Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Rev Cardiovasc Med ; 25(4): 121, 2024 Apr.
Article in English | MEDLINE | ID: mdl-39076555

ABSTRACT

Following long-term hypertension, mechanical stretching and neuroendocrine stimulation, cause multiple heterogeneous cells of the heart to interact, and result in myocardial remodeling with myocardial hypertrophy and fibrosis. The immune system, specifically macrophages, plays a vital role in this process. Macrophages are heterogeneous and plastic. Regulated by factors such as microenvironment and cytokines, polarization can be divided into two main forms: M1/M2, with different polarizations playing different roles in left ventricular structural remodeling associated with hypertension. However, descriptions of macrophage phenotypes in hypertension-induced myocardial hypertrophy models are not completely consistent. This article summarizes the phenotypes of macrophages in several models, aiming to assist researchers in studying macrophage phenotypes in hypertension-induced left ventricular structural remodeling models.

2.
Ageing Res Rev ; 93: 102160, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38065225

ABSTRACT

Central nervous system (CNS) diseases have become one of the leading causes of death in the global population. The pathogenesis of CNS diseases is complicated, so it is important to find the patterns of the disease to improve the treatment strategy. Microglia are considered to be a double-edged sword, playing both harmful and beneficial roles in CNS diseases. Therefore, it is crucial to understand the progression of the disease and the changes in the polar phenotype of microglia to provide guidance in the treatment of CNS diseases. Microglia activation may evolve into different phenotypes: M1 and M2 types. We focused on the roles that M1 and M2 microglia play in regulating intercellular dialogues, pathological reactions and specific diseases in CNS diseases. Importantly, we summarized the strategies used to modulate the polarization phenotype of microglia, including traditional pharmacological modulation, biological therapies, and physical strategies. This review will contribute to the development of potential strategies to modulate microglia polarization phenotypes and provide new alternative therapies for CNS diseases.


Subject(s)
Central Nervous System Diseases , Microglia , Humans , Microglia/pathology , Central Nervous System Diseases/therapy , Central Nervous System Diseases/pathology , Phenotype
3.
J Neuroimmune Pharmacol ; 18(4): 657-673, 2023 12.
Article in English | MEDLINE | ID: mdl-37955765

ABSTRACT

Cerebral ischemia reperfusion (I/R) is one of the neurovascular diseases which leads to severe brain deterioration. Haemorrhagic transformation (HT) is the main complication of ischemic stroke. It exacerbates by reperfusion, causing a more deleterious effect on the brain and death. The current study explored the protective effect of sertraline (Sert) against cerebral I/R in rats by inhibiting HT, together with the molecular pathways involved in this effect. Forty-eight wister male rats were divided into 4 groups: Sham, Sert + Sham, I/R, and Sert + I/R. The ischemic model was induced by bilateral occlusion of the common carotid artery for 20 min, then reperfusion for 24 h. Sertraline (20 mg/kg, p.o.) was administrated for 14 days before exposure to ischemia. Pre-treatment with Sert led to a significant attenuation of oxidative stress and inflammation. In addition, Sert attenuated phosphorylation of extracellular regulated kinases and nuclear factor kappa-p65 expression, consequently modulating microglial polarisation to M2 phenotype. Moreover, Sert prevented the hemorrhagic transformation of ischemic stroke as indicated by the notable decrease in neuronal expression of CD163, activity of Heme oxygenase-2 and matrix metalloproteinase-2 and 9 levels. In the same context, Sert decreased levels of autophagy and apoptotic markers. Furthermore, histological examination, Toluidine blue, and Prussian blue stain aligned with the results. In conclusion, Sert protected against cerebral I/R damage by attenuating oxidative stress, inflammation, autophagy, and apoptotic process. It is worth mentioning that our study was the first to show that Sert inhibited hemorrhagic transformation. The protective effect of sertraline against injury induced by cerebral ischemia reperfusion via inhibiting Hemorrhagic transformation.


Subject(s)
Brain Ischemia , Ischemic Stroke , Reperfusion Injury , Stroke , Rats , Male , Animals , Matrix Metalloproteinase 2/metabolism , Sertraline/pharmacology , Sertraline/therapeutic use , Down-Regulation , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Stroke/complications , Reperfusion , Ischemic Stroke/complications , Autophagy , Inflammation , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy
4.
Bull Exp Biol Med ; 175(5): 625-628, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37874494

ABSTRACT

Phenotypic characteristics of alveolar macrophages in the bronchoalveolar lavage fluid as well as their ability to acquire the M1 and M2 phenotypes during in vitro culturing with reprogramming factors were studied in rats with modeled diet-induced metabolic syndrome. A decrease in the number of alveolar macrophages with the M1 phenotype was found in animals with metabolic syndrome. The factors of metabolic syndrome do not affect phenotypic plasticity of cells in culture, but under the action of M2 reprogramming factors, the cells demonstrate a wide range of phenotypic plasticity by the CD80 and CD206 markers. The consistently high level of production of IL-6 and IL-10 by macrophages during culturing under different conditions indicates functional rigidity of the cells, which is probably a consequence of in vivo predetermined functional phenotype of these cells against the background of metabolic disorders.


Subject(s)
Macrophages, Alveolar , Metabolic Syndrome , Rats , Animals , Metabolic Syndrome/metabolism , Macrophages/metabolism , Phenotype , Bronchoalveolar Lavage Fluid
5.
Int J Mol Sci ; 24(19)2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37834030

ABSTRACT

Parkinson's disease is a neurodegenerative disease exhibiting the fastest growth in incidence in recent years. As with most neurodegenerative diseases, the pathophysiology is incompletely elucidated, but compelling evidence implicates inflammation, both in the central nervous system and in the periphery, in the initiation and progression of the disease, although it is not yet clear what triggers this inflammatory response and where it begins. Gut dysbiosis seems to be a likely candidate for the initiation of the systemic inflammation. The therapies in current use provide only symptomatic relief, but do not interfere with the disease progression. Nonetheless, animal models have shown promising results with therapies that target various vicious neuroinflammatory cascades. Translating these therapeutic strategies into clinical trials is still in its infancy, and a series of issues, such as the exact timing, identifying biomarkers able to identify Parkinson's disease in early and pre-symptomatic stages, or the proper indications of genetic testing in the population at large, will need to be settled in future guidelines.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Animals , Parkinson Disease/genetics , Neurodegenerative Diseases/therapy , Neuroinflammatory Diseases , Inflammation , Biomarkers , Microglia
6.
J Chem Neuroanat ; 133: 102312, 2023 11.
Article in English | MEDLINE | ID: mdl-37459999

ABSTRACT

Postoperative ileus (POI) is the cessation or reduction of gastrointestinal (GI) motility after surgery. Reactive enteric glial cells (EGCs) are critical for maintaining bowel function. However, the triggering mechanisms and downstream effects of reactive EGCs in POI were poorly understood. The goal of this current study was to investigate whether the inducible nitric oxide synthase (iNOS)-driven reactive EGCs participated in GI motility disorders and mechanisms underlying altered GI motility in POI. Intestinal manipulation (IM)-induced POI mice and iNOS-/- mice were used in the study. Longitudinal muscle and myenteric plexuses (LMMPs) from the distal small intestine were stained by immunofluorescence. Our results found that the GI motility disorders occurred in the IM-induced POI mice, and reactive EGCs were observed in LMMPs. Glial metabolic inhibitor gliotoxin fluorocitrate (FC) treatment or iNOS gene knockout attenuated GI motility dysfunction. In addition, we also found that FC treatment or iNOS gene knockout significantly inhibited the fluorescence intensity macrophage colony-stimulating factor (M-CSF), which reduced M2 phenotype macrophages activation in LMMPs of IM-induced POI mice. Our findings demonstrated that iNOS-driven reactive EGCs played a key role and were tightly linked to the MMs homeostasis in the POI mice. EGCs are emerging as a new frontier in neurogastroenterology and a potential therapeutic target.


Subject(s)
Ileus , Mice , Animals , Nitric Oxide Synthase Type II/metabolism , Ileus/metabolism , Gastrointestinal Motility/physiology , Neuroglia/metabolism , Intestine, Small/metabolism
7.
Biomed Pharmacother ; 165: 115042, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37379639

ABSTRACT

Mesenchymal stem cells (MSCs) are pluripotent stem cells derived from a variety of tissues, such as umbilical cord, fat, and bone marrow. Today, MSCs are widely recognized for their prominent anti-inflammatory properties in a variety of acute and chronic inflammatory diseases. In inflammatory diseases, monocytes/macrophages are an important part of the innate immune response in the body, and the alteration of the inflammatory phenotype plays a crucial role in the secretion of pro-inflammatory/anti-inflammatory factors, the repair of injured sites, and the infiltration of inflammatory cells. In this review, starting from the effect of MSCs on the monocyte/macrophage phenotype, we have outlined in detail the process by which MSCs influence the transformation of the monocyte/macrophage inflammatory phenotype, emphasizing the central role of monocytes/macrophages in MSC-mediated anti-inflammatory and damage site repair. MSCs are phagocytosed by monocytes/macrophages in various physiological states, the paracrine effect of MSCs and mitochondrial transfer of MSCs to macrophages to promote the transformation of monocytes/macrophages into anti-inflammatory phenotypes. We also review the clinical applications of the MSCs-monocytes/macrophages system and describe novel pathways between MSCs and tissue repair, the effects of MSCs on the adaptive immune system, and the effects of energy metabolism levels on monocyte/macrophage phenotypic changes.


Subject(s)
Mesenchymal Stem Cells , Monocytes , Monocytes/metabolism , Macrophages/metabolism , Phenotype , Anti-Inflammatory Agents/metabolism , Mesenchymal Stem Cells/metabolism
8.
Int J Mol Sci ; 24(12)2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37373050

ABSTRACT

Neuroinflammation is one of the postulated mechanisms for Pb neurotoxicity. However, the exact molecular mechanisms responsible for its pro-inflammatory effect are not fully elucidated. In this study, we examined the role of glial cells in neuroinflammation induced by Pb exposure. We investigated how microglia, a type of glial cell, responded to the changes caused by perinatal exposure to Pb by measuring the expression of Iba1 at the mRNA and protein levels. To assess the state of microglia, we analyzed the mRNA levels of specific markers associated with the cytotoxic M1 phenotype (Il1b, Il6, and Tnfa) and the cytoprotective M2 phenotype (Arg1, Chi3l1, Mrc1, Fcgr1a, Sphk1, and Tgfb1). Additionally, we measured the concentration of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α). To assess the reactivity and functionality status of astrocytes, we analyzed the GFAP (mRNA expression and protein concentration) as well as glutamine synthase (GS) protein level and activity. Using an electron microscope, we assessed ultrastructural abnormalities in the examined brain structures (forebrain cortex, cerebellum, and hippocampus). In addition, we measured the mRNA levels of Cxcl1 and Cxcl2, and their receptor, Cxcr2. Our data showed that perinatal exposure to Pb at low doses affected both microglia and astrocyte cells' status (their mobilization, activation, function, and changes in gene expression profile) in a brain-structure-specific manner. The results suggest that both microglia and astrocytes represent a potential target for Pb neurotoxicity, thus being key mediators of neuroinflammation and further neuropathology evoked by Pb poisoning during perinatal brain development.


Subject(s)
Astrocytes , Microglia , Pregnancy , Female , Humans , Astrocytes/metabolism , Microglia/metabolism , Lead/metabolism , Neuroinflammatory Diseases , Cytokines/metabolism , Prosencephalon/metabolism , RNA, Messenger/metabolism
9.
Toxicology ; 493: 153554, 2023 07.
Article in English | MEDLINE | ID: mdl-37236336

ABSTRACT

DDT, a persistent organic pollutant, remains affecting human health worldwide. DDT and its most persistent metabolite (p,p'-DDE) negatively affect the immune response regulation and mechanisms involved in protecting against pathogens Such metabolite decreases the capability to limit intracellular growth of Mycobacterium microti and yeast. However, the effect on unstimulated (M0) and anti-inflammatory macrophages (M2) has been evaluated scanty. Herein, we evaluated the impact of p,p'-DDE at environmentally relevant concentrations (0.125, 1.25, 2.5, and 5 µg/mL) on bone marrow-derived macrophages stimulated with IFNγ+LPS to M1 or with IL-4 +IL-13 to M2. Thus we study whether the p,p'-DDE induces M0 to a specific phenotype or modulates activation of the macrophage phenotypes and explains, at least partly, the reported effects of p,p'-DDE on the M1 function. The p,p'-DDE did not affect the cell viability of M0 or the macrophage phenotypes. In M1, the p,p'-DDE decreased NO•- production and IL-1ß secretion, but increasing cellular ROS and mitochondrial O2•-, but did not alter iNOS, TNF-α, MHCII, and CD86 protein expression nor affect M2 markers arginase activity, TGF-ß1, and CD206; p,p'-DDE, did not affect marker expression in M0 or M2, supporting that its effects on M1 parameters are not dependent on M0 nor M2 modulation. The decreasing of NO•- production by the p,p'-DDE without altering iNOS levels, Arginase activity, or TNF-α, but increasing cellular ROS and mitochondrial O2 suggests that p,p'-DDE interferes with the iNOS function but not with its transcription. The p,p'-DDE decreasing of IL-1ß secretion, without any effect on TNF-α, suggest that an alteration of specific targets involved in IL-1ß secretion may be affected and related to ROS induction. The p,p'-DDE effect on iNOS function and the IL-1ß secretion process, as the NLRP3 activation, deserves further study.


Subject(s)
Dichlorodiphenyl Dichloroethylene , Macrophages , Animals , Humans , Mice , Arginase/genetics , Arginase/metabolism , Arginase/pharmacology , DDT/metabolism , DDT/pharmacology , Dichlorodiphenyl Dichloroethylene/toxicity , Dichlorodiphenyl Dichloroethylene/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Phenotype , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/genetics
10.
Cells ; 12(7)2023 03 25.
Article in English | MEDLINE | ID: mdl-37048085

ABSTRACT

Parkinson's Disease (PD) is the second most common neurodegenerative disorder seen, especially in the elderly. Tremor, shaking, movement problems, and difficulty with balance and coordination are among the hallmarks, and dopaminergic neuronal loss in substantia nigra pars compacta of the brain and aggregation of intracellular protein α-synuclein are the pathological characterizations. Neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. It is a complex network of interactions comprising immune and non-immune cells in addition to mediators of the immune response. Microglia, the resident macrophages in the CNS, take on the leading role in regulating neuroinflammation and maintaining homeostasis. Under normal physiological conditions, they exist as "homeostatic" but upon pathological stimuli, they switch to the "reactive state". Pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes are used to classify microglial activity with each phenotype having its own markers and released mediators. When M1 microglia are persistent, they will contribute to various inflammatory diseases, including neurodegenerative diseases, such as PD. In this review, we focus on the role of microglia mediated neuroinflammation in PD and also signaling pathways, receptors, and mediators involved in the process, presenting the studies that associate microglia-mediated inflammation with PD. A better understanding of this complex network and interactions is important in seeking new therapies for PD and possibly other neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Humans , Parkinson Disease/metabolism , Microglia/metabolism , Neuroinflammatory Diseases , Neurodegenerative Diseases/metabolism , Inflammation/pathology
11.
Neurotoxicology ; 96: 81-91, 2023 05.
Article in English | MEDLINE | ID: mdl-37019307

ABSTRACT

Microglia, which are the primary inflammatory cells of the brain, can undergo phenotypic switching between M1 and M2 polarization, which have opposing effects on inflammation. Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the nuclear receptor family of ligand-inducible transcription factors, and PPARγ is known to regulate M2 macrophage polarization. Previous studies have shown that the natural pentacyclic triterpenoid ursolic acid (3ß-hydroxy-urs-12-en-28-oic acid; UA) influences microglial activation. Additionally, UA increases tissue inhibitor matrix metalloproteinase 1 (TIMP1), while greatly reducing the release of matrix metalloproteinase 2 (MMP2) and MMP9 in a PPARγ-dependent manner. Here, we examined the anti-inflammatory properties of UA by observing how well it promotes the phenotypic transition of lipopolysaccharide (LPS) and interferon gamma (IFNγ)-activated BV2 microglia from M1 to M2 polarization. To determine if PPARγ is involved in the underlying molecular pathway, we treated rats with UA and the PPARγ inhibitor BADGE. We also investigated the mechanisms by which PPARγ controls transcription from the MMP2 promoter. The in-vitro experiments showed that UA shifted LPS/IFNγ-activated BV2 microglia from the M1 to the M2 phenotype, which was associated with a reduction in the neurotoxic factors MMP2 and MMP9, and an increase in the anti-inflammatory factor TIMP1. Co-treatment with increased MMP2 and MMP9 synthesis while decreasing TIMP1 release, indicating that UA has anti-inflammatory effects on LPS/IFNγ-activated BV2 cells via activation of PPARγ. Next, we found that PPARγ directly influences MMP2 transcriptional activity by identifying the crucial peroxisome proliferator response element (PPRE) among five potential PPREs in the MMP2 promoter. These results suggest that UA has a protective anti-inflammatory effect against neuroinflammatory toxicity, which is exerted by direct activation of PPARγ and selectively modulates microglial polarization and suppresses MMP2 formation.


Subject(s)
Microglia , PPAR gamma , Rats , Animals , PPAR gamma/metabolism , PPAR gamma/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/pharmacology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Signal Transduction , Lipopolysaccharides , Anti-Inflammatory Agents/pharmacology , Phenotype , Ursolic Acid
12.
Bioact Mater ; 19: 38-49, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35415314

ABSTRACT

Ischemic stroke is an acute and serious cerebral vascular disease, which greatly affects people's health and brings huge economic burden to society. Microglia, as important innate immune components in central nervous system (CNS), are double-edged swords in the battle of nerve injury, considering their polarization between pro-inflammatory M1 or anti-inflammatory M2 phenotypes. High mobility group box 1 (HMGB1) is one of the potent pro-inflammatory mediators that promotes the M1 polarization of microglia. 18ß-glycyrrhetinic acid (GA) is an effective intracellular inhibitor of HMGB1, but of poor water solubility and dose-dependent toxicity. To overcome the shortcomings of GA delivery and to improve the efficacy of cerebral ischemia therapy, herein, we designed reactive oxygen species (ROS) responsive polymer-drug conjugate nanoparticles (DGA) to manipulate microglia polarization by suppressing the translocation of nuclear HMGB1. DGA presented excellent therapeutic efficacy in stroke mice, as evidenced by the reduction of infarct volume, recovery of motor function, suppressed of M1 microglia activation and enhanced M2 activation, and induction of neurogenesis. Altogether, our work demonstrates a close association between HMGB1 and microglia polarization, suggesting potential strategies for coping with inflammatory microglia-related diseases.

13.
Metab Brain Dis ; 38(2): 699-708, 2023 02.
Article in English | MEDLINE | ID: mdl-36454504

ABSTRACT

Microglia are resident immune cells in the brain and are closely associated with central nervous system inflammation and neurodegenerative diseases. It is known that mammalian target of rapamycin (mTOR) pathway plays an important role in the polarization of microglia. Castor1 has been identified as the cytosolic arginine sensor for the mTOR complex 1 (mTORC1) pathway, but the role of Castor1 in microglial polarization is still unknown. The purpose of this study was to explore the regulatory effect of Castor1 on microglial polarization and the underlying mechanism. The results demonstrated that Castor1 expression was significantly decreased in lipopolysaccharides (LPS) and interferon (IFN)-γ treated microglia. Castor1 overexpression inhibited the microglia M1 polarization by reducing the expression of M1 related markers. However, the expression of M2-related genes was promoted when Castor1 was overexpressed in IL-4 treated microglia. Mechanistically, Castor1 overexpression inhibited the activation of mTOR signaling pathway. In addition, after treatment with the mTOR activator MHY1485, the inhibitory effect of Castor1 overexpression on M1 polarization was attenuated, indicating that the regulation effects of Castor1 on M1 polarization was dependent on its inhibition of mTOR pathway. We propose that Castor1-mTOR signaling pathway could be considered as a potential target for treatment and intervention of central nervous system-related diseases by regulating microglia polarization.


Subject(s)
Microglia , TOR Serine-Threonine Kinases , Humans , Microglia/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Inflammation/metabolism , Mechanistic Target of Rapamycin Complex 1 , Intracellular Signaling Peptides and Proteins/metabolism
14.
Neural Regen Res ; 18(3): 643-651, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36018189

ABSTRACT

TMEM16F is involved in many physiological processes such as blood coagulation, cell membrane fusion and bone mineralization. Activation of TMEM16F has been studied in various central nervous system diseases. High TMEM16F level has been also found to participate in microglial phagocytosis and transformation. Microglia-mediated neuroinflammation is a key factor in promoting the progression of Alzheimer's disease. However, few studies have examined the effects of TMEM16F on neuroinflammation in Alzheimer's disease. In this study, we established TMEM16F-knockdown AD model in vitro and in vivo to investigate the underlying regulatory mechanism about TMEM16F-mediated neuroinflammation in AD. We performed a Morris water maze test to evaluate the spatial memory ability of animals and detected markers for the microglia M1/M2 phenotype and NLRP3 inflammasome. Our results showed that TMEM16F was elevated in 9-month-old APP/PS1 mice. After TMEM16F knockdown in mice, spatial memory ability was improved, microglia polarization to the M2 phenotype was promoted, NLRP3 inflammasome activation was inhibited, cell apoptosis and Aß plaque deposition in brain tissue were reduced, and brain injury was alleviated. We used amyloid-beta (Aß25-35) to stimulate human microglia to construct microglia models of Alzheimer's disease. The levels of TMEM16F, inducible nitric oxide synthase (iNOS), proinflammatory cytokines and NLRP3 inflammasome-associated biomarkers were higher in Aß25-35 treated group compared with that in the control group. TMEM16F knockdown enhanced the expression of the M2 phenotype biomarkers Arg1 and Socs3, reduced the release of proinflammatory factors interleukin-1, interleukin-6 and tumor necrosis factor-α, and inhibited NLRP3 inflammasome activation through reducing downstream proinflammatory factors interleukin-1ß and interleukin-18. This inhibitory effect of TMEM16F knockdown on M1 microglia was partially reversed by the NLRP3 agonist Nigericin. Our findings suggest that TMEM16F participates in neuroinflammation in Alzheimer's disease through participating in polarization of microglia and activation of the NLRP3 inflammasome. These results indicate that TMEM16F inhibition may be a potential therapeutic approach for Alzheimer's disease treatment.

15.
Toxicol Rep ; 9: 1380-1390, 2022.
Article in English | MEDLINE | ID: mdl-36518431

ABSTRACT

Systemic inflammation affects brain functions. In our previous study in which lipopolysaccharide (LPS) was injected intraperitoneally into mice at sublethal doses, choroid plexus macrophages produced interleukin-1ß and stimulated neighboring stromal cells. Activated stromal cells stimulate choroid plexus epithelial cells, and then choroid plexus epithelium-derived cytokines enter the brain parenchyma and stimulate astrocytes. Stimulated astrocytes then produce cytokines such as CCL11, CXCL10 and G-CSF and change the brain parenchymal microenvironment. However, the effects of an altered brain microenvironment on other brain cells remain to be determined. In the present study, we hypothesized that microglia are activated in response to astrocyte-induced changes in the brain microenvironment. Using the brains of mice treated with intraperitoneal LPS injection, Luminex multiplex cytokine immunoassays revealed increased hippocampal concentrations of CCL11, CXCL10 and G-CSF at 48 h after systemic LPS challenge. The concentrations of all cytokines examined returned to control levels at 72 h after LPS injection, which indicated a resolution of the neuroinflammation. Immunohistochemistry revealed that microglia were hypertrophied in mice at 48 h after systemic LPS challenge. Following isolation of microglial cells from the brain using magnetic-activated cell sorting, gene expression assays were performed with real-time reverse transcriptase-polymerase chain reaction. Isolated microglial cells exhibited much higher gene expression of the receptors for CCL11, CXCL10 and G-CSF than other brain cells. Microglial cells isolated from the brains of mice at 48 h after systemic LPS challenge exhibited the M2-like phenotype. In conclusion, microglial hypertrophy occurs following astrocytic reactions in a mouse model of sublethal endotoxemia-induced systemic inflammation, and hypertrophic microglia are polarized toward the M2-like phenotype and involved in the resolution of neuroinflammation.

16.
Int Immunopharmacol ; 113(Pt A): 109290, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36252498

ABSTRACT

There are two distinct phenotypes of activated microglia, pro-inflammatory M1 and anti-inflammatory M2. Accumulating evidence indicates that shifting the microglial polarization from M1 to M2 is a potential strategy for the treatment of neuroinflammation-associated brain diseases, including ischemic stroke. Cycloastragenol (CAG) is a hydrolysis product of astragaloside IV, the major active component of Astragalus radix. We have previously demonstrated that CAG has anti-inflammatory effect in a mouse model of ischemic stroke. This study investigated the effect of CAG on the phenotype polarization of microglia in lipopolysaccharide (LPS)-stimulated BV-2 mouse microglial cells and ischemic stroke mice. In LPS-treated BV-2 cells, we found that CAG significantly reduced the expression of M1 markers, including pro-inflammatory cytokines and enzymes. In contrast, CAG promoted the expression of M2 markers, including anti-inflammatory cytokines and growth factor. In addition, CAG inhibited the activation of nuclear factor-κB (NF-κB) and enhanced the activation of nuclear factor E2-related factor 2 (Nrf2) and the expression of its downstream heme oxygenase-1 (HO-1). Furthermore, CAG also inhibited levels of M1 markers, promoted those of M2 markers, and enhanced Nrf2 activation and HO-1 expression in ischemic mouse brain. Importantly, the effect of CAG on M2 markers, but not M1 markers, was reversed by Nrf2 siRNA in LPS-stimulated BV-2 cells. Together, our results suggested that CAG promoted microglial M2 and suppressed M1 polarization through activating Nrf2 and inhibiting NF-κB, respectively, in LPS-stimulated BV-2 cells and ischemic mouse brain. CAG is a promising candidate for the treatment of neuroinflammation-related diseases, including ischemic stroke.


Subject(s)
Ischemic Stroke , Sapogenins , Animals , Mice , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Lipopolysaccharides , Microglia , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Sapogenins/pharmacology
17.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35806302

ABSTRACT

Microglial polarization to the M1 phenotype (classically activated) or the M2 phenotype (alternatively activated) is critical in determining the fate of immune responses in neurodegenerative diseases (NDs). M1 macrophages contribute to neurotoxicity, neuronal and synaptic damage, and oxidative stress and are the first line of defense, and M2 macrophages elicit an anti-inflammatory response to regulate neuroinflammation, clear cell debris, and promote neuroregeneration. Various studies have focused on the ability of natural compounds to promote microglial polarization from the M1 phenotype to the M2 phenotype in several diseases, including NDs. However, studies on the roles of fatty acids in microglial polarization and their implications in NDs are a rare find. Most of the studies support the role of polyunsaturated fatty acids (PUFAs) in microglial polarization using cell and animal models. Thus, we aimed to collect data and provide a narrative account of microglial types, markers, and studies pertaining to fatty acids, particularly PUFAs, on microglial polarization and their neuroprotective effects. The involvement of only PUFAs in the chosen topic necessitates more in-depth research into the role of unexplored fatty acids in microglial polarization and their mechanistic implications. The review also highlights limitations and future challenges.


Subject(s)
Neurodegenerative Diseases , Neuroprotective Agents , Animals , Cell Polarity , Fatty Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Macrophages/metabolism , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology
18.
Pharmacol Rep ; 74(5): 909-919, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35796871

ABSTRACT

BACKGROUND: Microglia play a major role in the development of brain inflammation after central nervous system injury. On the other hand, microglia also participate in the repair process. The dualistic role of these cells results from the fact that various states of their activation are associated with specific phenotypes. The M1 phenotype is responsible for the production of proinflammatory mediators, whereas the M2 microglia release anti-inflammatory and trophic factors and take part in immunosuppressive and neuroprotective processes. The histone deacetylase inhibitor sodium butyrate (SB) shows anti-inflammatory and neuroprotective effects in some animal models of brain injury. The aim of this study was to examine the effects of sodium butyrate on the proliferation and M1/M2 polarization of primary microglial cells after oxygen and glucose deprivation (OGD) in vitro. METHODS: Primary microglial cultures were prepared from 1-day-old rats, subjected to the OGD procedure and treated with SB (0.1 mM, 1 mM and 10 mM). The effect of OGD and SB on microglial proliferation was assessed by double immunofluorescence, and microglial phenotypes were evaluated by qPCR. RESULTS: The OGD procedure stimulated the proliferation of microglia after 24 h of culturing, and SB treatment reduced the division of these cells. This effect was inversely proportional to the SB concentration. The OGD procedure increased proinflammatory CD86 and IL1ß gene expression and reduced the expression of the anti-inflammatory M2 markers arginase and CD200 in microglia. CONCLUSIONS: SB can change the polarization of microglia after OGD from an unfavourable M1 to a beneficial M2 phenotype. Our results show that SB is a potential immunosuppressive agent that can modulate microglial activation stimulated by ischaemic-like conditions.


Subject(s)
Microglia , Neuroprotective Agents , Rats , Animals , Butyric Acid/pharmacology , Butyric Acid/metabolism , Histone Deacetylase Inhibitors/pharmacology , Glucose/metabolism , Neuroprotective Agents/pharmacology , Oxygen/metabolism , Arginase/metabolism , Arginase/pharmacology , Immunosuppressive Agents/pharmacology
19.
J Agric Food Chem ; 70(23): 7110-7121, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35652418

ABSTRACT

ß-d-glucans have the potential of serving as both macrophage-targeted carriers and immune stimulators via inducing trained immunity in macrophages. In this study, a carboxymethylated ß-glucan from mushroom sclerotium of Pleurotus tuber-regium (CMPTR) was combined with iron oxide nanoparticles (IONPs) to form nanocomplexes (CMPTR/IONPs) with particle size around 193 ± 7 nm, which could exert a concerted effect on inducing proinflammatory M1 phenotype macrophages for immunotherapy. This nanocomplex exhibited good stability and low cytotoxicity (over 80% cellular viability of RAW 264.7 and THP-1) and higher cellular uptake by murine macrophages compared with B16F10 cells (p < 0.05). CMPTR/IONPs could convert M2-like bone marrow-derived macrophages into M1 phenotypes with upregulated expression of pro-inflammatory cytokines (IL12 and TNF-α, p < 0.05) and reduced immune-suppressive cytokines (IL10 and TGF-ß, p < 0.05). Such polarization was mediated by the combined signaling regulatory factors, including IONP-stimulated IRF5 and CMPTR-triggered TLRs-NF-κB pathways (p < 0.05). Accordingly, CMPTR could have a dual function as a macrophage-targeting carrier for IONPs and an immunostimulant to induce inflammatory M1 macrophage polarization for immunotherapy.


Subject(s)
Agaricales , Glucans , Agaricales/metabolism , Animals , Cytokines/genetics , Glucans/metabolism , Immunotherapy , Interferon Regulatory Factors/metabolism , Macrophages , Magnetic Iron Oxide Nanoparticles , Mice
20.
Int J Toxicol ; 41(5): 412-419, 2022.
Article in English | MEDLINE | ID: mdl-35652623

ABSTRACT

Macrophage play important role in acute lung injury (ALI). This study aims to explore the possible role of Shp2 in regulating macrophage 1 (M1) in ALI progression. ALI was induced in rats by intravenous injection of lipopolysacharide (LPS). Lentivirus was used to knock down Shp2 expression. Lungs from LPS-induced ALI rats were evaluated by H&E staining and wet/dry lung weight ratio (W/D ratio) measurement. The expression of inflammatory cytokines IL-1ß, TNF-α and IL-6 in bronchoalveolar lavage fluid were detected by ELISA. The expressions of M1 biomarker (iNOS) and macrophage 2 (M2) biomarker (Arg-1) in lung tissues and macrophages were measured by immunofluorescence and western blot. The ratio of M2/M1 was detected by flow cytometry. Inflammatory cytokines were highly expressed in ALI rat models, in which elevated expression of iNOS and decreased Arg-1 expression were detected. Shp2 was found to be highly expressed in lung tissues of ALI rat models. LPS treatment in NR8383 cells lead to increased M1 phenotype and elevated expression of Shp2. Suppression on Shp2 expression can counteract the LPS-induced effect and further attenuate ALI progression. Evidence collected from ALI rat and cell models showed that suppression Shp2 expression in macrophages can inhibit M1 phenotype to attenuate ALI progression.


Subject(s)
Acute Lung Injury , Macrophages, Alveolar , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Biomarkers/metabolism , Cytokines/metabolism , Lipopolysaccharides/toxicity , Lung , Macrophages , Macrophages, Alveolar/metabolism , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Rats
SELECTION OF CITATIONS
SEARCH DETAIL