Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Cell Rep ; 43(9): 114670, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39213156

ABSTRACT

Neutrophils from skull bone marrow (Nskull) are activated under some brain stresses, but their effects on traumatic brain injury (TBI) are lacking. Here, we find Nskull infiltrates brain tissue quickly and persistently after TBI, which is distinguished by highly and specifically expressed osteocalcin (OCN) from blood-derived neutrophils (Nblood). Reprogramming of glucose metabolism by reducing glycolysis-related enzyme glyceraldehyde 3-phosphate dehydrogenase expression is involved in the antiapoptotic and proliferative abilities of OCN-expressing Nskull. The transcription factor Fos-like 1 governs the specific gene profile of Nskull including C-C motif chemokine receptor-like 2 (CCRL2), arginase 1 (Arg1), and brain-derived neurotrophic factor (BDNF) in addition to OCN. Selective knockout of CCRL2 in Nskull demonstrates that CCRL2 mediates its recruitment, whereas high Arg1 expression is consistent with its immunosuppressive effects on Nblood, and the secretion of BDNF facilitating dendritic growth contributes to its neuroprotection. Thus, our findings provide insight into the roles of Nskull in TBI.

2.
Mil Med Res ; 11(1): 20, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38556884

ABSTRACT

BACKGROUND: Neutrophils are traditionally viewed as first responders but have a short onset of action in response to traumatic brain injury (TBI). However, the heterogeneity, multifunctionality, and time-dependent modulation of brain damage and outcome mediated by neutrophils after TBI remain poorly understood. METHODS: Using the combined single-cell transcriptomics, metabolomics, and proteomics analysis from TBI patients and the TBI mouse model, we investigate a novel neutrophil phenotype and its associated effects on TBI outcome by neurological deficit scoring and behavioral tests. We also characterized the underlying mechanisms both in vitro and in vivo through molecular simulations, signaling detections, gene expression regulation assessments [including dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays], primary cultures or co-cultures of neutrophils and oligodendrocytes, intracellular iron, and lipid hydroperoxide concentration measurements, as well as forkhead box protein O1 (FOXO1) conditional knockout mice. RESULTS: We identified that high expression of the FOXO1 protein was induced in neutrophils after TBI both in TBI patients and the TBI mouse model. Infiltration of these FOXO1high neutrophils in the brain was detected not only in the acute phase but also in the chronic phase post-TBI, aggravating acute brain inflammatory damage and promoting late TBI-induced depression. In the acute stage, FOXO1 upregulated cytoplasmic Versican (VCAN) to interact with the apoptosis regulator B-cell lymphoma-2 (BCL-2)-associated X protein (BAX), suppressing the mitochondrial translocation of BAX, which mediated the antiapoptotic effect companied with enhancing interleukin-6 (IL-6) production of FOXO1high neutrophils. In the chronic stage, the "FOXO1-transferrin receptor (TFRC)" mechanism contributes to FOXO1high neutrophil ferroptosis, disturbing the iron homeostasis of oligodendrocytes and inducing a reduction in myelin basic protein, which contributes to the progression of late depression after TBI. CONCLUSIONS: FOXO1high neutrophils represent a novel neutrophil phenotype that emerges in response to acute and chronic TBI, which provides insight into the heterogeneity, reprogramming activity, and versatility of neutrophils in TBI.


Subject(s)
Brain Injuries, Traumatic , Neutrophils , Animals , Humans , Mice , bcl-2-Associated X Protein/metabolism , Brain , Brain Injuries, Traumatic/complications , Depression , Forkhead Box Protein O1/metabolism , Iron
3.
Exp Neurol ; 374: 114691, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38224942

ABSTRACT

Blood-brain barrier (BBB) impairment and glutamate release are two pathophysiological features of traumatic brain injury (TBI), contributing to secondary brain damage and neuroinflammation. However, our knowledge of BBB integrity damage and dysfunction are still limited due to the diverse and fluctuating expression of glutamate receptors after trauma. Here, we confirmed the downregulation of metabotropic glutamate receptor 5 (mGluR5) on microvascular endothelial cell within the acute phase of TBI, and the recovered mGluR5 levels on BBB was positively associated with blood perfusion and neurological recovery. In whole body mGluR5-knockout mice, BBB dysfunction and neurological deficiency were exacerbated after TBI compared with wild type mice. In terms of mechanism, the amino acid sequence 201-259 of cytoskeletal protein Alpha-actinin-1 (ACTN1) interacted with mGluR5, facilitating mGluR5 translocation from cytoplasmic compartment to plasma membrane in endothelial cells. Activation of plasma membrane mGluR5 triggers the PLC/PKCµ/c-Jun signaling pathway, leading to increased expression of the tight junction-actin cytoskeleton connecting protein zonula occludens-1 (ZO-1). Our findings uncover a novel mechanism mediated by membrane and cytoplasmic mGluR5 in endothelial cell integrity maintenance and repair, providing the potential therapeutic target for TBI treatment targeting at mGluR5 and mGluR5/ACTN1 complex in BBB.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Animals , Mice , Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Brain Injuries, Traumatic/metabolism , Endothelial Cells/metabolism , Mice, Knockout , Receptor, Metabotropic Glutamate 5/metabolism
4.
Int J Mol Sci ; 24(21)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37958896

ABSTRACT

Aortic dissection (AD) is a cardiovascular disease that seriously endangers the lives of patients. The mortality rate of this disease is high, and the incidence is increasing annually, but the pathogenesis of AD is complicated. In recent years, an increasing number of studies have shown that immune cell infiltration in the media and adventitia of the aorta is a novel hallmark of AD. These cells contribute to changes in the immune microenvironment, which can affect their own metabolism and that of parenchymal cells in the aortic wall, which are essential factors that induce degeneration and remodeling of the vascular wall and play important roles in the formation and development of AD. Accordingly, this review focuses on the independent and interactive roles of immunity and metabolism in AD to provide further insights into the pathogenesis, novel ideas for diagnosis and new strategies for treatment or early prevention of AD.


Subject(s)
Aortic Dissection , Humans , Aorta
5.
Biochem Mol Biol Educ ; 51(5): 557-565, 2023.
Article in English | MEDLINE | ID: mdl-37409779

ABSTRACT

The development of information technology and portable devices has sparked a revolution in the field of education, facilitating access to diverse educational resources and lifelong learning. In particular, the COVID-19 pandemic has accelerated the transition from face-to-face to distance teaching, which requires online education to be provided worldwide. Biochemistry and Molecular Biology are key basic medical courses in laboratory-based science that cover complicated theories and applications. The balance between traditional and online courses, and the effectiveness of online courses, are fundamental to the teaching quality of Biochemistry and Molecular Biology. In this study, we explored the concepts, designs, and practices of a new blended online course and identified potential challenges. We hope that our experiences will provide new ideas for online teaching and promote teaching reform and the development of Medical Biochemistry and Molecular Biology education.

6.
Neurosci Bull ; 38(10): 1199-1214, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35819574

ABSTRACT

The brain pericyte is a unique and indispensable part of the blood-brain barrier (BBB), and contributes to several pathological processes in traumatic brain injury (TBI). However, the cellular and molecular mechanisms by which pericytes are regulated in the damaged brain are largely unknown. Here, we show that the formation of neutrophil extracellular traps (NETs) induces the appearance of CD11b+ pericytes after TBI. These CD11b+ pericyte subsets are characterized by increased permeability and pro-inflammatory profiles compared to CD11b- pericytes. Moreover, histones from NETs by Dectin-1 facilitate CD11b induction in brain pericytes in PKC-c-Jun dependent manner, resulting in neuroinflammation and BBB dysfunction after TBI. These data indicate that neutrophil-NET-pericyte and histone-Dectin-1-CD11b are possible mechanisms for the activation and dysfunction of pericytes. Targeting NETs formation and Dectin-1 are promising means of treating TBI.


Subject(s)
Brain Injuries, Traumatic , Extracellular Traps , Blood-Brain Barrier/metabolism , Brain/pathology , Brain Injuries, Traumatic/metabolism , Extracellular Traps/metabolism , Histones , Humans , Lectins, C-Type , Pericytes/pathology
7.
Cancer Sci ; 113(6): 1955-1967, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35363928

ABSTRACT

Neutrophils are the first defenders of the innate system for injury and infection. They have gradually been recognized as important participants in tumor initiation and development due to their heterogeneity and plasticity. In the tumor microenvironment (TME), neutrophils can exert antitumor and protumor functions, depending on the surroundings. Tumor cells systemically alter intracellular amino acid (AA) metabolism and extracellular AA distribution to meet their proliferation need, leading to metabolic reprogramming and TME reshaping. However, the underlying mechanisms that determine how altered AAs affect neutrophils in TME are less-explored. Here, we identified that abundant glutamate releasing from tumor cells blunted neutrophils' cell-killing effects toward tumor cells in vitro and in vivo. Mass spectrometric detection, flow cytometry, and western blot experiments proved that increased levels of pSTAT3/RAB10/ARF4, mediated by glutamate, were accompanied with immunosuppressive phenotypes of neutrophils in TME. We also discovered that riluzole, an FDA-approved glutamate release inhibitor, significantly inhibited tumor growth by restoring neutrophils' cell-killing effects and decreasing glutamate secretion from tumor cells. These findings highlight the importance of tumor-released glutamate on neutrophil transformation in TME, providing new possible cancer treatments targeting altered glutamate metabolism.


Subject(s)
Neoplasms , Tumor Microenvironment , Apoptosis , Glutamic Acid , Humans , Neoplasms/pathology , Neutrophils/metabolism
8.
MedComm (2020) ; 2(3): 402-413, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34766153

ABSTRACT

Neutrophils are essential components of the immune system and have vital roles in the pathogenesis of autoimmune disorders. As effector cells, neutrophils promote autoimmune disease by releasing cytokines and chemokines cascades that accompany inflammation, neutrophil extracellular traps (NETs) regulating immune responses through cell-cell interactions. More recent evidence has extended functions of neutrophils. Accumulating evidence implicated neutrophils contribute to tissue damage during a broad range of disorders, involving rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), primary sjögren's syndrome (pSS), multiple sclerosis (MS), crohn's disease (CD), and gout. A variety of studies have reported on the functional role of neutrophils as therapeutic targets in autoimmune diseases. However, challenges and controversies in the field remain. Enhancing our understanding of neutrophils' role in autoimmune disorders may further advance the development of new therapeutic approaches.

9.
Brain Behav Immun ; 79: 114-124, 2019 07.
Article in English | MEDLINE | ID: mdl-30682501

ABSTRACT

Antidepressant-like effects of metabotropic glutamate receptor 5 (mGluR5) have been verified by specific antagonists or whole body knock-out (KO) mice. Previous experiments indicate that blocking mGluR5 exerts antidepressant-like effects through neuronal mechanisms, like modulating NMDA receptor activity or 5-HT system. Here we found that transplanting bone marrow from mGluR5 KO mice to WT mice could also show antidepressant-like effects, which were confirmed by sucrose preference test and tail suspension test. Furthermore, mGluR5 deficiency dramatically inhibits cytokines release from bone marrow cells, such as IL-1ß, TNF-α and IL-6, alleviating proinflammatory responses in LPS-induced depression model. In addition, inhibited cytokines could decrease the activation of brain endothelial cells in ERK-dependent manner. These data provide the evidence that blocking mGluR5 could improve depression through inhibiting peripheral immune responses, confirming the causal relationship between peripheral immune phenotype and brain behavior.


Subject(s)
Antidepressive Agents/metabolism , Depression/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Bone Marrow/metabolism , Bone Marrow Transplantation/methods , Brain/metabolism , Cytokines/metabolism , Depression/drug therapy , Depression/etiology , Depressive Disorder/drug therapy , Depressive Disorder/etiology , Depressive Disorder/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Receptor, Metabotropic Glutamate 5/genetics , Receptor, Metabotropic Glutamate 5/physiology , Receptors, N-Methyl-D-Aspartate/metabolism
10.
J Neuroinflammation ; 15(1): 146, 2018 May 17.
Article in English | MEDLINE | ID: mdl-29776443

ABSTRACT

Our knowledge of the pathophysiology about traumatic brain injury (TBI) is still limited. Neutrophils, as the most abundant leukocytes in circulation and the first-line transmigrated immune cells at the sites of injury, are highly involved in the initiation, development, and recovery of TBI. Nonetheless, our understanding about neutrophils in TBI is obsolete, and mounting evidences from recent studies have challenged the conventional views. This review summarizes what is known about the relationships between neutrophils and pathophysiology of TBI. In addition, discussions are made on the complex roles as well as the controversial views of neutrophils in TBI.


Subject(s)
Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Neutrophils/physiology , Animals , Humans
11.
Sci Rep ; 7(1): 9998, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28855570

ABSTRACT

Both brain native inflammatory cells and infiltrated peripheral white blood cells (WBCs) are primary participants in the brain inflammatory damage post-TBI. Metabotropic glutamate receptor 5 (mGluR5) has been reported to regulate microglias and astrocytes to affect inflammation after TBI, but its effect on modulating infiltrated peripheral WBCs remains unclear. In a mouse moderate TBI model, we found that mGluR5 knockout (KO) significantly reduced neutrophil infiltration and inflammatory cytokine expression in the brain at 24 hours post TBI, which was accompanied by improved neurological dysfunction. Further investigation indicated that mGluR5 KO reduced the permeability of blood-brain barrier (BBB), the entrance for neutrophils to enter brain, and markedly decreased the mRNA levels of neutrophil-associated chemokines in brain tissue, including CXCL1, CXCL2, CCL2, CCL4 and CCL5. Using brain microvascular endothelial cells (BMECs), neutrophils and a BBB model in vitro, we confirmed the inhibitory effect of mGluR5 deficiency on neutrophil infiltration and demonstrated that blockade of protein kinase C (PKC) signaling was involved in it. These results provide insight into the role of mGluR5 in the regulation of inflammation in the acute phase of TBI, which may provide novel clues for TBI therapy.


Subject(s)
Blood-Brain Barrier/pathology , Brain Injuries, Traumatic/pathology , Neutrophil Infiltration , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Cells, Cultured , Cytokines/biosynthesis , Disease Models, Animal , Endothelial Cells/physiology , Mice , Mice, Knockout , Receptor, Metabotropic Glutamate 5/deficiency
12.
Sci Rep ; 6: 33614, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27647162

ABSTRACT

Systemic inflammatory response syndrome (SIRS) is an overwhelming whole body inflammation caused by infectious diseases or sterile insults. Neutrophils are the dominant participants during inflammation, and their survival and death determine the initiation as well as resolution of SIRS. Apoptosis and autophagy are two fundamental cellular processes that modulating cell fate, but their correlation and regulators in neutrophils under SIRS condition have not been elucidated. In this study, we demonstrated that high dose of LPS induced both apoptosis and autophagy of neutrophils in a mouse SIRS model and LPS-stimulated neutrophils in vitro. Moreover, we found that the adenosine 2A receptor (A2AR), a known anti-inflammatory G protein-coupled receptor (GPCR), could inhibit LPS-induced neutrophil apoptosis by suppressing the LPS-induced autophagy. Activation of A2AR suppressed LPS-induced autophagy by inhibiting the ROS-JNK pathway as well as promoting GPCR ßϒ subunit-AKT signaling. The A2AR-inhibited autophagy suppressed apoptosis of neutrophils by blocking caspase8, caspase3 and PARP signaling. These findings not only increase our understandings of neutrophils' fate and function in response to systemic inflammation, but also identify a novel anti-inflammatory role of A2AR in modulating neutrophils' survival during inflammation.


Subject(s)
Apoptosis , Autophagy , Neutrophils/metabolism , Receptor, Adenosine A2A/metabolism , Systemic Inflammatory Response Syndrome/etiology , Systemic Inflammatory Response Syndrome/metabolism , ADP Ribose Transferases , Animals , Apoptosis/immunology , Autophagy/immunology , Caspase 3/metabolism , Caspase 8/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/immunology , Mice , Neutrophils/immunology , Neutrophils/pathology , Neutrophils/ultrastructure , Phosphorylation , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
13.
Cell Signal ; 27(10): 2026-34, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26171727

ABSTRACT

Inflammation is a pathological course involved in several diseases. Both adenosine A2A receptor (A2AR) and miR-214 play important roles in regulation of inflammation. However, the internal link between them and their synergic modulation in inflammatory response has not been elucidated. In this study, we found that miR-214 and A2AR activation could downregulate the expressions of each other in murine macrophages. Comparing with the well known anti-inflammatory role of A2AR, miR-214 promoted the release of inflammatory cytokines TNF-α and IL-6. Further investigation demonstrated that miR-214 downregulated A2AR expression by directly targeting the 3'-untranslated region of A2AR mRNA. Instead of directly interacting with miR-214, A2AR activation repressed miR-214 expression by stimulating PKA signaling to suppress the nuclear translocation of NF-κB which could enhance the transcript activity of miR-214 gene promoter. Then using an LPS-induced ALI mouse model, in which inflammation is a hallmark, we confirmed their negative relationship and demonstrated that combination of miR-214 antagomir and A2AR agonist CGS21680 exerts more anti-inflammatory effect including alleviating the pathological changes, suppressing the neutrophil infiltration and the expression of inflammatory cytokines than using one of them alone. These findings for the first time uncovered a mutual suppression feedback loop between A2AR signaling and miR-214 in inflammation, which may provide new insight of inflammatory regulation and potential therapeutic significance for some inflammation-associated diseases.


Subject(s)
MicroRNAs/genetics , Receptor, Adenosine A2A/metabolism , Signal Transduction , 3' Untranslated Regions , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/genetics , Cytokines/metabolism , Feedback, Physiological , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA Interference , Transcription, Genetic
14.
Cell Signal ; 25(9): 1913-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712033

ABSTRACT

Although peroxisome proliferator-activated receptor-γ (PPARγ) and adenosine A2A receptor (A2AR) are reported to be anti-inflammatory factors in acute lung injury (ALI), their internal link and synergic or antagonistic effect after activation are poorly understood. Here, we found that PPARγ and A2AR could upregulate the mRNA and protein expressions of each other in lung tissues of LPS-induced mouse ALI model and murine macrophages. Further investigation demonstrated that PPARγ upregulated A2AR expression by directly binding to a DR10 response element (-218 to -197) within A2AR gene promoter region. Instead of directly interacting with PPARγ, A2AR stimulated PPARγ expression via protein kinase A (PKA)-cAMP response element binding protein (CREB) signaling by provoking the binding of CREB to a cAMP responsive element (CRE)-like site in PPARγ gene promoter region. In addition, combination of PPARγ and A2AR agonists was found to exert obviously better effect on suppressing neutrophil infiltration and inflammatory cytokine expressions, attenuating lung edema, pathological changes and improving lung function of blood gas exchange than their single application. These findings reveal a novel functional positive feedback loop between PPARγ and A2AR signaling to potentialize their effect on inhibiting inflammation and attenuating lung damages in ALI. It suggests that targeting this PPARγ-A2AR signaling rather than PPARγ or A2AR alone may be a more attractive and efficient potential therapeutic strategy for ALI.


Subject(s)
Acute Lung Injury/immunology , Acute Lung Injury/pathology , Lung/pathology , PPAR gamma/immunology , Receptor, Adenosine A2A/immunology , Acute Lung Injury/genetics , Acute Lung Injury/prevention & control , Adenosine A2 Receptor Agonists/therapeutic use , Animals , Cell Line , Cells, Cultured , Cytokines/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , PPAR gamma/agonists , PPAR gamma/genetics , RNA, Messenger/genetics , Receptor, Adenosine A2A/genetics , Signal Transduction , Transcriptional Activation , Up-Regulation
15.
PLoS One ; 8(3): e59257, 2013.
Article in English | MEDLINE | ID: mdl-23555007

ABSTRACT

Sinomenine (SIN) is a bioactive alkaloid extracted from the Chinese medicinal plant Sinomenium acutum, which is widely used in the clinical treatment of rheumatoid arthritis (RA). However, its role in acute lung injury (ALI) is unclear. In this study, we investigate the role of SIN in lipopolysaccharide (LPS)-induced ALI in mice. After ALI, lung water content and histological signs of pulmonary injury were attenuated, whereas the PaO2/FIO2 (P/F) ratios were elevated significantly in the mice pretreated with SIN. Additionally, SIN markedly inhibited inflammatory cytokine TNF-α and IL-1ß expression levels as well as neutrophil infiltration in the lung tissues of the mice. Microarray analysis and real-time PCR showed that SIN treatment upregulated adenosine A(2A) receptor (A(2A)R) expression, and the protective effect of SIN was abolished in A(2A)R knockout mice. Further investigation in isolated mouse neutrophils confirmed the upregulation of A(2A)R by SIN and showed that A(2A)R-cAMP-PKA signaling was involved in the anti-inflammatory effect of SIN. Taken together, these findings demonstrate an A(2A)R-associated anti-inflammatory effect and the protective role of SIN in ALI, which suggests a potential novel approach to treat ALI.


Subject(s)
Acute Lung Injury/prevention & control , Gene Expression Regulation/drug effects , Morphinans/pharmacology , Receptor, Adenosine A2A/genetics , Signal Transduction/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Profiling , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Lipopolysaccharides , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/metabolism , Receptor, Adenosine A2A/deficiency , Sinomenium/chemistry , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
16.
J Exp Med ; 210(4): 839-51, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23478188

ABSTRACT

The bone marrow-derived cell (BMDC)-associated inflammatory response plays a key role in the development of acute lung injury (ALI). Activation of adenosine A2A receptor (A2AR) is generally considered to be antiinflammatory, inhibiting BMDC activities to protect against ALI. However, in the present study, we found that in a mouse model of neurogenic ALI induced by severe traumatic brain injury (TBI), BMDC A2AR exerted a proinflammatory effect, aggravating lung damage. This is in contrast to the antiinflammatory effect observed in the mouse oleic acid-induced ALI model (a nonneurogenic ALI model.) Moreover, the A2AR agonist CGS21680 aggravated, whereas the antagonist ZM241385 attenuated, the severe TBI-induced lung inflammatory damage in mice. Further investigation of white blood cells isolated from patients or mouse TBI models and of cultured human or mouse neutrophils demonstrated that elevated plasma glutamate after severe TBI induced interaction between A2AR and the metabotropic glutamate receptor 5 (mGluR5) to increase phospholipase C-protein kinase C signaling, which mediated the proinflammatory effect of A2AR. These results are in striking contrast to the well-known antiinflammatory and protective role of A2AR in nonneurogenic ALI and indicate different therapeutic strategies should be used for nonneurogenic and neurogenic ALI treatment when targeting A2AR.


Subject(s)
Acute Lung Injury/blood , Bone Marrow Cells/metabolism , Brain Injuries/blood , Glutamic Acid/blood , Receptor, Adenosine A2A/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction , Acute Lung Injury/etiology , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adult , Animals , Bone Marrow Cells/pathology , Brain Injuries/complications , Brain Injuries/genetics , Brain Injuries/pathology , Disease Models, Animal , Female , Glutamic Acid/genetics , Humans , Male , Mice , Mice, Knockout , Middle Aged , Phenethylamines/pharmacology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics , Triazines/pharmacology , Triazoles/pharmacology , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
17.
Cell Signal ; 25(1): 159-67, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22986000

ABSTRACT

Proliferation of vascular smooth muscle cells (VSMCs) plays key roles in the progression of intimal hyperplasia, but the molecular mechanisms that trigger VSMC proliferation after vascular injury remain unclear. c-Ski, a co-repressor of transforming growth factor ß (TGF-ß)/Smad signaling, was detected to express in VSMC of rat artery. During the course of arterial VSMC proliferation induced by balloon injury in rat, the endogenous protein expressions of c-Ski decreased markedly in a time-dependent manner. In vivo c-Ski gene delivery was found to significantly suppress balloon injury-induced VSMC proliferation and neointima formation. Further investigation in A10 rat aortic smooth muscle cells demonstrated that overexpression of c-Ski gene inhibited TGF-ß1 (1 ng/ml)-induced A10 cell proliferation while knockdown of c-Ski by RNAi enhanced the stimulatory effect of TGF-ß1 on A10 cell growth. Western blot for signaling detection showed that suppression of Smad3 phosphorylation while stimulating p38 signaling associated with upregulation of cyclin-dependent kinase inhibitors p21 and p27 was responsible for the inhibitory effect of c-Ski on TGF-ß1-induced VSMC proliferation. These data suggest that the decrease of endogenous c-Ski expression is implicated in the progression of VSMC proliferation after arterial injury and c-Ski administration represents a promising role for treating intimal hyperplasia via inhibiting the proliferation of VSMC.


Subject(s)
Proto-Oncogene Proteins/metabolism , Smad3 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Neointima , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
18.
Cell Physiol Biochem ; 30(4): 1059-71, 2012.
Article in English | MEDLINE | ID: mdl-23052247

ABSTRACT

BACKGROUND/AIM: Both peroxisome proliferator-activated receptor (PPAR) δ and Ski are investigate the interaction of PPARδ and Ski and this interaction-associated effect in wound healing. METHODS: Effect of PPARδ activation on Ski expression was detected in rat skin fibroblasts by real-time PCR and western blot. Luciferase assay, electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assay were performed to identify the binding site of PPARδ in the promoter region of rat Ski gene. And the functional activity of PPARδ regulation to Ski was detected in fibroblast proliferation and rat skin wound healing model. RESULTS: PPARδ agonist GW501516 upregulated Ski expression in a dose-dependent manner. Direct repeat-1 (DR1) response element locating at -865∼-853 in Ski promoter region was identified to mediate PPARδ binding to Ski and associated induction of Ski. Furthermore, PPARδ upregulated Ski to promote fibroblasts proliferation and rat skin wound repair, which could be largely blocked by pre-treated with Ski RNA interference. CONCLUSION: This study demonstrates that Ski is a novel target gene for PPARδ and upregulation of Ski to promote fibroblast proliferation is implicated in the PPARδ-mediated wound healing.


Subject(s)
Fibroblasts/metabolism , PPAR delta/metabolism , Proto-Oncogene Proteins/genetics , Skin/cytology , Transcriptional Activation , Wound Healing , Animals , Cell Proliferation , Cells, Cultured , Female , Fibroblasts/cytology , PPAR delta/agonists , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , Rats , Rats, Wistar , Thiazoles/pharmacology , Up-Regulation/drug effects
19.
Rev Neurosci ; 22(2): 231-9, 2011.
Article in English | MEDLINE | ID: mdl-21476942

ABSTRACT

This review summarizes recent developments that have contributed to our understanding of how adenosine 2A receptors (A2ARs) modulate brain damage in various animal models of acute neurological injuries, including brain ischemia, traumatic brain injury, spinal cord injury and hemorrhage stroke. The main conclusions are: (1) pharmacological, neurochemical and molecular/genetic approaches to the complex actions of A2AR in different cellular elements suggest that A2AR activation exerts bidirectional effect (detrimental or protective) after brain insults; (2) modulation of glutamate excitotoxicity and neuroinflammation are involved in the protection of A2AR agonists or antagonists, but the bidirectional effect of A2AR is largely due to the bidirectional regulation of neuroinflammation (anti-inflammation or proinflammation) by A2AR on immune cells such as microglia cells and peripheral bone marrow cells; and (3) the bidirectional effect of A2AR on neuroinflammation and brain injury depends on the distinct and sometimes opposite actions of A2AR in various cellular elements and on different injury models and associated pathological conditions. The local glutamate level in the brain injury is one of the crucial factors that contribute to the direction of A2AR effect on neuroinflammation and brain injury outcome. These developments presented here clearly highlight the complexity of using A2AR agents therapeutically in acute neuronal injuries and confirm that A2AR ligands have many promising characteristics that encourage the pursuit of their full therapeutic potential.


Subject(s)
Brain Injuries/metabolism , Encephalitis/metabolism , Receptors, Adenosine A2/metabolism , Adenosine A2 Receptor Agonists/therapeutic use , Adenosine A2 Receptor Antagonists/therapeutic use , Animals , Brain Injuries/therapy , Encephalitis/therapy , Humans , Models, Biological , Neuroprotective Agents/therapeutic use , Receptors, Adenosine A2/genetics
20.
Sheng Li Xue Bao ; 63(1): 62-8, 2011 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-21340436

ABSTRACT

TGFß/smad pathway is recognized as an important signal pathway to promote the pathogenesis of atherosclerosis (AS). Peroxisome proliferator-activated receptor γ (PPARγ) activation is considered to be important in modulating AS. Herein, we investigated the regulation of PPARγ on c-Ski, the repressor of TGFß/smad pathway, in rat AS model and cultured vascular smooth muscle cells (VSMCs). c-Ski mRNA and protein expression were detected by real-time PCR and Western blot, respectively, in vivo and in vitro with treatment of PPARγ agonist rosiglitazone and antagonist GW9662. The proliferation and collagen secretion of VSMCs after c-Ski transfection were investigated. The underlying mechanism was further investigated by online program NUBIScan and luciferase reporter gene analysis. Results showed that both mRNA and protein expressions of c-Ski in the AS lesions was down-regulated in vivo, while in cultured VSMCs, c-Ski transfection significantly suppressed the proliferation and collagen secretion of rat VSMCs. Rosiglitazone significantly up-regulated mRNA and protein levels of c-Ski in VSMCs, which could be blocked by GW9662. Online NUBIScan analysis suggested possible PPARγ binding sites in the promoter region of c-Ski. In addition, luciferase activity of c-Ski reporter gene was also increased obviously in the presence of rosiglitazone. These results indicate that c-Ski is one of the newly found target genes of PPARγ and thus involved in the anti-AS effect of PPARγ.


Subject(s)
Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , PPAR gamma/physiology , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Anilides/pharmacology , Animals , Atherosclerosis/physiopathology , Cells, Cultured , Male , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rosiglitazone , Smad Proteins/metabolism , Thiazolidinediones/pharmacology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL