Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(30): 15128-15133, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31289228

ABSTRACT

Immune cells infiltrate adipose tissues and provide a framework to regulate energy homeostasis. However, the precise underlying mechanisms and signaling by which the immune system regulates energy homeostasis in metabolic tissues remain poorly understood. Here, we show that the AT-rich interactive domain 5A (Arid5a), a cytokine-induced nucleic acid binding protein, is important for the maintenance of adipose tissue homeostasis. Long-term deficiency of Arid5a in mice results in adult-onset severe obesity. In contrast, transgenic mice overexpressing Arid5a are highly resistant to high-fat diet-induced obesity. Inhibition of Arid5a facilitates the in vitro differentiation of 3T3-L1 cells and fibroblasts to adipocytes, whereas its induction substantially inhibits their differentiation. Molecular studies reveal that Arid5a represses the transcription of peroxisome proliferator activated receptor gamma 2 (Ppar-γ2) due to which, in the absence of Arid5a, Ppar-γ2 is persistently expressed in fibroblasts. This phenomenon is accompanied by enhanced fatty acid uptake in Arid5a-deficient cells, which shifts metabolic homeostasis toward prolipid metabolism. Furthermore, we show that Arid5a and Ppar-γ2 are dynamically counterregulated by each other, hence maintaining adipogenic homeostasis. Thus, we show that Arid5a is an important negative regulator of energy metabolism and can be a potential target for metabolic disorders.


Subject(s)
Adipogenesis/genetics , Adipose Tissue/metabolism , DNA-Binding Proteins/genetics , Feedback, Physiological , Obesity/genetics , PPAR gamma/genetics , Transcription Factors/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Biological Transport , Cell Differentiation , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Fatty Acids/metabolism , Female , Gene Expression Regulation , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Obesity/pathology , PPAR gamma/metabolism , Signal Transduction , Transcription Factors/metabolism
2.
PLoS One ; 14(4): e0215981, 2019.
Article in English | MEDLINE | ID: mdl-31026283

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is a widespread neurological autoimmune disease that includes episodes of demyelination in the central nervous system (CNS). The accumulated evidence has suggested that aryl hydrocarbon receptor (Ahr), a ligand-activated transcription factor, is a promising treatment target for MS. Thus, the current study aimed to identify a novel Ahr ligand with anti-inflammatory potential in experimental autoimmune encephalomyelitis (EAE). METHODS: An in silico analysis was carried out to predict interactions between Ahr and potential natural ligands. The effects of a predicted interaction were examined in vitro using CD4+ T cells under T helper17 (Th17) cell-polarizing conditions and lipopolysaccharide (LPS)-stimulated macrophages. Silencing Ahr and microRNA (miR)-132 was achieved by electroporation. Myelin oligodendrocyte glycoprotein (MOG)35-55 and the adoptive transfer of encephalitogenic CD4+ T cells were used to induce EAE. RESULTS: Molecular docking analysis and in vitro data identified gallic acid (GA) as a novel Ahr ligand with potent activation potential. GA induced the expression of Ahr downstream genes, including cytochrome P450 family 1 subfamily A member 1 (Cyp1a1) and the miR-212/132 cluster, and promoted the formation of the Ahr/Ahr nuclear translocator (Arnt) complex. In vivo, GA-treated mice were resistant to EAE and exhibited reduced levels of proinflammatory cytokines and increased levels of transforming growth factor-ß (TGF-ß). Furthermore, GA reduced infiltration of CD4+CD45+ T cells and monocytes into the CNS. The anti-inflammatory effects of GA were concomitant with miR-132-potentiated cholinergic anti-inflammation and the regulation of the pathogenic potential of astrocytes and microglia. Inducing EAE by adoptive transfer revealed that CD4+ T cells were not entirely responsible for the ameliorative effects of GA. CONCLUSION: Our findings identify GA as a novel Ahr ligand and provide molecular mechanisms elucidating the ameliorative effects of GA on EAE, suggesting that GA is a potential therapeutic agent to control inflammation in autoimmune diseases such as MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Animals , Astrocytes/pathology , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Gene Expression Regulation/drug effects , Humans , Ligands , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Microglia/pathology , Protein Domains , Receptors, Aryl Hydrocarbon/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL