Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros











Intervalo de año de publicación
1.
Ocul Surf ; 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39306240

RESUMEN

PURPOSE: To evaluate the effects of dry eye on conjunctival immune cell number and transcriptional profiles with attention to mononuclear phagocytes. METHODS: Expression profiling was performed by single-cell RNA sequencing on sorted conjunctival immune cells from non-stressed and C57BL/6 mice subjected to desiccating stress (DS). Monocle 3 modeled cell trajectory, scATAC-seq assessed chromatin accessibility and IPA identified canonical pathways. Inflammation and goblet cells were measured after depletion of MRC1+ MΦs with mannosylated clodronate liposomes. RESULTS: Mononuclear phagocytes (monocytes, MΦs, DCs) comprised 72% of immune cells and showed the greatest changes with DS. Distinct DS induced gene expression patterns were seen in phagocytes classified by expression of Ccr2 and [Timd4, Lyve1, Folr2 (TLR)]. Expression of phagocytosis/efferocytosis genes increased in TLF+CCR2- MΦs. Monocytes showed the highest expression of Ace, Cx3cr1, Vegfa, Ifngr1,2, and Stat1 and TLF-CCR2+ cells expressed higher levels of inflammatory mediators (Il1a, Il1b, Il1rn, Nfkb1, Ccl5, MHCII, Cd80, Cxcl10, Icam1). A trajectory from monocyte precursors branched to terminate in regulatory MΦs or in mDCs via transitional MΦ and cDC clusters. Activated pathways in TLF+ cells include phagocytosis, PPAR/RXRα activation, IL-10 signaling, alternate MΦ activation, while inflammatory pathways were suppressed. Depletion of MRC1+ MΦs increased IL-17 and IFN-γ expression and cytokine-expressing T cells, reduced IL-10 and worsened goblet loss. CONCLUSIONS: Dryness stimulates distinct gene expression patterns in conjunctival phagocytes, increasing expression of regulatory genes in TLF+ cells regulated in part by RXRα, and inflammatory genes in CCR2+ cells. Regulatory MΦs depletion worsens DS induced inflammation and goblet cell loss.

2.
Cell Commun Signal ; 22(1): 395, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39123188

RESUMEN

Esophageal cancer is common worldwide, with ESCC being the most frequent tumor in East Asia. Tumor-associated macrophages are an important component of the ESCC microenvironment. SUMOylation is a post-translational modification of proteins, and SUMO-specific proteases (SENPs) play an important role in de-SUMOylation. In human patients, we discovered that the levels of SENP3 were upregulated in the tumor-associated macrophages. Furthermore, the loss of SENP3 enhanced the alternative activation of macrophages in the 4-NQO-induced ESCC mice model. This is the first study to identify SENP3-mediated macrophage polarization via the de-SUMOylation of interferon regulatory factor 4 (IRF4) at the K349 site. Alternative activation of macrophages increases the migration and invasion potential of ESCC cells and promotes their progression in vivo. Moreover, patients with relatively low SENP3 expression in macrophages exhibit higher primary PET SUVmax value and lymph node metastasis rates. In summary, this study revealed that SENP3-mediated IRF4 de-SUMOylation is crucial for the alternative activation of macrophages and influences the progression of ESCC.


Asunto(s)
Cisteína Endopeptidasas , Factores Reguladores del Interferón , Activación de Macrófagos , Sumoilación , Animales , Femenino , Humanos , Masculino , Ratones , Línea Celular Tumoral , Movimiento Celular , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Progresión de la Enfermedad , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Macrófagos/metabolismo , Macrófagos Asociados a Tumores/metabolismo
3.
Inflamm Res ; 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052063

RESUMEN

BACKGROUND: Inflammation, a biological response of the immune system, can be triggered by various factors such as pathogens, damaged cells, and toxic compounds. These factors can lead to chronic inflammatory responses, potentially causing tissue damage or disease. Both infectious and non-infectious agents, as well as cell damage, activate inflammatory cells and trigger common inflammatory signalling pathways, including NF-κB, MAPK, and JAK-STAT pathways. These pathways are activated through adaptor proteins, which possess distinct protein binding domains that connect corresponding interacting molecules to facilitate downstream signalling. Adaptor molecules have gained widespread attention in recent years due to their key role in chronic inflammatory diseases. METHODS: In this review, we explore potential pharmacological agents that can be used to target adaptor molecules in chronic inflammatory responses. A comprehensive analysis of published studies was performed to obtain information on pharmacological agents. CONCLUSION: This review highlights the therapeutic strategies involving small molecule inhibitors, antisense oligonucleotide therapy, and traditional medicinal compounds that have been found to inhibit the inflammatory response and pro-inflammatory cytokine production. These strategies primarily block the protein-protein interactions in the inflammatory signaling cascade. Nevertheless, extensive preclinical studies and risk assessment methodologies are necessary to ensure their safety.

4.
Front Immunol ; 15: 1355012, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38482001

RESUMEN

Macrophages play a central role in initiating, maintaining, and terminating inflammation. For that, macrophages respond to various external stimuli in changing environments through signaling pathways that are tightly regulated and interconnected. This process involves, among others, autoregulatory loops that activate and deactivate macrophages through various cytokines, stimulants, and other chemical mediators. Adaptor proteins play an indispensable role in facilitating various inflammatory signals. These proteins are dynamic and flexible modulators of immune cell signaling and act as molecular bridges between cell surface receptors and intracellular effector molecules. They are involved in regulating physiological inflammation and also contribute significantly to the development of chronic inflammatory processes. This is at least partly due to their involvement in the activation and deactivation of macrophages, leading to changes in the macrophages' activation/phenotype. This review provides a comprehensive overview of the 20 adaptor molecules and proteins that act as negative regulators of inflammation in macrophages and effectively suppress inflammatory signaling pathways. We emphasize the functional role of adaptors in signal transduction in macrophages and their influence on the phenotypic transition of macrophages from pro-inflammatory M1-like states to anti-inflammatory M2-like phenotypes. This endeavor mainly aims at highlighting and orchestrating the intricate dynamics of adaptor molecules by elucidating the associated key roles along with respective domains and opening avenues for therapeutic and investigative purposes in clinical practice.


Asunto(s)
Citocinas , Macrófagos , Humanos , Citocinas/metabolismo , Transducción de Señal , Inflamación , Proteínas Adaptadoras Transductoras de Señales/metabolismo
5.
Front Immunol ; 15: 1201439, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38482013

RESUMEN

Introduction: Obesity is associated with chronic low-grade inflammation of adipose tissue (AT) and an increase of AT macrophages (ATMs) that is linked to the onset of type 2 diabetes. We have recently shown that neutralization of interleukin (IL)-6 in obese AT organ cultures inhibits proliferation of ATMs, which occurs preferentially in alternatively activated macrophage phenotype. Methods: In this study, we investigated AT biology and the metabolic phenotype of mice with myeloid cell-specific IL-6Rα deficiency (Il6ra Δmyel) after normal chow and 20 weeks of high-fat diet focusing on AT inflammation, ATM polarization and proliferation. Using organotypical AT culture and bone marrow derived macrophages (BMDMs) of IL-4Rα knockout mice (Il4ra -/-) we studied IL-6 signaling. Results: Obese Il6ra Δmyel mice exhibited no differences in insulin sensitivity or histological markers of AT inflammation. Notably, we found a reduction of ATMs expressing the mannose receptor 1 (CD206), as well as a decrease of the proliferation marker Ki67 in ATMs of Il6ra Δmyel mice. Importantly, organotypical AT culture and BMDM data of Il4ra -/- mice revealed that IL-6 mediates a shift towards the M2 phenotype independent from the IL-6/IL-4Rα axis. Discussion: Our results demonstrate IL-4Rα-independent anti-inflammatory effects of IL-6 on macrophages and the ability of IL-6 to maintain proliferation rates in obese AT.


Asunto(s)
Diabetes Mellitus Tipo 2 , Interleucina-6 , Ratones , Animales , Interleucina-6/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Tejido Adiposo/metabolismo , Macrófagos/metabolismo , Inflamación/metabolismo , Ratones Noqueados , Obesidad/metabolismo
6.
Microbiol Spectr ; 12(1): e0347523, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38018982

RESUMEN

IMPORTANCE: Sepsis is the consequence of a systemic bacterial infection that exacerbates the immune cell's activation via bacterial products, resulting in the augmented release of inflammatory mediators. A critical factor in the pathogenesis of sepsis is the primary component of the outer membrane of Gram-negative bacteria known as lipopolysaccharide (LPS), which is sensed by TLR4. For this reason, scientists have aimed to develop antagonists able to block TLR4 and, thereby the cytokine storm. We report here that a mixture of mu-class isoforms from the F. hepatica GST protein family administered intraperitoneally 1 h prior to a lethal LPS injection can modulate the dynamics and abundance of large peritoneal macrophages in the peritoneal cavity of septic mice while significantly suppressing the LPS-induced cytokine storm in a mouse model of septic shock. These results suggest that native F. hepatica glutathione S-transferase is a promising candidate for drug development against endotoxemia and other inflammatory diseases.


Asunto(s)
Fasciola hepatica , Sepsis , Animales , Ratones , Macrófagos Peritoneales/metabolismo , Lipopolisacáridos/metabolismo , Fasciola hepatica/metabolismo , Escherichia coli/metabolismo , Síndrome de Liberación de Citoquinas/metabolismo , Receptor Toll-Like 4/metabolismo , Macrófagos
7.
Front Immunol ; 14: 1293828, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38162675

RESUMEN

Alterations in macrophage (Mφ) polarization, function, and metabolic signature can foster development of chronic diseases, such as autoimmunity or fibrotic tissue remodeling. Thus, identification of novel therapeutic agents that modulate human Mφ biology is crucial for treatment of such conditions. Herein, we demonstrate that the soluble CD83 (sCD83) protein induces pro-resolving features in human monocyte-derived Mφ biology. We show that sCD83 strikingly increases the expression of inhibitory molecules including ILT-2 (immunoglobulin-like transcript 2), ILT-4, ILT-5, and CD163, whereas activation markers, such as MHC-II and MSR-1, were significantly downregulated. This goes along with a decreased capacity to stimulate alloreactive T cells in mixed lymphocyte reaction (MLR) assays. Bulk RNA sequencing and pathway analyses revealed that sCD83 downregulates pathways associated with pro-inflammatory, classically activated Mφ (CAM) differentiation including HIF-1A, IL-6, and cytokine storm, whereas pathways related to alternative Mφ activation and liver X receptor were significantly induced. By using the LXR pathway antagonist GSK2033, we show that transcription of specific genes (e.g., PPARG, ABCA1, ABCG1, CD36) induced by sCD83 is dependent on LXR activation. In summary, we herein reveal for the first time mechanistic insights into the modulation of human Mφ biology by sCD83, which is a further crucial preclinical study for the establishment of sCD83 as a new therapeutical agent to treat inflammatory conditions.


Asunto(s)
Antígeno CD83 , Macrófagos , Linfocitos T , Humanos , Diferenciación Celular , Fenotipo
8.
Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi ; 34(4): 438-444, 2022 Aug 19.
Artículo en Chino | MEDLINE | ID: mdl-36116939

RESUMEN

As a type of highly plastic innate immune cells, macrophages may be differentiated into M1 and M2 macrophages upon different stimuli, and M2 macrophages are involved in immune regulation, tissue remodeling and regeneration, and wound healing. Previous epidemiological studies have shown a significant negative correlation between the prevalence of helminth infections and the incidence of inflammatory diseases, such as allergy and autoimmune diseases. As a common type of intestinal helminths, hookworm infection may trigger high levels of type II host immune responses, with alternative activation of macrophages, which are effective to inhibit the development and progression of inflammatory diseases. This review summarizes the advances in alternative activation of macrophages in hookworm therapy for inflammatory diseases.


Asunto(s)
Helmintiasis , Infecciones por Uncinaria , Ancylostomatoidea , Animales , Diferenciación Celular , Macrófagos
9.
Front Immunol ; 13: 860262, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35444643

RESUMEN

Activation of tissue repair program in macrophages requires the integration of IL-4/IL-13 cytokines and tissue-specific signals. In the lung, surfactant protein A (SP-A) is a tissue factor that amplifies IL-4Rα-dependent alternative activation and proliferation of alveolar macrophages (AMs) through the myosin18A receptor. However, the mechanism by which SP-A and IL-4 synergistically increase activation and proliferation of AMs is unknown. Here we show that SP-A amplifies IL-4-mediated phosphorylation of STAT6 and Akt by binding to myosin18A. Blocking PI3K activity or the myosin18A receptor abrogates SP-A´s amplifying effects on IL-4 signaling. SP-A alone activates Akt, mTORC1, and PKCζ and inactivates GSK3α/ß by phosphorylation, but it cannot activate arginase-1 activity or AM proliferation on its own. The combined effects of IL-4 and SP-A on the mTORC1 and GSK3 branches of PI3K-Akt signaling contribute to increased AM proliferation and alternative activation, as revealed by pharmacological inhibition of Akt (inhibitor VIII) and mTORC1 (rapamycin and torin). On the other hand, the IL-4+SP-A-driven PKCζ signaling axis appears to intersect PI3K activation with STAT6 phosphorylation to achieve more efficient alternative activation of AMs. Consistent with IL-4+SP-A-driven activation of mTORC1 and mTORC2, both agonists synergistically increased mitochondrial respiration and glycolysis in AMs, which are necessary for production of energy and metabolic intermediates for proliferation and alternative activation. We conclude that SP-A signaling in AMs activates PI3K-dependent branched pathways that amplify IL-4 actions on cell proliferation and the acquisition of AM effector functions.


Asunto(s)
Activación de Macrófagos , Proteína A Asociada a Surfactante Pulmonar , Glucógeno Sintasa Quinasa 3/metabolismo , Interleucina-4/metabolismo , Macrófagos Alveolares/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Transducción de Señal
10.
Immunol Cell Biol ; 100(5): 323-337, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35238065

RESUMEN

Patients with hepatitis B virus-related acute-on-chronic liver failure (HBV-ACLF) are characterized by immune paralysis and susceptibility to infections. Macrophages are important mediators of immune responses can be subclassified into two main phenotypes: classically activated and alternatively activated. However, few studies have investigated changes to macrophage polarization in HBV-related liver diseases. Therefore, we investigated the functional status of monocyte-derived macrophages (MDMs) from patients with mild chronic hepatitis B (n = 226), HBV-related compensated cirrhosis (n = 36), HBV-related decompensated cirrhosis (n = 40), HBV-ACLF (n = 62) and healthy controls (n = 10), as well as Kupffer cells (KCs) from patients with HBV-ACLF (n = 3). We found that during the progression of HBV-related liver diseases, the percentage of CD163+ CD206+ macrophages increased, while the percentage of CD80+ human leukocyte antigen-DR+ macrophages decreased significantly. MDMs and KCs mainly exhibited high CD163+ CD206+ expression in patients with HBV-ACLF, which predicted poor clinical outcome and higher liver transplantation rate. Transcriptome sequencing analysis revealed that chloride intracellular channel-3 (CLIC3) was reduced in patients with HBV-ACLF, indicating a poor prognosis. To further study the effect of CLIC3 on macrophage polarization, human monocytic THP-1 cell-derived macrophages were used. We found that classical and alternative macrophage activation occurred through nuclear factor kappa B (NF-κB) and phosphoinositide 3-kinase/protein kinase B pathways, respectively. CLIC3 suppression inhibited NF-κB activation and promoted the alternative activation. In conclusion, macrophage polarization gradually changed from classically activated to alternatively activated as HBV-related liver diseases progressed. Both CLIC3 suppression and increased alternatively activated macrophage percentage were potential indicators of the poor prognosis of patients with HBV-ACLF.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada , Canales de Cloruro/metabolismo , Hepatitis B Crónica , Cloruros , Virus de la Hepatitis B , Hepatitis B Crónica/complicaciones , Humanos , Cirrosis Hepática , Activación de Macrófagos , Macrófagos , FN-kappa B , Fosfatidilinositol 3-Quinasas
11.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-942374

RESUMEN

As a type of highly plastic innate immune cells, macrophages may be differentiated into M1 and M2 macrophages upon different stimuli, and M2 macrophages are involved in immune regulation, tissue remodeling and regeneration, and wound healing. Previous epidemiological studies have shown a significant negative correlation between the prevalence of helminth infections and the incidence of inflammatory diseases, such as allergy and autoimmune diseases. As a common type of intestinal helminths, hookworm infection may trigger high levels of type II host immune responses, with alternative activation of macrophages, which are effective to inhibit the development and progression of inflammatory diseases. This review summarizes the advances in alternative activation of macrophages in hookworm therapy for inflammatory diseases.

12.
Front Immunol ; 12: 771453, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34880868

RESUMEN

Interleukin (IL)-4 is a cytokine that affects both adaptive and innate immune responses. In the central nervous system, microglia express IL-4 receptors and it has been described that IL-4-exposed microglia acquire anti-inflammatory properties. We here demonstrate that IL-4 exposure induces changes in the cell surface protein expression profile of primary rhesus macaque microglia and enhances their potential to induce proliferation of T cells with a regulatory signature. Moreover, we show that Toll like receptor (TLR)-induced cytokine production is broadly impaired in IL-4-exposed microglia at the transcriptional level. IL-4 type 2 receptor-mediated signaling is shown to be crucial for the inhibition of microglial innate immune responses. TLR-induced nuclear translocalization of NF-κB appeared intact, and we found no evidence for epigenetic modulation of target genes. By contrast, nuclear extracts from IL-4-exposed microglia contained significantly less NF-κB capable of binding to its DNA consensus site. Further identification of the molecular mechanisms that underlie the inhibition of TLR-induced responses in IL-4-exposed microglia may aid the design of strategies that aim to modulate innate immune responses in the brain, for example in gliomas.


Asunto(s)
Citocinas/inmunología , Microglía/inmunología , FN-kappa B/inmunología , Receptores Toll-Like/inmunología , Animales , Proliferación Celular , Células Cultivadas , Femenino , Histona Desacetilasas/genética , Lipopolisacáridos/farmacología , Macaca mulatta , Masculino , Linfocitos T/inmunología , Transcripción Genética
13.
mBio ; 12(4): e0165721, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34253061

RESUMEN

Extracellular vesicles (EVs) have recently garnered attention for their participation in host-microbe interactions in pneumococcal infections. However, the effect of EVs on the host immune system remain poorly understood. Our studies focus on EVs produced by Streptococcus pneumoniae (pEVs), and reveal that pEVs are internalized by macrophages, T cells, and epithelial cells. In vitro, pEVs induce NF-κB activation in a dosage-dependent manner and polarize human macrophages to an alternative (M2) phenotype. In addition, pEV pretreatment conditions macrophages to increase bacteria uptake and such macrophages may act as a reservoir for pneumococcal cells by increasing survival of the phagocytosed bacteria. When administered systemically in mice, pEVs induce cytokine release; when immobilized locally, they recruit lymphocytes and macrophages. Taken together, pEVs emerge as critical contributors to inflammatory responses and tissue damage in mammalian hosts. IMPORTANCE Over the last decade, pathogen-derived extracellular vesicles (EVs) have emerged as important players in several human diseases. Therefore, a thorough understanding of EV-mediated mechanisms could provide novel insights into vaccine/therapeutic development. A critical question in the field is: do pathogen-derived EVs help the pathogen evade the harsh environment in the host or do they help the host to mount a robust immune response against the pathogen? This study is a step towards answering this critical question for the Gram-positive pathogen, Streptococcus pneumoniae. Our study shows that while S. pneumoniae EVs (pEVs) induce inflammatory response both in vitro and in vivo, they may also condition the host macrophages to serve as a reservoir for the bacteria.


Asunto(s)
Vesículas Extracelulares/inmunología , Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Streptococcus pneumoniae/inmunología , Animales , Femenino , Macrófagos/clasificación , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitosis , Fenotipo , Infecciones Neumocócicas/inmunología , Transducción de Señal/inmunología
14.
Front Immunol ; 12: 618501, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149684

RESUMEN

Macrophage polarization is mainly steered by metabolic reprogramming in the tissue microenvironment, thus leading to distinct outcomes of various diseases. However, the role of lipid metabolism in the regulation of macrophage alternative activation is incompletely understood. Using human THP-1 and mouse bone marrow derived macrophage polarization models, we revealed a pivotal role for arachidonic acid metabolism in determining the phenotype of M2 macrophages. We demonstrated that macrophage M2 polarization was inhibited by arachidonic acid, but inversely facilitated by its derived metabolite prostaglandin E2 (PGE2). Furthermore, PPARγ bridges these two seemingly unrelated processes via modulating oxidative phosphorylation (OXPHOS). Through inhibiting PPARγ, PGE2 enhanced OXPHOS, resulting in the alternative activation of macrophages, which was counterweighted by the activation of PPARγ. This connection between PGE2 biosynthesis and macrophage M2 polarization also existed in human and mouse esophageal squamous cell carcinoma. Our results highlight the critical role of arachidonic acid and metabolic PGE2 as immune regulators in modulating tissue homeostasis and pathological process.


Asunto(s)
Ácido Araquidónico/metabolismo , Carcinoma de Células Escamosas/inmunología , Dinoprostona/metabolismo , Neoplasias Esofágicas/inmunología , Inflamación/metabolismo , Macrófagos/fisiología , PPAR gamma/metabolismo , Animales , Diferenciación Celular , Homeostasis , Humanos , Metabolismo de los Lípidos , Activación de Macrófagos , Ratones , Fosforilación Oxidativa , Transducción de Señal , Células THP-1 , Células Th2/inmunología
15.
FEBS J ; 288(11): 3530-3546, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33314622

RESUMEN

The alternative activation of macrophages in the lungs has been considered as a major factor promoting pulmonary fibrogenesis; however, the mechanisms underlying this phenomenon are still elusive. In this study, we investigated the interaction between macrophages and fibrosis-associated alveolar epithelial cells using a bleomycin-induced mouse pulmonary fibrosis model and a coculture system. We demonstrated that fibrosis-promoting macrophages are spatially proximate to alveolar type II (ATII) cells, permissive for paracrine-induced macrophage polarization. Importantly, we revealed that fibrosis-associated ATII cells secrete Sonic hedgehog (Shh), a hedgehog pathway ligand, and that ATII cell-derived Shh promotes the development of pulmonary fibrosis by osteopontin (OPN)-mediated macrophage alternative activation. Mechanistically, Shh promotes the secretion of OPN in macrophages via Shh/Gli signaling cascade. The secreted OPN acts on the surrounding macrophages in an autocrine or paracrine manner and induces macrophage alternative activation through activating the JAK2/STAT3 signaling pathway. Tissue samples from idiopathic pulmonary fibrosis patients confirmed the increased expression of Shh and OPN in ATII cells and macrophages, respectively. Together, our study illustrated an alveolar epithelium-dependent mechanism for macrophage M2 polarization and pulmonary fibrogenesis and suggested that targeting Shh may offer a selective and efficient therapeutic strategy for the development and progression of pulmonary fibrosis.


Asunto(s)
Proteínas Hedgehog/genética , Janus Quinasa 2/genética , Osteopontina/genética , Fibrosis Pulmonar/genética , Factor de Transcripción STAT3/genética , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Modelos Animales de Enfermedad , Humanos , Activación de Macrófagos/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Fibrosis Pulmonar/patología , Transducción de Señal/genética
16.
Innate Immun ; 27(2): 133-142, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33241977

RESUMEN

The capacity for macrophages to polarize into distinct functional activation states (e.g., M1, M2) is critical to tune an inflammatory response to the relevant infection or injury. Alternative or M2 polarization of macrophages is most often achieved in vitro in response to IL-4/IL-13 and results in the transcriptional up-regulation of a constellation of characteristic M2 marker genes. In vivo, additional signals from the inflammatory milieu can further increase or decrease M2 marker expression. Particularly, activation of cAMP-generating G protein-coupled receptors is reported to increase M2 markers, but whether this is strictly dependent upon cAMP production is unclear. We report herein that increased cAMP alone can increase IL-4-dependent M2 marker expression through a PKA/C/EBPß/CREB dependent pathway in murine macrophages.


Asunto(s)
Biomarcadores/metabolismo , AMP Cíclico/metabolismo , Macrófagos/metabolismo , Animales , Diferenciación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Interleucina-4/metabolismo , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células RAW 264.7 , Transducción de Señal , Esteroide Isomerasas/metabolismo , Células Th2/inmunología
17.
Front Immunol ; 11: 2015, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072069

RESUMEN

Trichinella infection can induce macrophages into the alternatively activated phenotype, which is primarily associated with the development of a polarized Th2 immune response. In the present study, we examined the immunomodulatory effect of T. spiralis thioredoxin peroxidase-2 (TsTPX2), a protein derived from T. spiralis ES products, in the regulation of Th2 response through direct activation of macrophages. The location of TsTPX2 was detected by immunohistochemistry and immunofluorescence analyses. The immune response in vivo induced by rTsTPX2 was characterized by analyzing the Th2 cytokines and Th1 cytokines in the peripheral blood. The rTsTPX2-activated macrophages (MrTsTPX2) were tested for polarization, their ability to evoke naïve CD4+ T cells, and resistance to the larval infection after adoptive transfer in BALB/c mice. The immunolocalization analysis showed TsTPX2 in cuticles and stichosome of T. spiralis ML. The immunostaining was detected in cuticles and stichosome of T. spiralis Ad3 and ML, as well as in tissue-dwellings around ML after the intestines and muscle tissues of infected mice were incubated with anti-rTsTPX2 antibody. Immunization of BALB/c mice with rTsTPX2 could induce a Th1-suppressing mixed immune response given the increased levels of Th2 cytokines (IL-4 and IL-10) production along with the decreased levels of Th1 cytokines (IFN-γ, IL-12, and TNF-α). In vitro studies showed that rTsTPX2 could directly drive RAW264.7 and peritoneal macrophages to the M2 phenotype. Moreover, MrTsTPX2 could promote CD4+ T cells polarized into Th2 type in vitro. Adoptive transfer of MrTsTPX2 into mice suppressed Th1 responses by enhancing Th2 responses and exhibited a 44.7% reduction in adult worm burden following challenge with T. spiralis infective larval, suggesting that the TsTPX2 is a potential vaccine candidate against trichinosis. Our study showed that TsTPX2 would be at least one of the molecules to switch macrophages into the M2 phenotype during T. spiralis infection, which provides a new therapeutic approach to various inflammatory disorders like allergies or autoimmune diseases.


Asunto(s)
Proteínas del Helminto/metabolismo , Macrófagos/inmunología , Peroxirredoxinas/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Trichinella spiralis/fisiología , Triquinelosis/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Resistencia a la Enfermedad , Femenino , Proteínas del Helminto/genética , Inmunidad Celular , Inmunomodulación , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Peroxirredoxinas/genética
18.
Brain Res Bull ; 161: 21-32, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32389801

RESUMEN

Traumatic brain injury (TBI) causes substantial mortality and long-term disability worldwide. TGFß1 is a unique molecular and functional signature in microglia, but the role of TGFß1 in TBI is not clear. The purpose of this study was to investigate the role of TGFß1 in TBI. The weight dropping device was used to establish TBI model of rats. Hematoxylin eosin staining and Bielschowsky silver staining were used to assess tissue loss. Beam walking and muscle strength tests were used to assess neurological deficits. Immunohistochemical staining was used to assess axonal injures. Western blotting was used to detect expression of related proteins. RT-PCR was used to detect expression of cytokines. Immunofluorescence staining was used to assess the microglia/macrophages activation. We observed obvious axonal injury and microglia/macrophages activation in the peri-lesion cortex. The expression of inflammatory cytokines was markedly high after TBI. The expression of TGFß1 and TGFßRI were significantly reduced after TBI. TGFß1 promoted the functional recovery and alleviated axonal injury 1 day after TBI. TGFß1 promoted microglia/macrophages polarizing to alternative activation and alleviated neuroinflammation. These effects of TGFß1 could be inhibited by LY2109761, the inhibitor of TGFRI/II. These results suggested that TGFß1 played a protective role in axonal injury and could be a potential therapeutic target in early stages following TBI.


Asunto(s)
Axones/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Animales , Axones/efectos de los fármacos , Axones/patología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/prevención & control , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Microglía/efectos de los fármacos , Microglía/patología , Pirazoles/farmacología , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley
19.
Methods Mol Biol ; 2137: 117-131, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32399925

RESUMEN

The liver fluke, Fasciola hepatica, infects a wide range of mammals including humans and leads to chronic disease. Like other helminths, F. hepatica migrates and survives in the host tissues after penetrating the intestinal wall to enter the peritoneal cavity, and then migrates through the liver before finally inhabiting the bile ducts. To avoid the antihelminthic immune response during migration, F. hepatica releases excretory-secretory products (FhESP) that exert various immunomodulatory effects, such as alternative macrophage activation or programmed cell death induction. Here, we describe the currently available techniques for studying macrophage activation and apoptotic cell death triggered by purified FhESP originating from the adult F. hepatica fluke.


Asunto(s)
Antígenos Helmínticos/inmunología , Fasciola hepatica/inmunología , Inmunomodulación/inmunología , Macrófagos/inmunología , Animales , Apoptosis/inmunología , Femenino , Inmunidad/inmunología , Macrófagos/parasitología , Ratones , Ratones Endogámicos BALB C
20.
Immunol Rev ; 295(1): 54-67, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32242952

RESUMEN

We have only recently started to appreciate the extent to which immune cell activation involves significant changes in cellular metabolism. We are now beginning to understand how commitment to specific metabolic pathways influences aspects of cellular biology that are the more usual focus of immunological studies, such as activation-induced changes in gene transcription, post-transcriptional regulation of transcription, post-translational modifications of proteins, cytokine secretion, etc. Here, we focus on metabolic reprogramming in mononuclear phagocytes downstream of stimulation with inflammatory signals (such as LPS and IFNγ) vs alternative activation signals (IL-4), with an emphasis on work on dendritic cells and macrophages from our laboratory, and related studies from others. We cover aspects of glycolysis and its branching pathways (glycogen synthesis, pentose phosphate, serine synthesis, hexose synthesis, and glycerol 3 phosphate shuttle), the tricarboxylic acid pathway, fatty acid synthesis and oxidation, and mitochondrial biology. Although our understanding of the metabolism of mononuclear phagocytes has progressed significantly over the last 10 years, major challenges remain, including understanding the effects of tissue residence on metabolic programming related to cellular activation, and the translatability of findings from mouse to human biology.


Asunto(s)
Sistema Mononuclear Fagocítico/inmunología , Sistema Mononuclear Fagocítico/metabolismo , Fagocitos/inmunología , Fagocitos/metabolismo , Animales , Metabolismo Energético , Humanos , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Sistema Mononuclear Fagocítico/citología , Fagocitos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA