Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Biochem Mol Toxicol ; 34(11): e22573, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32659049

ABSTRACT

Docosahexaenoic acid (DHA) is reported to have the potential to ameliorate pulmonary arterial hypertension (PAH), while the specific mechanism is still obscure. This study aims to investigate the function of DHA in pulmonary artery smooth muscle cells (PASMCs) and explore the underlying mechanism. In our study, DHA was used to incubate PASMCs. Cytosolic-free Ca2+ concentration ([Ca2+ ]cyt) was measured using Fluo-3 AM method. Real-time polymerase chain reaction was used to detect microRNA-16 (miR-16) and calcium-sensing receptor (CaSR) messenger RNA expression levels. CCK-8 assay, BrdU assay, and Transwell assay were employed to detect the effects of DHA on proliferation and migration of PASMCs. CaSR was confirmed as a direct target of miR-16 using dual-luciferase assay, polymerase chain reaction, and Western blot analysis. It was found that DHA significantly inhibited PASMC proliferation and migration and decreased [Ca2+ ]cyt. After transfection of miR-16 mimics, proliferation and migration ability of PASMCs were significantly inhibited, whereas opposite effects were observed after miR-16 inhibition. [Ca2+ ]cyt was also inhibited by miR-16 transfection. DHA then promoted the expression of miR-16, and the effects of DHA on PASMCs were annulled when miR-16 was inhibited. CaSR was identified as a direct target of miR-16. CaSR was inhibited directly by miR-16 and indirectly by DHA. In conclusion, DHA inhibits the proliferation and migration of PASMCs, and probably ameliorates PAH via regulating miR-16/CaSR axis.


Subject(s)
Calcium/metabolism , Down-Regulation/drug effects , MicroRNAs/metabolism , Muscle, Smooth/drug effects , Pulmonary Artery/drug effects , Receptors, Calcium-Sensing/metabolism , Binding Sites , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Humans , Ion Transport , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism
2.
J Biosci ; 452020.
Article in English | MEDLINE | ID: mdl-32098916

ABSTRACT

This paper explores the potential mechanism of microRNA-143-5p regulation effects on pulmonary artery smooth muscle cells (PASMCs) functions in hypoxic pulmonary hypertension (HPH) via targeting HIF-1a, which may offer a new idea for HPH therapy. PASMCs were transfected with mimics control/miR-143-5p mimics or inhibitor control/miR-143-5p inhibitor. We used Western blotting and RT-qPCR to detect the protein and mRNA expressions, CCK-8 assay to detect cellular viability, Annexin V-FITC/PI staining and caspase- 3/cleaved caspase-3 protein to evaluate cellular apoptosis, transwell migration experiment for cellular migration measurement and Dual luciferase reporter gene assay to prove the target of miR-143-5p. Cells under hypoxic condition presented the decreased protein and mRNA expressions of α-smooth muscle actin (SM-α-actin), Myocardin, smooth muscle myosin heavy chain (SMMHC), and smooth muscle-22α (SM22α), Calponin1 and Hypoxia-inducible factor-1α(HIF-1α), the increased cell viability and miR-143-5p level; Overexpression of miR-143-5p obviously reduced vascular smooth muscle-specific contraction marker protein levels and cellular apoptosis, increased cellular migration of PASMCs with hypoxia stimulation; Low-expression of miR-143-5p caused the opposite changes, while co-transfected with Si HIF-1 α blocked the beneficial effects of miR-143-5p inhibition on PASMCs under hypoxia. MicroRNA-143-5p can promote the phenotype conversion, proliferation and migration of pulmonary artery smooth muscle cells under hypoxic condition through direct targeting of HIF-1α.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/physiology , Oxygen/pharmacology , Pulmonary Artery , Cell Migration Assays , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MicroRNAs/genetics
3.
Front Cell Neurosci ; 12: 255, 2018.
Article in English | MEDLINE | ID: mdl-30135647

ABSTRACT

Recent studies indicate that disturbed structure and function of microglia can cause depression and associated neurogenesis impairments. Our previous work has demonstrated that exogenous fibroblast growth factor 2 (FGF2) reverses the depressive-like behaviors and the impaired hippocampal neurogenesis in a neuroinflammatory model of depression. However, whether and how the antidepressant effects of FGF2 involve the modulation of microglia activation has not been elucidated. In this study, to examine the effects of FGF2 on microglia activation, exogenous FGF2 was supplemented to the lateral ventricle of rats during the neuroinflammatory state induced by central lipopolysaccharides (LPS) administrations. It was found that FGF2 infusions reversed the LPS-induced depressive-like behaviors and inhibited the hippocampal microglia activation. In LPS-treated rats, FGF2 decreased the level of pro-inflammatory cytokines including interlukin-1ß (IL-1ß), IL-6 and tumor necrosis factor (TNF)-α, increased the level of IL-10, the anti-inflammatory cytokine and reversed the decreased expression of CX3CL1, a chemokine mainly expressed by neurons and keeping microglia in surveillance. Further, we examined the effects of inhibited FGF2 signaling by administration of SU5402, an FGFR inhibitor. It was found that SU5402 itself evoked depressive-like behaviors, induced microglia activation, increased production of pro-inflammatory cytokines including IL-1ß, IL-6 and TNF-α, and decreased the expression of CX3CL1. Two lines of results that FGF2 signaling and FGFR inhibitor can effectively but oppositely modulate the regulation of microglia and the generation of depressive-like behavior, suggesting that microglia-regulated mechanisms may underlie the antidepressant role of FGF2. The present data provide novel insights into the understanding of mechanism of neuroinflammation-associated depression and may serve as a novel mechanism-based target for the treatment of inflammation-related depression.

4.
Front Pharmacol ; 9: 511, 2018.
Article in English | MEDLINE | ID: mdl-29867510

ABSTRACT

Increasing evidence has demonstrated that neuroinflammation contributes to the development of depressive-like behaviors, in both animal models and human patients; however, the brain areas and signaling pathways involved are still elusive. Recent studies have suggested novel roles of the habenula in the onset of depression and other psychiatric disorders; however, there is no evidence for whether the habenula has a function in neuroinflammation-induced depression. Using an animal model of depression, which is induced by the repeated central administration of lipopolysaccharide (LPS), we examined whether cytokine expression and p38 signal activation in the habenula were involved in the depressive-like behaviors. Body weight, saccharin preference test, and tail suspension test were used to measure depressive-like behaviors. Immunohistochemistry, quantitative-polymerase chain reaction (q-PCR), and western blot were used to measure the expression of tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10), and the phosphorylation of p38 in the habenula. The results showed that central LPS administration induced depressive-like behaviors, characterized by anhedonia in the saccharin preference test and increased immobility in the tail suspension test. Central LPS administration also significantly increased the p-p38 level in microglial cells and increased TNF-α expression in the habenula. Treatment with fluoxetine, a widely prescribed antidepressant, or SB203580, a p38-specific inhibitor, reversed the depressive-like behaviors, normalized the alterations in p-p38 and TNF-α levels and increased the levels of the anti-inflammatory cytokine IL-10 in the habenula. The present findings suggest that the habenula is involved in the pathophysiology of behavioral depression induced by neuroinflammation, and the p38 pathway may serve as a novel mechanism-based target for the treatment of inflammation-related depression.

5.
Sci Rep ; 8(1): 3110, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449595

ABSTRACT

Intercropping studies usually focus on yield advantage and interspecific interactions but few quantify temporal niche differentiation and its relationship with intercropping yield advantage. A field experiment conducted in northwest China in 2013 and 2014 examined four intercropping systems (oilseed rape/maize, oilseed rape/soybean, potato/maize, and soybean/potato) and the corresponding monocultures. Total dry matter data collected every 20 d after maize emergence were fitted to logistic models to investigate the temporal dynamics of crop growth and interspecific interactions. All four intercropping systems showed significant yield advantages. Temporal niche complementarity between intercropped species was due to differences in sowing and harvesting dates or the time taken to reach maximum daily growth rate or both. Interspecific interactions between intercropped species amplified temporal niche differentiation as indicated by postponement of the time taken to reach maximum daily growth rate of late-maturing crops (i.e. 21 to 41 days in maize associated with oilseed rape or potato). Growth trajectories of intercropped maize or soybean recovered after the oilseed rape harvest to the same values as in their monoculture on a per plant basis. Amplified niche differentiation between crop species depends on the identity of neighboring species whose relative growth rate is crucial in determining the differentiation.


Subject(s)
Agriculture/methods , Crop Production/methods , Brassica rapa/growth & development , China , Crops, Agricultural/growth & development , Fertilizers , Nitrogen/analysis , Soil/chemistry , Solanum tuberosum/growth & development , Glycine max/growth & development , Time Factors , Zea mays/growth & development
6.
Brain Behav Immun ; 66: 322-331, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28529071

ABSTRACT

Our previous work demonstrated that neuroinflammation evoked by triple repeated central LPS challenges inhibited adult hippocampal neurogenesis that were correlated with the depressive-like behavioral symptoms induced by neuroinflammation. These findings suggest that hippocampal neurogenesis might be one of biological mechanisms underlying depression induced by neuroinflammation and targeting neurogenesis might lead to new therapeutic strategies for the treatment of depression. In this study, we manipulated adult hippocampal neurogenesis using fibroblast growth factor 2 (FGF2), one crucial molecule modulating cell proliferation and survival in central nervous system, and investigate the involvement and the potential therapeutic effects of FGF2 on neuroinflammation-induced depression. Central lipopolysaccharides (LPS) challenges were used as previously to evoke the neuroinflammatory state in the brain of rat. Exogenous FGF2 was infused into lateral ventricle during the neuroinflammatory state. It was found that the protein expression of FGF2 in hippocampus was inhibited by neuroinflammation. The activation of extracellular signal-regulated kinase (ERK), the downstream molecule of FGF2, was also inhibited by neuroinflammation. Exogenous FGF2 infusions prevented the decrease in phosphorylation of ERK1/2 under neuroinflammation state. Exogenous FGF2 reversed depressive-like behaviors and the impaired hippocampal neurogenesis induced by neuroinflammation. These findings provide evidence that the FGF2-ERK1/2 pathway is involved in the pathophysiology of depressive-like behaviors, and manipulating the neurogenesis pathway is a viable therapeutic approach to inflammation-associated depression.


Subject(s)
Depression/metabolism , Encephalitis/metabolism , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/metabolism , Hippocampus/metabolism , MAP Kinase Signaling System , Neurogenesis , Animals , Depression/prevention & control , Encephalitis/chemically induced , Lipopolysaccharides/administration & dosage , Male , Phosphorylation , Rats, Sprague-Dawley
7.
Oncotarget ; 7(29): 45052-45059, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27281608

ABSTRACT

Latent transforming growth factor (TGF)-beta binding protein 2 (LTBP2) belongs to the fibrillin/LTBP extracellular matrix glycoprotein superfamily. It plays vital roles in tumorigenesis through regulating TGFß activity, elastogenesis and maintenance of the extracellular matrix (ECM) structure. In this study, we determined the expression levels of LTBP2 mRNA and protein in head and neck squamous cell carcinoma (HNSCC) tissues and adjacent normal tissues by quantitative reverse transcription PCR (qRT-PCR) and tissue microarray immunohistochemistry analysis (TMA-IHC) respectively. LTBP2 protein levels in cancer tissues were correlated with HNSCC patients' clinical characteristics and overall survival. Both LTBP2 mRNA and protein levels were significantly higher in HNSCC tissues than in adjacent normal tissues. High LTBP2 protein level was associated with lymph node metastasis and higher pTNM stages. High LTBP2 protein level is an independent prognostic marker in HNSCC. Our data suggest that LTBP2 acts as an oncogene in HNSCC development and progression. Detection of LTBP2 expression could be a useful prognosis marker and targeting LTBP2 may represent a novel strategy for cancer treatment through regulating activities of TGFß.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Latent TGF-beta Binding Proteins/biosynthesis , Adult , Aged , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Humans , Infant , Kaplan-Meier Estimate , Latent TGF-beta Binding Proteins/analysis , Male , Middle Aged , Prognosis , Squamous Cell Carcinoma of Head and Neck
8.
Physiol Behav ; 161: 166-173, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27106565

ABSTRACT

Our previous work found that triple central lipopolysaccharide (LPS) administration could induce depressive-like behaviors and increased central pro-inflammatory cytokines mRNA, hippocampal cytokine mRNA in particular. Since several neuroinflammation-associated conditions have been reported to impair neurogenesis, in this study, we further investigated whether the neuroinflammation induced depression would be associated with hippocampal neurogenesis dysfunction. An animal model of depression induced by triple central lipopolysaccharide (LPS) administration was used. In the hippocampus, the neuroinflammatory state evoked by LPS was marked by an increased production of pro-inflammatory cytokines, including interleukin-1ß, interleukin-6, and tumor necrosis factor-α. It was found that rats in the neuroinflammatory state exhibited depressive-like behaviors, including reduced saccharin preference and locomotor activity as well as increased immobility time in the tail suspension test and latency to feed in the novelty suppressed feeding test. Adult hippocampal neurogenesis was concomitantly inhibited, including decreased cell proliferation and newborn cell survival. We also demonstrated that the decreased hippocampal neurogenesis in cell proliferation was significantly correlated with the depressive-like phenotypes of decreased saccharine preference and distance travelled, the core and characteristic symptoms of depression, under neuro inflammation state. These findings provide the first evidence that hippocampal neurogenesis dysfunction is correlated with neuroinflammation-induced depression, which suggests that hippocampal neurogenesis might be one of biological mechanisms underlying depression induced by neruoinflammation.


Subject(s)
Cytokines/metabolism , Depression/complications , Encephalitis/etiology , Encephalitis/pathology , Hippocampus/pathology , Neurogenesis/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Exploratory Behavior/drug effects , Food Preferences/drug effects , Hindlimb Suspension , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Male , Rats , Rats, Sprague-Dawley , Saccharin/administration & dosage , Time Factors
9.
Psych J ; 4(3): 113-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26354152

ABSTRACT

Although proinflammatory cytokine changes in depression have been studied widely, few investigations have searched for specific and common changes in cytokines. In the present study, two animal models of depression were compared: a chronic stress model using forced swim stress and an immune activation model using repeated central lipopolysaccharide (LPS) infusion. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 mRNA were examined in the brain regions of the prefrontal cortex, amygdala, and hippocampus using real-time polymerase chain reaction (RT-PCR). It was found that both chronic swim stress and repeated central LPS infusion induced depressive-like behaviors, including decreased body weight, reduced saccharin preference, and increased immobility time or shortened latency of immobility in the tail suspension test. Central TNF-α mRNA expression was elevated in both models and central IL-6 mRNA expression was unchanged in both models. Central IL-1ß mRNA expression was increased only in the chronic immune activation model. The findings from this study suggest that TNF-α may be a common risk factor for inflammation in depressive disorders.


Subject(s)
Cytokines/metabolism , Depression/immunology , Inflammation/immunology , Lipopolysaccharides/immunology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/immunology , Depression/chemically induced , Inflammation/chemically induced , Male , RNA, Messenger/metabolism , Random Allocation , Rats , Real-Time Polymerase Chain Reaction , Stress, Physiological/immunology , Stress, Psychological/immunology , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL
...