Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
CNS Neurosci Ther ; 25(9): 976-986, 2019 09.
Article in English | MEDLINE | ID: mdl-30972969

ABSTRACT

AIM: Mitochondrial autophagy (mitophagy) clears damaged mitochondria and attenuates ischemic neuronal injury. Urolithin A (Uro-A) activates mitophagy in mammal cells and Caenorhabditis elegans. We explored neuroprotection of Uro-A against ischemic neuronal injury. METHODS: Mice were subjected to middle cerebral artery occlusion. The brain infarct and neurological deficit scores were measured. The N2a cells and primary cultured mice cortical neurons were subjected to oxygen-glucose deprivation and reperfusion (OGD/R). Uro-A was incubated during OGD/R, and cell injury was determined by MTT and LDH. Autophagosomes were visualized by transfecting mCherry-microtubule-associated protein 1 light chain 3 (LC3). The protein levels of LC3-II, p62, Translocase Of Inner Mitochondrial Membrane 23 (TIMM23), and cytochrome c oxidase subunit 4 isoform 1 (COX4I1) were detected by Western blot. The ER stress markers, activating transcription factor 6 (ATF6) and C/EBP homologous protein (CHOP), were determined by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: Urolithin A alleviated OGD/R-induced injury in N2a cells and neurons and reduced ischemic brain injury in mice. Uro-A reinforced ischemia-induced autophagy. Furthermore, Uro-A-conferred protection was abolished by 3-methyladenine, suggesting the requirement of autophagy for neuroprotection. However, mitophagy was not further activated by Uro-A. Instead, Uro-A attenuated OGD/R-induced ER stress, which was abolished by 3-methyladenosine. Additionally, neuroprotection was reversed by ER stress inducer. CONCLUSION: Urolithin A protected against ischemic neuronal injury by reinforcing autophagy rather than mitophagy. Autophagy activation by Uro-A attenuated ischemic neuronal death by suppressing ER stress.


Subject(s)
Autophagy/drug effects , Brain Ischemia/prevention & control , Coumarins/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Mitophagy/drug effects , Neuroprotective Agents/therapeutic use , Animals , Autophagy/physiology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Coumarins/pharmacology , Endoplasmic Reticulum Stress/physiology , Male , Mice , Mice, Inbred C57BL , Mitophagy/physiology , Neuroprotective Agents/pharmacology
2.
Sci Rep ; 6: 18694, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728971

ABSTRACT

T helper 9 (Th9) cells, a recently recognized Th cell subset, are involved in autoimmune diseases. We aimed to investigate the role of Th9/interleukin-9 (IL-9) in the pathogenesis of hepatic fibrosis. Th9 and Th17 cells were quantified in chronic hepatitis B (CHB) patients with hepatic fibrosis, HBV-associated liver cirrhosis (LC) patients and healthy controls (HC). The percentages of Th9 and Th17 cells, concentrations of IL-9 and IL-17, as well as expression of IL-17, TNF-α, IL-6, IL-4, IL-21, TGF-ß1 and IFN-γ were significantly increased in plasma of CHB and LC patients compared with those in HC. Splenic Th9 and Th17 cells, plasma concentrations and liver expression of IL-9 and IL-17A were significantly elevated in mice with hepatic fibrosis compared with controls. Neutralization of IL-9 in mice ameliorated hepatic fibrosis, attenuated the activation of hepatic stellate cells, reduced frequencies of Th9, Th17 and Th1 cells in spleen, and suppressed expression of IL-9, IL-17A, IFN-γ, TGF-ß1, IL-6, IL-4 and TNF-α in plasma and liver respectively. Our data suggest a deleterious role of Th9/IL-9 in increasing hepatic fibrosis and exacerbating disease endpoints, indicating that Th9/IL9 based immunotherapy may be a promising approach for treating hepatic fibrosis.


Subject(s)
Interleukin-9/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , T-Lymphocyte Subsets , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Adolescent , Adult , Aged , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Case-Control Studies , Cytokines/metabolism , Disease Models, Animal , Female , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/metabolism , Humans , Immunophenotyping , Interleukin-17/antagonists & inhibitors , Interleukin-17/blood , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-9/antagonists & inhibitors , Interleukin-9/blood , Interleukin-9/genetics , Liver Cirrhosis/pathology , Lymphocyte Count , Male , Mice , Middle Aged , RNA, Messenger/genetics , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult
3.
World J Gastroenterol ; 21(5): 1531-45, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-25663772

ABSTRACT

AIM: To investigate the effect of interleukin (IL)-22 on hepatic fibrosis in mice and the possible mechanism involved. METHODS: Liver fibrosis was induced in male BALB/c mice by CCl4. Recombinant IL-22 (rmIL-22) was administered intraperitoneally in CCl4-treated mice. Fibrosis was assessed by histology and Masson staining. The activation of hepatic stellate cells (HSCs) was investigated by analysis of α-smooth muscle actin expression. The frequencies of T helper (Th) 22 cells, Th17 cells and Th1 cells, the expression of inflammatory cytokines [IL-22, IL-17A, interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), IL-6, IL-1ß] and transcription factors [aryl hydrocarbon receptor (AHR), RAR-related orphan receptor (RORγt), T-bet] mRNA in the liver were investigated. In addition, the plasma levels of IL-22, IL-17A, IFN-γ, TNF-α, IL-6 and IL-1ß were evaluated. RESULTS: Significant elevations in circulating Th22 cells, Th17 cells, Th1 cells, IL-22, IL-17A, and IFN-γ were observed in the hepatic fibrosis group compared with the control group (P < 0.01). Treatment with rmIL-22 in mice with hepatic fibrosis ameliorated the severity of hepatic fibrosis, which was confirmed by lower hepatic fibrosis pathological scores (P < 0.01). RmIL-22 decreased the frequencies of Th22 cells (6.71% ± 0.97% vs 8.09% ± 0.74%, P < 0.01), Th17 cells (4.34% ± 0.37% vs 5.71% ± 0.24%, P < 0.01), Th1 cells (3.09% ± 0.49% vs 4.91% ± 0.73%, P < 0.01), and the levels of IL-22 (56.23 ± 3.08 vs 70.29 ± 3.01, P < 0.01), IL-17A (30.74 ± 2.77 vs 45.68 ± 2.71, P < 0.01), and IFN-γ (74.78 ± 2.61 vs 124.89 ± 2.82, P < 0.01). Down-regulation of IL-22, IL-17A, IFN-γ, TNF-α, IL-6, IL-1ß, AHR RORγt, and T-bet gene expression in the liver was observed in the rmIL-22 group (P < 0.01). CONCLUSION: The frequencies of Th22, Th17 and Th1 cells are elevated in hepatic fibrosis. RmIL-22 can attenuate HSC activation and down-regulate the levels of inflammatory cytokines, thereby ameliorating liver fibrogenesis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Hepatic Stellate Cells/drug effects , Inflammation Mediators/metabolism , Interleukins/pharmacology , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Animals , Carbon Tetrachloride , Cytokines/genetics , Cytokines/immunology , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Inflammation Mediators/immunology , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred BALB C , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Severity of Illness Index , Signal Transduction/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Time Factors , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...