Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.232
Filtrar
1.
PLoS One ; 19(1): e0270779, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38271449

RESUMEN

Macrophages show high plasticity and result in heterogenic subpopulations or polarized states identified by specific cellular markers. These immune cells are typically characterized as pro-inflammatory, or classically activated M1, and anti-inflammatory, or alternatively activated M2. However, a more precise definition places them along a spectrum of activation where they may exhibit a number of pro- or anti-inflammatory roles. To understand M1-M2 dynamics in the context of a localized response and explore the results of different mathematical modeling approaches based on the same biology, we utilized two different modeling techniques, ordinary differential equation (ODE) modeling and agent-based modeling (ABM), to simulate the spectrum of macrophage activation to general pro- and anti-inflammatory stimuli on an individual and multi-cell level. The ODE model includes two hallmark pro- and anti-inflammatory signaling pathways and the ABM incorporates similar M1-M2 dynamics but in a spatio-temporal platform. Both models link molecular signaling with cellular-level dynamics. We then performed simulations with various initial conditions to replicate different experimental setups. Similar results were observed in both models after tuning to a common calibrating experiment. Comparing the two models' results sheds light on the important features of each modeling approach. When more data is available these features can be considered when choosing techniques to best fit the needs of the modeler and application.


Asunto(s)
Activación de Macrófagos , Macrófagos , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Antiinflamatorios/metabolismo , Transducción de Señal
2.
Mediators Inflamm ; 2023: 8821610, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37332618

RESUMEN

Macrophages are innate immune cells in the organism and can be found in almost tissues and organs. They are highly plastic and heterogeneous cells and can participate in the immune response, thereby playing a crucial role in maintaining the immune homeostasis of the body. It is well known that undifferentiated macrophages can polarize into classically activated macrophages (M1 macrophages) and alternatively activated macrophages (M2 macrophages) under different microenvironmental conditions. The directions of macrophage polarization can be regulated by a series of factors, including interferon, lipopolysaccharide, interleukin, and noncoding RNAs. To elucidate the role of macrophages in various autoimmune diseases, we searched the literature on macrophages with the PubMed database. Search terms are as follows: macrophages, polarization, signaling pathways, noncoding RNA, inflammation, autoimmune diseases, systemic lupus erythematosus, rheumatoid arthritis, lupus nephritis, Sjogren's syndrome, Guillain-Barré syndrome, and multiple sclerosis. In the present study, we summarize the role of macrophage polarization in common autoimmune diseases. In addition, we also summarize the features and recent advances with a particular focus on the immunotherapeutic potential of macrophage polarization in autoimmune diseases and the potentially effective therapeutic targets.


Asunto(s)
Artritis Reumatoide , Lupus Eritematoso Sistémico , Nefritis Lúpica , Humanos , Macrófagos/metabolismo , Inflamación/metabolismo , Nefritis Lúpica/metabolismo , Artritis Reumatoide/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Activación de Macrófagos/fisiología
3.
BMB Rep ; 55(11): 519-527, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36195564

RESUMEN

Macrophage activation has long been implicated in a myriad of human pathophysiology, particularly in the context of the dysregulated capacities of an unleashing intracellular or/and extracellular inflammatory response. A growing number of studies have functionally coupled the macrophages' inflammatory capacities with dynamic metabolic reprogramming which occurs during activation, albeit the results have been mostly interpreted through classic metabolism point of view; macrophages take advantage of the rewired metabolism as a source of energy and for biosynthetic precursors. However, a specific subset of metabolic products, namely immune-modulatory metabolites, has recently emerged as significant regulatory signals which control inflammatory responses in macrophages and the relevant extracellular milieu. In this review, we introduce recently highlighted immuno-modulatory metabolites, with the aim of understanding their physiological and pathological relevance in the macrophage inflammatory response. [BMB Reports 2022; 55(11): 519-527].


Asunto(s)
Activación de Macrófagos , Macrófagos , Humanos , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Inmunomodulación , Inflamación/metabolismo
4.
Res Vet Sci ; 145: 91-101, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35180662

RESUMEN

Brucella are serious intracellular pathogens that parasitize macrophages and cause persistent infection in humans and animals. Although macrophages are an important bridge between natural and acquired immunity, their role in Brucella infection is not completely clear. Recently, studies have reported that Brucella can induce macrophage polarization, although the specific molecular mechanism involved is not known. Therefore, in the current study the replication ability of Brucella melitensis strain M5 (Brucella M5) was examined as well as its macrophage polarization and cytokine production, in a host. The role of Signal transducers and activators of transcription 6 (STAT6) in macrophage polarization induced by Brucella infection was also investigated. The results showed that Brucella M5 survived in vivo for a prolonged period of time and caused damage to the spleen and uterus tissues. The expression of type M2 cytokines was induced after Brucella M5 infection. Immunohistochemistry showed that STAT6 was upregulated in spleen and uterus tissues. At the cellular level, Brucella M5 induced macrophagetransformation from M1 to M2-type during the later stage of infection. When STAT6 was silenced, the polarization of M2-type was inhibited, and the intracellular survival rate of Brucella decreased significantly. In conclusion, these findings demonstrate that STAT6 is the key factor regulates M2 polarization of macrophages and promotes the intracellular survival of Brucella in the late stage of infection and provides an explanation of the mechanism responsible for persistent Brucella infection.


Asunto(s)
Brucella melitensis , Macrófagos , Animales , Citocinas/metabolismo , Activación de Macrófagos/fisiología , Transducción de Señal
5.
Biochem Biophys Res Commun ; 598: 32-39, 2022 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-35151201

RESUMEN

Alveolar macrophage activation and apoptosis are vital contributors to sepsis-associated acute lung injury (ALI). However, the mechanisms of alveolar macrophage activation are yet to be clarified. Death-associated protein kinase 1 (DAPK1) is one of the potential candidates that play crucial roles in regulating alveolar macrophage inflammation. Herein, we found that primary human bone mesenchymal stem cell (BMSC)-derived extracellular vesicles (EVs) antagonize LPS-induced inflammation in the THP-1 human macrophage-like cell line. Mechanistically, LPS stimulation elevates the expression of DAPK1 and the inflammation markers in THP-1 cells, while BMSC-derived EVs inhibit the expression of DAPK1 and inflammation through delivering miR-191, which can target the 3'-UTR of the DAPK1 mRNA and therefore suppress its translation. The importance of DAPK1 in the activation of THP-1 is also stressed in this study. Our findings provide evidence that BMSC-derived EVs regulate the alveolar macrophage inflammation and highlight BMSC-derived EVs as a potential vehicle to deliver biomacromolecules to macrophages.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/genética , Vesículas Extracelulares/genética , Inflamación/etiología , Activación de Macrófagos/fisiología , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Regiones no Traducidas 3' , Medios de Cultivo Condicionados/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Lipopolisacáridos/toxicidad , Activación de Macrófagos/genética , MicroARNs/farmacología , Regiones Promotoras Genéticas , Células THP-1
6.
Neurobiol Dis ; 163: 105608, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34979258

RESUMEN

Tissue damage after spinal cord injury (SCI) elicits a robust inflammatory cascade that fails to resolve in a timely manner, resulting in impaired wound healing and cellular regeneration. This inflammatory response is partly mediated by infiltrating immune cells, including macrophages. As professional phagocytes, macrophages initially play an important role in debris clearance at the injury site, which would be necessary for proper tissue regeneration. After SCI, most macrophages become filled with lipid droplets due to excessive uptake of lipid debris, assuming a "foamy" phenotype that is associated with a proinflammatory state. Myelin has been assumed to be the main source of lipid that induces foamy macrophage formation after injury given its abundance in the spinal cord. This assumption has led to the widespread use of purified myelin treatment to model foamy macrophage formation in vitro. However, the assumption that myelin is necessary for foamy macrophage formation remains untested. To this end, we developed a novel foamy macrophage assay utilizing total spinal cord homogenate to include all sources of lipid present at the injury site. Using the myelin basic protein knockout (MBP KO, i.e., Shiverer) mice that lack myelin, we investigated lipid accumulation in foamy macrophages. Primary macrophages treated with myelin-deficient spinal cord homogenate still formed large lipid droplets typically observed in foamy macrophages, although to a lesser degree than cells treated with normal homogenate. Similarly, MBP KO mice subjected to contusive spinal cord injury also formed foamy macrophages that exhibited reduced lipid content and associated with improved histological outcomes and reduced immune cell infiltration. Therefore, the absence of myelin does not preclude foamy macrophage formation, indicating that myelin is not the only major source of lipid that contributes this pathology, even though myelin may alter certain aspects of its inflammatory profile.


Asunto(s)
Macrófagos/patología , Vaina de Mielina/patología , Traumatismos de la Médula Espinal/patología , Médula Espinal/patología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inflamación/metabolismo , Inflamación/patología , Lípidos , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Masculino , Ratones , Vaina de Mielina/metabolismo , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo
7.
Oxid Med Cell Longev ; 2022: 9964689, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35096275

RESUMEN

OBJECTIVE: Apolipoprotein (a)/lipoprotein(a) (Lp(a)), a major carrier of oxidized phospholipids, and α7-nicotinic acetylcholine receptor (α7-nAChR) may play an important role in the development of coronary artery spasm (CAS). In CAS, the association between Lp(a) and the α7-nAChR-modulated inflammatory macrophage polarization and activation and smooth muscle cell dysfunction remains unknown. METHODS: We investigated the relevance of Lp(a)/α7-nAChR signaling in patient monocyte-derived macrophages and human coronary artery smooth muscle cells (HCASMCs) using expression profile correlation analyses, fluorescence-assisted cell sorting flow cytometry, immunoblotting, quantitative real-time polymerase chain reaction, and clinicopathological analyses. RESULTS: There are increased serum Lp(a) levels (3.98-fold, p = 0.011) and macrophage population (3.30-fold, p = 0.013) in patients with CAS compared with patients without CAS. Serum Lp(a) level was positively correlated with high-sensitivity C-reactive protein (r 2 = 0.48, p < 0.01), IL-6 (r 2 = 0.38, p = 0.03), and α7-nAChR (r 2 = 0.45, p < 0.01) in patients with CAS, but not in patients without CAS. Compared with untreated or low-density lipoprotein- (LDL-) treated macrophages, Lp(a)-treated macrophages exhibited markedly enhanced α7-nAChR mRNA expression (p < 0.01) and activity (p < 0.01), in vitro and ex vivo. Lp(a) but not LDL preferentially induced CD80+ macrophage (M1) polarization and reduced the inducible nitric oxide synthase expression and the subsequent NO production. While shRNA-mediated loss of α7-nAChR function reduced the Lp(a)-induced CD80+ macrophage pool, both shRNA and anti-IL-6 receptor tocilizumab suppressed Lp(a)-upregulated α7-nAChR, p-p38 MAPK, IL-6, and RhoA-GTP protein expression levels in cultures of patient monocyte-derived macrophages and HCASMCs. CONCLUSIONS: Elevated Lp(a) levels upregulate α7-nAChR/IL-6/p38 MAPK signaling in macrophages of CAS patients and HCASMC, suggesting that Lp(a)-triggered inflammation mediates CAS through α7-nAChR/p38 MAPK/IL-6/RhoA-GTP signaling induction, macrophage M1 polarization, and HCASMC activation.


Asunto(s)
Apoproteína(a)/efectos adversos , Vasos Coronarios/patología , Interleucina-6/metabolismo , Lipoproteína(a)/efectos adversos , Sistema de Señalización de MAP Quinasas/fisiología , Activación de Macrófagos/fisiología , Espasmo/patología , Anciano , Estudios de Cohortes , Femenino , Humanos , Inflamación , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Transfección
8.
Mol Cell Biol ; 42(2): e0028221, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34842443

RESUMEN

Glycemic variability has been considered one of the predictors of diabetes complications in patients with diabetes mellitus (DM). In this work, we evaluated whether glycemic variability induces cardiac fibrosis through regulating cardiac fibroblast activation and macrophage polarization. Moreover, we determined whether glucose transporter sodium-glucose cotransporter 1 (SGLT1) plays an important role in this process. Glycemic variability-induced mice were established using DM mice (GVDM mice), and intermittent high-glucose (IHG) treatment was used to simulate glycemic variability in RAW264.7 macrophages and cardiac fibroblasts. The short hairpin RNA for SGLT1 was used to knock down SGLT1. The results showed that glycemic variability aggravated the cardiac fibrosis in GVDM mice. Additionally, glycemic variability promoted the expression of fibrogenic cytokine and the extracellular matrix proteins in left ventricular tissues and cardiac fibroblasts. GVDM mice showed a higher incidence of macrophage infiltration and M1 polarization in left ventricular tissues. Moreover, IHG-promoted RAW264.7 macrophages tended to differentiate to M1 phenotype. SGLT1 knockdown alleviated cardiac fibrosis in GVDM mice and inhibited activations of cardiac fibroblast and macrophage M1 polarization. Our results indicated that glycemic variability aggravates cardiac fibrosis through activating cardiac fibroblast and macrophage M1 polarization, which could be partially inhibited by SGLT1 knockdown.


Asunto(s)
Glucemia/metabolismo , Fibroblastos/metabolismo , Activación de Macrófagos/fisiología , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Animales , Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamiento del Gen/métodos , Glucosa/metabolismo , Corazón/fisiopatología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Miocardio/metabolismo , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/metabolismo
9.
Naunyn Schmiedebergs Arch Pharmacol ; 395(1): 13-25, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34851450

RESUMEN

Macrophages are myeloid immune cells, present in virtually all tissues which exhibit considerable functional plasticity and diversity. Macrophages are often subdivided into two distinct subsets described as classically activated (M1) and alternatively activated (M2) macrophages. It has recently emerged that metabolites regulate the polarization and function of macrophages by altering metabolic pathways. These metabolites often cannot freely pass the cell membrane and are therefore transported by the corresponding metabolite transporters. Here, we reviewed how glucose, glutamate, lactate, fatty acid, and amino acid transporters are involved in the regulation of macrophage polarization. Understanding the interactions among metabolites, metabolite transporters, and macrophage function under physiological and pathological conditions may provide further insights for novel drug targets for the treatment of macrophage-associated diseases. In Brief Recent studies have shown that the polarization and function of macrophages are regulated by metabolites, most of which cannot pass freely through biofilms. Therefore, metabolite transporters required for the uptake of metabolites have emerged seen as important regulators of macrophage polarization and may represent novel drug targets for the treatment of macrophage-associated diseases. Here, we summarize the role of metabolite transporters as regulators of macrophage polarization.


Asunto(s)
Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , Membrana Celular/metabolismo , Humanos
10.
Cytokine ; 149: 155726, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34666235

RESUMEN

BACKGROUND: There is growing evidence that exposure to low-grade inflammation may be associated with adverse health outcomes. METHODS: We conducted a cross-sectional study within the California Teachers Study prospective cohort, among female participants who had completed a questionnaire that asked about their health behaviors (e.g., diabetes, physical activity, body mass index, medication use) and who had donated blood within a year of their questionnaire. 822 women with stored serum were evaluated for 16 immune biomarkers. In addition, four immune pathways were constructed: Th1, pro-inflammatory/macrophage activation, B-cell activation, and T-cell activation. Odds ratios (ORs) and 95% confidence intervals (CI) for the association between host characteristics and immune biomarkers were assessed using logistic regression models. RESULT: Compared to women of a normal BMI, obese women (>30 kg/m2) were positively associated with sTNFR2, CD27, IL6, CXCL13, sIL-2Rα, and IL6Ra levels above the median, with odds ratios ranging from 1.5 to 6.0. The pro-inflammatory/macrophage activation pathway was positively associated with diabetes (OR = 2.12, 95% CI = 1.14-3.95), fueled by individual associations between diabetes and sTNF-R2, TNFα and sCD27. Physical activity was inversely associated with sTNF-R2, TNFα, CXCL13, IL6, IL10, and IFN-γ levels, particularly for the highest category of activity (5.88+ hours/week) (ORs = 0.32-0.69). In pathway-based analyses, the Th1 pathway which includes decreased levels of IL4 and IL10 was positively associated with elevated physical activity (OR = 1.5). In contrast, the pro-inflammatory, B- and T-cell activation pathways were positively associated with higher BMI (OR ranging from 1.6 to 3) and inversely associated with increasing levels of physical activity. CONCLUSIONS: Several host characteristics were associated with circulating levels of immune biomarkers, including markers of inflammation. Further understanding of associations between immune marker profiles with human disease are warranted.


Asunto(s)
Biomarcadores/metabolismo , Inflamación/metabolismo , Linfocitos B/metabolismo , Índice de Masa Corporal , Estudios Transversales , Citocinas/metabolismo , Ejercicio Físico/fisiología , Femenino , Humanos , Modelos Logísticos , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Oportunidad Relativa , Estudios Prospectivos , Linfocitos T/metabolismo
11.
Hepatology ; 75(3): 550-566, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34510498

RESUMEN

BACKGROUND AND AIMS: Hepatic ischemia-reperfusion injury (IRI) is the leading cause of early posttransplantation organ failure as mitochondrial respiration and ATP production are affected. A shortage of donors has extended liver donor criteria, including aged or steatotic livers, which are more susceptible to IRI. Given the lack of an effective treatment and the extensive transplantation waitlist, we aimed at characterizing the effects of an accelerated mitochondrial activity by silencing methylation-controlled J protein (MCJ) in three preclinical models of IRI and liver regeneration, focusing on metabolically compromised animal models. APPROACH AND RESULTS: Wild-type (WT), MCJ knockout (KO), and Mcj silenced WT mice were subjected to 70% partial hepatectomy (Phx), prolonged IRI, and 70% Phx with IRI. Old and young mice with metabolic syndrome were also subjected to these procedures. Expression of MCJ, an endogenous negative regulator of mitochondrial respiration, increases in preclinical models of Phx with or without vascular occlusion and in donor livers. Mice lacking MCJ initiate liver regeneration 12 h faster than WT and show reduced ischemic injury and increased survival. MCJ knockdown enables a mitochondrial adaptation that restores the bioenergetic supply for enhanced regeneration and prevents cell death after IRI. Mechanistically, increased ATP secretion facilitates the early activation of Kupffer cells and production of TNF, IL-6, and heparin-binding EGF, accelerating the priming phase and the progression through G1 /S transition during liver regeneration. Therapeutic silencing of MCJ in 15-month-old mice and in mice fed a high-fat/high-fructose diet for 12 weeks improves mitochondrial respiration, reduces steatosis, and overcomes regenerative limitations. CONCLUSIONS: Boosting mitochondrial activity by silencing MCJ could pave the way for a protective approach after major liver resection or IRI, especially in metabolically compromised, IRI-susceptible organs.


Asunto(s)
Hígado Graso/metabolismo , Regeneración Hepática/fisiología , Activación de Macrófagos/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales , Chaperonas Moleculares , Daño por Reperfusión/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Silenciador del Gen/fisiología , Rechazo de Injerto/prevención & control , Hígado/metabolismo , Trasplante de Hígado/métodos , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Daño por Reperfusión/prevención & control
12.
Proc Natl Acad Sci U S A ; 118(52)2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34934001

RESUMEN

Biomaterial characteristics such as surface topographies have been shown to modulate macrophage phenotypes. The standard methodologies to measure macrophage response to biomaterials are marker-based and invasive. Raman microspectroscopy (RM) is a marker-independent, noninvasive technology that allows the analysis of living cells without the need for staining or processing. In the present study, we analyzed human monocyte-derived macrophages (MDMs) using RM, revealing that macrophage activation by lipopolysaccharides (LPS), interferons (IFN), or cytokines can be identified by lipid composition, which significantly differs in M0 (resting), M1 (IFN-γ/LPS), M2a (IL-4/IL-13), and M2c (IL-10) MDMs. To identify the impact of a biomaterial on MDM phenotype and polarization, we cultured macrophages on titanium disks with varying surface topographies and analyzed the adherent MDMs with RM. We detected surface topography-induced changes in MDM biochemistry and lipid composition that were not shown by less sensitive standard methods such as cytokine expression or surface antigen analysis. Our data suggest that RM may enable a more precise classification of macrophage activation and biomaterial-macrophage interaction.


Asunto(s)
Lipidómica/métodos , Activación de Macrófagos/fisiología , Macrófagos , Espectrometría Raman/métodos , Materiales Biocompatibles/farmacología , Citocinas/farmacología , Femenino , Humanos , Inmunidad Innata , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino
13.
Biomed Res Int ; 2021: 9910596, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34722776

RESUMEN

Macrophages are important immune cells that participate in the regulation of inflammation in implant dentistry, and their activation/polarization state is considered to be the basis for their functions. The classic dichotomy activation model is commonly accepted, however, due to the discovery of macrophage heterogeneity and more functional and iconic exploration at different technologies; some studies have discovered the shortcomings of the dichotomy model and have put forward the concept of alternative activation models through the application of advanced technologies such as cytometry by time-of-flight (CyTOF), single-cell RNA-seq (scRNA-seq), and hyperspectral image (HSI). These alternative models have great potential to help macrophages divide phenotypes and functional genes.


Asunto(s)
Activación de Macrófagos/inmunología , Macrófagos/clasificación , Macrófagos/fisiología , Animales , Implantación Dental/métodos , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Humanos , Activación de Macrófagos/fisiología , Macrófagos/inmunología , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética
14.
Int Immunopharmacol ; 101(Pt B): 108372, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34810128

RESUMEN

Citrate has a prominent role as a substrate in cellular energy metabolism. Recently, citrate has been shown to drive inflammation. However, the role of citrate in lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains unclear. Here, we aimed to clarify whether extracellular citrate aggravated the LPS-induced ALI and the potential mechanism. Our findings demonstrated that extracellular citrate aggravated the pathological lung injury induced by LPS in mice, characterized by up-regulation of pro-inflammatory factors and over-activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in the lungs. In vitro, we found that citrate treatment significantly augmented the expression of NLRP3 and pro-IL-1ß and enhanced the translocation of NF-κB/p65 into the nucleus. Furthermore, extracellular citrate plus adenosine-triphosphate (ATP) significantly increased the production of reactive oxygen species (ROS) in primary murine macrophages. Inhibiting the production of ROS with a ROS scavenger N-acetyl-L-cysteine (NAC) attenuated the activation of NLRP3 inflammasome. Altogether, we conclude that extracellular citrate may serve as a damage-associated molecular pattern (DAMP) and aggravates LPS-induced ALI by activating the NLRP3 inflammasome.


Asunto(s)
Alarminas/metabolismo , Ácido Cítrico/metabolismo , Lipopolisacáridos/toxicidad , Lesión Pulmonar/inducido químicamente , Activación de Macrófagos/fisiología , Macrófagos/efectos de los fármacos , Adenosina Trifosfato , Animales , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Distribución Aleatoria
15.
Nat Commun ; 12(1): 6535, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764296

RESUMEN

Super-enhancers (SEs) govern macrophage polarization and function. However, the mechanism underlying the signal-dependent latent SEs remodeling in macrophages remains largely undefined. Here we show that the epigenetic reader ZMYND8 forms liquid compartments with NF-κB/p65 to silence latent SEs and restrict macrophage-mediated inflammation. Mechanistically, the fusion of ZMYND8 and p65 liquid condensates is reinforced by signal-induced acetylation of p65. Then acetylated p65 guides the ZMYND8 redistribution onto latent SEs de novo generated in polarized macrophages, and consequently, recruit LSD1 to decommission latent SEs. The liquidity characteristic of ZMYND8 is critical for its regulatory effect since mutations coagulating ZMYND8 into solid compartments disable the translocation of ZMYND8 and its suppressive function. Thereby, ZMYND8 serves as a molecular rheostat to switch off latent SEs and control the magnitude of the immune response. Meanwhile, we propose a phase separation model by which the latent SEs are fine-tuned in a spatiotemporal manner.


Asunto(s)
Proteínas Supresoras de Tumor/metabolismo , Animales , Western Blotting , Movimiento Celular/genética , Movimiento Celular/fisiología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Inmunoprecipitación , Activación de Macrófagos/genética , Activación de Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , RNA-Seq , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Supresoras de Tumor/genética
16.
Neurosci Lett ; 765: 136283, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34624395

RESUMEN

Intracerebral hemorrhage (ICH) is a leading medical problem and has no effective treatment approach up until now. The transcription factor androgen receptor (AR) has been indicated in the cerebrovascular function recently. However, its participation in ICH remains unclear. The present study aims to expound the regulation of AR in microglia/macrophage phenotypes and the secondary brain injury in a rat model with ICH, and to discuss the involved pathway. Following the induction of ICH in rats, we found that ICH led to increased mNSS score, enhanced microglial activity, and promoted levels of inflammatory factors and apoptosis of brain cells. Using microarray analysis, AR was found to be significantly overexpressed in ICH rat brain tissues. AR repressed the transcription of Jumonji d3 (JMJD3, histone 3 demethylase). JMJD3 inhibited the methylation of Botch and promoted the activity of Notch1. JMJD3 hampered microglial activity and ameliorated secondary brain injury in rats, whereas upregulation of AR or downregulation of Botch reversed the protective effects of JMJD3. In conclusion, we found that AR promoted microglial activation and secondary brain injury via transcriptionally repressing JMJD3 and mediating the subsequent Botch/Notch1 pathway, which may provide novel insights into therapeutic options for the treatment of ICH.


Asunto(s)
Hemorragias Intracraneales/metabolismo , Activación de Macrófagos/fisiología , Microglía/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal/fisiología , Animales , Histona Demetilasas con Dominio de Jumonji/metabolismo , Macrófagos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptor Notch1/metabolismo , gamma-Glutamilciclotransferasa/metabolismo
17.
Theranostics ; 11(19): 9243-9261, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34646369

RESUMEN

The coagulation protein tissue factor (TF) regulates inflammation and angiogenesis via its cytoplasmic domain in infection, cancer and diabetes. While TF is highly abundant in the heart and is implicated in cardiac pathology, the contribution of its cytoplasmic domain to post-infarct myocardial injury and adverse left ventricular (LV) remodeling remains unknown. Methods: Myocardial infarction was induced in wild-type mice or mice lacking the TF cytoplasmic domain (TF∆CT) by occlusion of the left anterior descending coronary artery. Heart function was monitored with echocardiography. Heart tissue was collected at different time-points for histological, molecular and flow cytometry analysis. Results: Compared with wild-type mice, TF∆CT had a higher survival rate during a 28-day follow-up after myocardial infarction. Among surviving mice, TF∆CT mice had better cardiac function and less LV remodeling than wild-type mice. The overall improvement of post-infarct cardiac performance in TF∆CT mice, as revealed by speckle-tracking strain analysis, was attributed to reduced myocardial deformation in the peri-infarct region. Histological analysis demonstrated that TF∆CT hearts had in the infarct area greater proliferation of myofibroblasts and better scar formation. Compared with wild-type hearts, infarcted TF∆CT hearts showed less infiltration of proinflammatory cells with concomitant lower expression of protease-activated receptor-1 (PAR1) - Rac1 axis. In particular, infarcted TF∆CT hearts displayed markedly lower ratios of inflammatory M1 macrophages and reparative M2 macrophages (M1/M2). In vitro experiment with primary macrophages demonstrated that deletion of the TF cytoplasmic domain inhibited macrophage polarization toward the M1 phenotype. Furthermore, infarcted TF∆CT hearts presented markedly higher peri-infarct vessel density associated with enhanced endothelial cell proliferation and higher expression of PAR2 and PAR2-associated pro-angiogenic pathway factors. Finally, the overall cardioprotective effects observed in TF∆CT mice could be abolished by subcutaneously infusing a cocktail of PAR1-activating peptide and PAR2-inhibiting peptide via osmotic minipumps. Conclusions: Our findings demonstrate that the TF cytoplasmic domain exacerbates post-infarct cardiac injury and adverse LV remodeling via differential regulation of inflammation and angiogenesis. Targeted inhibition of the TF cytoplasmic domain-mediated intracellular signaling may ameliorate post-infarct LV remodeling without perturbing coagulation.


Asunto(s)
Infarto del Miocardio/patología , Tromboplastina/metabolismo , Remodelación Ventricular/fisiología , Animales , Proliferación Celular/fisiología , Inflamación/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/prevención & control , Dominios Proteicos/fisiología , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Transducción de Señal/fisiología , Tromboplastina/fisiología , Función Ventricular Izquierda/fisiología
18.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34445695

RESUMEN

Accumulating evidence indicates that the molecular pathways mediating wound healing induce cell migration and localization of cytokines to sites of injury. Macrophages are immune cells that sense and actively respond to disturbances in tissue homeostasis by initiating, and subsequently resolving, inflammation. Hypoxic conditions generated at a wound site also strongly recruit macrophages and affect their function. Hypoxia inducible factor (HIF)-1α is a transcription factor that contributes to both glycolysis and the induction of inflammatory genes, while also being critical for macrophage activation. For the latter, HIF-1α regulates sphingosine 1-phosphate (S1P) to affect the migration, activation, differentiation, and polarization of macrophages. Recently, S1P and HIF-1α have received much attention, and various studies have been performed to investigate their roles in initiating and resolving inflammation via macrophages. It is hypothesized that the HIF-1α/S1P/S1P receptor axis is an important determinant of macrophage function under inflammatory conditions and during disease pathogenesis. Therefore, in this review, biological regulation of monocytes/macrophages in response to circulating HIF-1α is summarized, including signaling by S1P/S1P receptors, which have essential roles in wound healing.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Cicatrización de Heridas/fisiología , Animales , Diferenciación Celular/genética , Movimiento Celular/fisiología , Citocinas/metabolismo , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Inflamación/metabolismo , Lisofosfolípidos/fisiología , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Macrófagos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Esfingosina/metabolismo , Esfingosina/fisiología
19.
Mol Nutr Food Res ; 65(20): e2001010, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34390195

RESUMEN

SCOPE: Konjac glucomannan oligosaccharides (KMOS) are prebiotics and may improve intestinal immunity through modulation of macrophage function. However, the underlying molecular mechanisms were unclear. METHODS AND RESULTS: Using a mouse model of dextran sulfated sodium (DSS)-induced acute colitis, the study demonstrates here that KMOS (400 mg-1 kg-1 d-1 ) can ameliorate intestinal inflammation in a macrophage dependent manner. Oral exposure to KMOS prevents DSS-induced intestinal pathology, improves epithelial integrity, and decreases accumulation of colonic inflammatory leukocytes and cytokines. The therapeutic effects of KMOS are dependent on the function of macrophages, as depletion of macrophages abolished the effects. In colonic lamina propria of DSS-treated mice, as well as in vitro culture of bone marrow derived macrophages (BMDMs), KMOS skews reprogramming of classically activated macrophages (CAM/M1) into alternatively activated macrophages (AAM/M2). The study further determines that the activation of SIGNR1/phospho-c-Raf (S338)/phospho-p65 (S276)/acetyl-p65 (K310) pathway is responsible for KMOS-induced AAM/M2 polarization. Blockage of SIGNR1 abolishes KMOS-induced AAM/M2 polarization of activated macrophages, expression of phospho-p65 (S276) in colonic macrophages, and alleviation of DSS-induced colitis in mice, suggesting that SIGNR1 is critical for macrophage responses to KMOS. CONCLUSIONS: This study reveals a SIGNR1-mediated macrophage-dependent pathway that supports regulatory function of KMOS in host immunity and intestinal homeostasis.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Colitis/prevención & control , Lectinas Tipo C/fisiología , Activación de Macrófagos/fisiología , Mananos/farmacología , Oligosacáridos/farmacología , Prebióticos , Receptores de Superficie Celular/fisiología , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
20.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209797

RESUMEN

Macrophages play critical roles in both innate and adaptive immunity and are known for their high plasticity in response to various external signals. Macrophages are involved in regulating systematic iron homeostasis and they sequester iron by phagocytotic activity, which triggers M1 macrophage polarization and typically exerts antitumor effects. We previously developed a novel cryo-thermal therapy that can induce the mass release of tumor antigens and damage-associated molecular patterns (DAMPs), promoting M1 macrophage polarization. However, that study did not examine whether iron released after cryo-thermal therapy induced M1 macrophage polarization; this question still needed to be addressed. We hypothesized that cryo-thermal therapy would cause the release of a large quantity of iron to augment M1 macrophage polarization due to the disruption of tumor cells and blood vessels, which would further enhance antitumor immunity. In this study, we investigated iron released in primary tumors, the level of iron in splenic macrophages after cryo-thermal therapy and the effect of iron on macrophage polarization and CD4+ T cell differentiation in metastatic 4T1 murine mammary carcinoma. We found that a large amount of iron was released after cryo-thermal therapy and could be taken up by splenic macrophages, which further promoted M1 macrophage polarization by inhibiting ERK phosphorylation. Moreover, iron promoted DC maturation, which was possibly mediated by iron-induced M1 macrophages. In addition, iron-induced M1 macrophages and mature DCs promoted the differentiation of CD4+ T cells into the CD4 cytolytic T lymphocytes (CTL) subset and inhibited differentiation into Th2 and Th17 cells. This study explains the role of iron in cryo-thermal therapy-induced antitumor immunity from a new perspective.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Crioterapia/efectos adversos , Hierro/metabolismo , Hierro/farmacología , Activación de Macrófagos/efectos de los fármacos , Linfocitos T Citotóxicos/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Femenino , Quelantes del Hierro/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Macrófagos/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Linfocitos T Citotóxicos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...