Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Neuropharmacol ; 20(5): 983-997, 2022.
Article in English | MEDLINE | ID: mdl-34370639

ABSTRACT

BACKGROUND: High methionine-diet (HMD) causes Alzheimer's disease (AD)-like symptoms. Previous studies have shown that Dendrobium nobile Lindle. alkaloids (DNLA) have potential benefits for AD Object: The objective of this study has been to explore whether DNLA can improve AD-like symptoms induced by HMD. METHODS: Mice were fed with 2% HMD diet for 11 weeks; the DNLA20 control group (20 mg/kg), DNLA10 group (10 mg/kg), and DNLA20 group (20 mg/kg) were administered DNLA for 3 months. Morris water maze test was used to detect learning and memory ability. Neuron damage was evaluated by HE and Nissl staining. Levels of homocysteine (Hcy), beta-amyloid 1-42 (Aß1-42), S-adenosine methionine (SAM) and S-adenosine homocysteine (SAH) were detected by ELISA. Immunofluorescence and western blotting (WB) were used to determine the expression of proteins. CPG island methylation levels were accessed by Methylation-specific PCR (MSP) and MethylTarget methylation detection. RESULTS: Morris water maze test revealed that DNLA improved learning and memory dysfunction. HE, Nissl, and immunofluorescence staining showed that DNLA alleviated neuron damage and reduced the 5-methylcytosine (5-mC), Aß1-40) and Aß1-42) levels. DNLA also decreased the levels of Hcy and Aß1-42) in the serum, along with decreasing SAM/SAH level in the liver tissue. WB results showed that DNLA down-regulated the expression of amyloid-precursor protein (APP), presenilin-1 (PS1), beta-secretase-1 (BACE1), DNA methyltransferase1 (DNMT1), Aß1-40) and Aß1-42) proteins. DNLA also up-regulated the proteins expression of insulin-degrading enzyme (IDE), neprilysin (NEP), DNMT3a and DNMT3b. Meanwhile, DNLA increased CPG island methylation levels of APP and BACE1 genes. CONCLUSION: DNLA alleviated AD-like symptoms induced by HMD via the DNA methylation pathway.


Subject(s)
Alkaloids , Alzheimer Disease , Dendrobium , Adenosine , Alkaloids/pharmacology , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases , Dendrobium/chemistry , Diet , Disease Models, Animal , Homocysteine , Methionine/adverse effects , Mice
2.
Neural Regen Res ; 16(12): 2499-2504, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33907040

ABSTRACT

Microglial cells are important resident innate immune components in the central nervous system that are often activated during neuroinflammation. Activated microglia can display one of two phenotypes, M1 or M2, which each play distinct roles in neuroinflammation. Rutin, a dietary flavonoid, exhibits protective effects against neuroinflammation. However, whether rutin is able to influence the M1/M2 polarization of microglia remains unclear. In this study, in vitro BV-2 cell models of neuroinflammation were established using 100 ng/mL lipopolysaccharide to investigate the effects of 1-hour rutin pretreatment on microglial polarization. The results revealed that rutin pretreatment reduced the expression of the proinflammatory cytokines tumor necrosis factor-α, interleukin-1ß, and interleukin-6 and increased the secretion of interleukin-10. Rutin pretreatment also downregulated the expression of the M1 microglial markers CD86 and inducible nitric oxide synthase and upregulated the expression of the M2 microglial markers arginase 1 and CD206. Rutin pretreatment inhibited the expression of Toll-like receptor 4 and myeloid differentiation factor 88 and blocked the phosphorylation of I kappa B kinase and nuclear factor-kappa B. These results showed that rutin pretreatment may promote the phenotypic switch of microglia M1 to M2 by inhibiting the Toll-like receptor 4/nuclear factor-kappa B signaling pathway to alleviate lipopolysaccharide-induced neuroinflammation.

3.
Diabetes Metab Syndr Obes ; 14: 431-442, 2021.
Article in English | MEDLINE | ID: mdl-33564250

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a metabolic disease characterized by hyperglycemia which is caused by insufficient insulin secretion or insulin resistance. Interaction of genetic, epigenetic and environmental factors plays a significant role in the development of T2DM. Several environmental factors including diet and lifestyle, as well as age have been associated with an increased risk for T2DM. It has been demonstrated that these environmental factors may affect global epigenetic status, and alter the expression of susceptible genes, thereby contributing to the pathogenesis of T2DM. In recent years, a growing body of molecular and genetic studies in diabetes have been focused on the ways to restore the numbers or function of ß-cells in order to reverse a range of metabolic consequences of insulin deficiency. The pancreatic duodenal homeobox 1 (PDX-1) is a transcriptional factor that is essential for the development and function of islet cells. A number of studies have shown that there is a significant increase in the level of DNA methylation of PDX-1 resulting in reduced activity in T2DM islets. The decrease in PDX-1 activity may be a critical mediator causing dysregulation of pancreatic ß cells in T2DM. This article reviews the epigenetic mechanisms of PDX-1 involved in T2DM, focusing on diabetes and DNA methylation, and discusses some potential strategies for the application of PDX-1 in the treatment of diabetes.

4.
J Neuroinflammation ; 17(1): 292, 2020 Oct 07.
Article in English | MEDLINE | ID: mdl-33028343

ABSTRACT

BACKGROUND: Sepsis-associated encephalopathy (SAE) is an early and frequent event of infection-induced systemic inflammatory response syndrome. Phosphoinositide 3-kinase γ (PI3Kγ) is linked to neuroinflammation and inflammation-related microglial activity. In homeotherms, variations in ambient temperature (Ta) outside the thermoneutral zone lead to thermoregulatory responses, mainly driven by a gradually increasing sympathetic activity, and may affect disease severity. We hypothesized that thermoregulatory response to hypothermia (reduced Ta) aggravates SAE in PI3Kγ-dependent manner. METHODS: Experiments were performed in wild-type, PI3Kγ knockout, and PI3Kγ kinase-dead mice, which were kept at neutral (30 ± 0.5 °C) or moderately lowered (26 ± 0.5 °C) Ta. Mice were exposed to lipopolysaccharide (LPS, 10 µg/g, from Escherichia coli serotype 055:B5, single intraperitoneal injection)-evoked systemic inflammatory response (SIR) and monitored 24 h for thermoregulatory response and blood-brain barrier integrity. Primary microglial cells and brain tissue derived from treated mice were analyzed for inflammatory responses and related cell functions. Comparisons between groups were made with one-way or two-way analysis of variance, as appropriate. Post hoc comparisons were made with the Holm-Sidak test or t tests with Bonferroni's correction for adjustments of multiple comparisons. Data not following normal distribution was tested with Kruskal-Wallis test followed by Dunn's multiple comparisons test. RESULTS: We show that a moderate reduction of ambient temperature triggers enhanced hypothermia of mice undergoing LPS-induced systemic inflammation by aggravated SAE. PI3Kγ deficiency enhances blood-brain barrier injury and upregulation of matrix metalloproteinases (MMPs) as well as an impaired microglial phagocytic activity. CONCLUSIONS: Thermoregulatory adaptation in response to ambient temperatures below the thermoneutral range exacerbates LPS-induced blood-brain barrier injury and neuroinflammation. PI3Kγ serves a protective role in suppressing release of MMPs, maintaining microglial motility and reinforcing phagocytosis leading to improved brain tissue integrity. Thus, preclinical research targeting severe brain inflammation responses is seriously biased when basic physiological prerequisites of mammal species such as preferred ambient temperature are ignored.


Subject(s)
Body Temperature Regulation/physiology , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Lipopolysaccharides/toxicity , Sepsis-Associated Encephalopathy/enzymology , Sepsis-Associated Encephalopathy/physiopathology , Animals , Animals, Newborn , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/physiopathology , Body Temperature/drug effects , Body Temperature/physiology , Body Temperature Regulation/drug effects , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sepsis-Associated Encephalopathy/chemically induced
5.
Front Immunol ; 10: 2492, 2019.
Article in English | MEDLINE | ID: mdl-31781091

ABSTRACT

Trained immunity and immune tolerance have been identified as long-term response patterns of the innate immune system. The causes of these opposing reactions remain elusive. Here, we report about differential inflammatory responses of microglial cells derived from neonatal mouse brain to increasing doses of the endotoxin LPS. Prolonged priming with ultra-low LPS doses provokes trained immunity, i.e., increased production of pro-inflammatory mediators in comparison to the unprimed control. In contrast, priming with high doses of LPS induces immune tolerance, implying decreased production of inflammatory mediators and pronounced release of anti-inflammatory cytokines. Investigation of the signaling processes and cell functions involved in these memory-like immune responses reveals the essential role of phosphoinositide 3-kinase γ (PI3Kγ), one of the phosphoinositide 3-kinase species highly expressed in innate immune cells. Together, our data suggest profound influence of preceding contacts with pathogens on the immune response of microglia. The impact of these interactions-trained immunity or immune tolerance-appears to be shaped by pathogen dose.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/immunology , Microglia/enzymology , Microglia/immunology , Animals , Animals, Newborn , Brain/cytology , Brain/enzymology , Brain/immunology , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Class Ib Phosphatidylinositol 3-Kinase/genetics , Cytokines/metabolism , Immune Tolerance/drug effects , Immunity, Innate/drug effects , Immunologic Memory/drug effects , Immunologic Memory/physiology , Inflammation Mediators/metabolism , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects
6.
Cell Rep ; 27(3): 835-846.e5, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995480

ABSTRACT

Interleukin-38 (IL-38) is a cytokine of the IL-1 family with a role in chronic inflammation. However, its main cellular targets and receptors remain obscure. IL-38 is highly expressed in the skin and downregulated in psoriasis patients. We report an investigation in cellular targets of IL-38 during the progression of imiquimod-induced psoriasis. In this model, IL-38 knockout (IL-38 KO) mice show delayed disease resolution with exacerbated IL-17-mediated inflammation, which is reversed by the administration of mature IL-38 or γδ T cell-receptor-blocking antibodies. Mechanistically, X-linked IL-1 receptor accessory protein-like 1 (IL1RAPL1) is upregulated upon γδ T cell activation to feedforward-amplify IL-17 production and is required for IL-38 to suppress γδ T cell IL-17 production. Accordingly, psoriatic IL1RAPL1 KO mice show reduced inflammation and IL-17 production by γδ T cells. Our findings indicate a role for IL-38 in the regulation of γδ T cell activation through IL1RAPL1, with consequences for auto-inflammatory disease.


Subject(s)
Interleukin-17/metabolism , Interleukin-1/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Skin/pathology , T-Lymphocytes/metabolism , Animals , Antibodies/immunology , Antibodies/therapeutic use , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Humans , Imiquimod/toxicity , Inflammation/prevention & control , Interleukin-1/genetics , Interleukin-1/pharmacology , Interleukin-1 Receptor Accessory Protein/deficiency , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Psoriasis/chemically induced , Psoriasis/pathology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Regeneration/drug effects , Skin/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology
7.
Basic Res Cardiol ; 114(3): 26, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31016449

ABSTRACT

Sepsis-induced myocardial depression (SIMD) is an early and frequent consequence of the infection-induced systemic inflammatory response syndrome. In homiotherms, variations in ambient temperature (Ta) outside the thermoneutral zone induce thermoregulatory responses mainly driven by a gradually increased sympathetic activity, which may affect disease severity. We hypothesized that thermoregulatory responses upon reduced Ta exposition aggravate SIMD in mice. Mice were kept at neutral Ta (30 ± 0.5 °C), moderately lowered Ta (26 ± 0.5 °C) or markedly lowered Ta (22 ± 0.5 °C), exposed to lipopolysaccharide- (LPS, 10 µg/g, from Escherichia coli serotype 055:B5, single intraperitoneal injection) evoked shock and monitored for survival, cardiac autonomic nervous system function and left ventricular performance. Primary adult cardiomyocytes and heart tissue derived from treated mice were analyzed for inflammatory responses and signaling pathways of myocardial contractility. We show that a moderate reduction of Ta to 26 °C led to a 40% increased mortality of LPS-treated mice when compared to control mice and that a marked reduction of Ta to 22 °C resulted in an early mortality of all mice. Mice kept at 26 °C exhibited increased heart rate and altered indices of heart rate variability (HRV), indicating sympathovagal imbalance along with aggravated LPS-induced SIMD. This SIMD was associated with reduced myocardial ß-adrenergic receptor expression and suppressed adrenergic signaling, as well as with increased myocardial iNOS expression, nitrotyrosine formation and leukocyte invasion as well as enhanced apoptosis and appearance of contraction band necrosis in heart tissue. While ineffective separately, combined treatment with the ß2-adrenergic receptor (AR) antagonist ICI 118551 (10 ng/gbw) and the inducible nitric oxide synthase (iNOS) inhibitor 1400 W (5 µg/gbw) reversed the increase in LPS-induced mortality and aggravation of SIMD at reduced Ta. Thus, consequences of thermoregulatory adaptation in response to ambient temperatures below the thermoneutral range increase the mortality from LPS-evoked shock and markedly prolong impaired myocardial function. These changes are mitigated by combined ß2-AR and iNOS inhibition.


Subject(s)
Autonomic Nervous System/physiopathology , Body Temperature Regulation , Heart Diseases/chemically induced , Heart/innervation , Housing, Animal , Myocardial Contraction , Systemic Inflammatory Response Syndrome/chemically induced , Temperature , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Heart Diseases/metabolism , Heart Diseases/physiopathology , Hemodynamics , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Systemic Inflammatory Response Syndrome/metabolism , Systemic Inflammatory Response Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...