMC Med ; 17(1): 204, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31727112

ABSTRACT

BACKGROUND: Brain innate immunity is vital for maintaining normal brain functions. Immune homeostatic imbalances play pivotal roles in the pathogenesis of neurological diseases including Parkinson's disease (PD). However, the molecular and cellular mechanisms underlying the regulation of brain innate immunity and their significance in PD pathogenesis are still largely unknown. METHODS: Cre-inducible diphtheria toxin receptor (iDTR) and diphtheria toxin-mediated cell ablation was performed to investigate the impact of neuron-glial antigen 2 (NG2) glia on the brain innate immunity. RNA sequencing analysis was carried out to identify differentially expressed genes in mouse brain with ablated NG2 glia and lipopolysaccharide (LPS) challenge. Neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice were used to evaluate neuroinflammatory response in the presence or absence of NG2 glia. The survival of dopaminergic neurons or glial cell activation was evaluated by immunohistochemistry. Co-cultures of NG2 glia and microglia were used to examine the influence of NG2 glia to microglial activation. RESULTS: We show that NG2 glia are required for the maintenance of immune homeostasis in the brain via transforming growth factor-ß2 (TGF-ß2)-TGF-ß type II receptor (TGFBR2)-CX3C chemokine receptor 1 (CX3CR1) signaling, which suppresses the activation of microglia. We demonstrate that mice with ablated NG2 glia display a profound downregulation of the expression of microglia-specific signature genes and remarkable inflammatory response in the brain following exposure to endotoxin lipopolysaccharides. Gain- or loss-of-function studies show that NG2 glia-derived TGF-ß2 and its receptor TGFBR2 in microglia are key regulators of the CX3CR1-modulated immune response. Furthermore, deficiency of NG2 glia contributes to neuroinflammation and nigral dopaminergic neuron loss in MPTP-induced mouse PD model. CONCLUSIONS: These findings suggest that NG2 glia play a critical role in modulation of neuroinflammation and provide a compelling rationale for the development of new therapeutics for neurological disorders.


Subject(s)
Antigens/physiology , Brain/immunology , Immunity, Innate , Neuroglia/physiology , Parkinson Disease/immunology , Proteoglycans/physiology , Receptor, Transforming Growth Factor-beta Type II/metabolism , Transforming Growth Factor beta2/metabolism , Animals , Brain/metabolism , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Dopaminergic Neurons/physiology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Microglia/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction
9.
Transl Neurodegener ; 8: 3, 2019.
Article in English | MEDLINE | ID: mdl-30675347

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is characterized by a chronic loss of dopaminergic neurons and the presence of proteinaceous inclusions (Lewy bodies) within some remaining neurons in the substantia nigra. Recently, astroglial inclusion body has also been found in some neurodegenerative diseases including PD. However, the underlying molecular mechanisms of how astroglial protein aggregation forms remain largely unknown. Here, we investigated the contribution of αB-crystallin (CRYAB), a small heat shock protein, in α-synuclein inclusion formation in astrocytes. METHODS: Small interfering RNA (siRNA)-mediated CRYAB (siCRYAB) knockdown or CRYAB overexpression was performed to investigate the impact of CRYAB on the autophagy in human glioblastoma cell line U251 cells. Co-immunoprecipitation (co-IP) and immunoblotting were used to dissect the interaction among multiple proteins. The clearance of α-synuclein in vitro was evaluated by immunocytochemistry. CRYAB transgenic mice and transgenic mice overexpressing A30P mutant form of human α-synuclein were used to examine the influence of CRYAB to α-synuclein accumulation in vivo. RESULTS: We found that knockdown of CRYAB in U251 cells or primary cultured astrocytes resulted in a marked augmentation of autophagy activity. In contrast, exogenous CRYAB disrupted the assembly of the BAG3-HSPB8-HSC70 complex via binding with BAG3, thereby suppressing the autophagy activity. Furthermore, CRYAB-regulated autophagy has relevance to PD pathogenesis. Knockdown of CRYAB remarkably promoted cytoplasmic clearance of α-synuclein preformed fibrils (PFFs). Conversely, selective overexpression of CRYAB in astrocytes markedly suppressed autophagy leading to the accumulation of α-synuclein aggregates in the brain of transgenic mice expressing human α-synuclein A30P mutant. CONCLUSIONS: This study reveals a novel function for CRYAB as a natural inhibitor of astrocytic autophagy and shows that knockdown of CYRAB may provide a therapeutic target against proteinopathies such as synucleinopathies.

10.
Biochem Biophys Res Commun ; 508(1): 282-288, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30497777

ABSTRACT

Neuroinflammation is considered a challenging clinical problem. Chronic inflammatory responses play important roles in the onset and progression of various neurodegenerative diseases, including multiple sclerosis (MS). Previous studies have shown that astrocytes express small heat shock protein αB-crystallin (CRYAB) which is capable of inhibiting inflammatory responses in astrocytes per se. However, the underlying mechanisms of CRYAB-induced modulation of neuroinflammation are still not fully understood. In the present study, we investigated the role of extracellular CRYAB in the interaction between microglia and astrocytes in the context of MS-associated neuroinflammation. We found that the expression of CRYAB was profoundly increased in EAE mice. CRYAB was preferentially expressed in astrocytes and could be secreted via exosomes. Levels of exosomal CRYAB secreted from astrocytes were markedly increased under stress conditions. Furthermore, incubation of immortalized astrocytes or microglia cell lines with CRYAB remarkably suppressed astrocytes and microglia-mediated inflammatory responses in both autocrine and paracrine manners. Our results reveal a novel function for extracellular CRYAB in the regulation of neuroinflammation. Targeting extracellular CRYAB-modulated neuroinflammation is a potential therapeutic intervention for MS.


Subject(s)
Inflammation/metabolism , alpha-Crystallin B Chain/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Inflammation/chemically induced , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism
12.
Carbohydr Polym ; 98(1): 861-9, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23987422

ABSTRACT

The inclusion complexation behavior, characterization and binding ability of naringenin with ß-cyclodextrin and its derivatives were investigated in both solution and the solid state by means of XRD, DSC, SEM, (1)H and 2D NMR and UV-vis spectroscopy. The results showed that the water solubility and thermal stability of naringenin were obviously increased in the inclusion complex with cyclodextrins. This satisfactory water solubility and high thermal stability of the naringenin/CD complexes will be potentially useful for their application as herbal medicines or healthcare products.


Subject(s)
Flavanones/chemistry , Water/chemistry , beta-Cyclodextrins/chemistry , Biological Availability , Flavanones/pharmacokinetics , Solubility
13.
Nature ; 494(7435): 90-4, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23242137

ABSTRACT

Chronic neuroinflammation is a common feature of the ageing brain and some neurodegenerative disorders. However, the molecular and cellular mechanisms underlying the regulation of innate immunity in the central nervous system remain elusive. Here we show that the astrocytic dopamine D2 receptor (DRD2) modulates innate immunity through αB-crystallin (CRYAB), which is known to suppress neuroinflammation. We demonstrate that knockout mice lacking Drd2 showed remarkable inflammatory response in multiple central nervous system regions and increased the vulnerability of nigral dopaminergic neurons to neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity. Astrocytes null for Drd2 became hyper-responsive to immune stimuli with a marked reduction in the level of CRYAB. Preferential ablation of Drd2 in astrocytes robustly activated astrocytes in the substantia nigra. Gain- or loss-of-function studies showed that CRYAB is critical for DRD2-mediated modulation of innate immune response in astrocytes. Furthermore, treatment of wild-type mice with the selective DRD2 agonist quinpirole increased resistance of the nigral dopaminergic neurons to MPTP through partial suppression of inflammation. Our study indicates that astrocytic DRD2 activation normally suppresses neuroinflammation in the central nervous system through a CRYAB-dependent mechanism, and provides a new strategy for targeting the astrocyte-mediated innate immune response in the central nervous system during ageing and disease.


Subject(s)
Astrocytes/immunology , Astrocytes/metabolism , Inflammation/immunology , Receptors, Dopamine D2/metabolism , alpha-Crystallin B Chain/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Astrocytes/drug effects , Dopaminergic Neurons/drug effects , Immunity, Innate/drug effects , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/immunology , Neuroprotective Agents/metabolism , Quinpirole/pharmacology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/deficiency , Receptors, Dopamine D2/genetics , Substantia Nigra/cytology , Substantia Nigra/drug effects , alpha-Crystallin B Chain/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...