Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Semin Immunol ; 54: 101523, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-34776300

RESUMEN

Granulocyte macrophage-colony stimulating factor (GM-CSF) was originally identified as a growth factor for its ability to promote the proliferation and differentiation in vitro of bone marrow progenitor cells into granulocytes and macrophages. Many preclinical studies, using GM-CSF deletion or depletion approaches, have demonstrated that GM-CSF has a wide range of biological functions, including the mediation of inflammation and pain, indicating that it can be a potential target in many inflammatory and autoimmune conditions. This review provides a brief overview of GM-CSF biology and signaling, and summarizes the findings from preclinical models of a range of inflammatory and autoimmune disorders and the latest clinical trials targeting GM-CSF or its receptor in these disorders.


Asunto(s)
Enfermedades Autoinmunes , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Enfermedades Autoinmunes/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Granulocitos/metabolismo , Humanos , Inflamación , Macrófagos
2.
Artículo en Inglés | MEDLINE | ID: mdl-38844159

RESUMEN

OBJECTIVE: We have previously reported that the interleukin-23 p19 subunit (IL-23p19) is required for experimental inflammatory arthritic pain-like behavior and disease. Even though inflammation is often a characteristic feature of osteoarthritis (OA), IL-23 is not usually considered as a therapeutic target in OA. We began to explore the role of IL-23p19 in OA pain and disease utilizing mouse models of OA and patient samples. DESIGN: The role of IL-23p19 in two mouse models of OA, namely collagenase-induced OA and monosodium iodoacetate-induced OA, was investigated using gene-deficient male mice. Pain-like behavior and arthritis were assessed by relative static weight distribution and histology, respectively. In knee synovial tissues from a small cohort of human OA patients, a correlation analysis was performed between IL-23A gene expression and Oxford knee score (OKS), a validated Patient Reported Outcome Measure. RESULTS: We present evidence that i) IL-23p19 is required for the development of pain-like behavior and optimal disease, including cartilage damage and osteophyte formation, in two experimental OA models and ii) IL-23A gene expression in OA knee synovial tissues correlates with a lower OKS (r = -0.742, p = 0.0057). CONCLUSIONS: The findings support the possible targeting of IL-23 as a treatment for OA pain and disease progression.

3.
Immunol Cell Biol ; 101(7): 600-609, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36975092

RESUMEN

Chemokine (C-C) ligand 17 (CCL17) was first identified as thymus- and activation-regulated chemokine when it was found to be constitutively expressed in the thymus and identified as a T-cell chemokine. This chemoattractant molecule has subsequently been found at elevated levels in a range of autoimmune and inflammatory diseases, as well as in cancer. CCL17 is a C-C chemokine receptor type 4 (CCR4) ligand, with chemokine (C-C) ligand 22 being the other major ligand and, as CCR4 is highly expressed on helper T cells, CCL17 can play a role in T-cell-driven diseases, usually considered to be via its chemotactic activity on T helper 2 cells; however, given that CCR4 is also expressed by other cell types and there is elevated expression of CCL17 in many diseases, a broader CCL17 biology is suggested. In this review, we summarize the biology of CCL17, its regulation and its potential contribution to the pathogenesis of various preclinical models. Reference is made, for example, to recent literature indicating a role for CCL17 in the control of pain as part of a granulocyte macrophage-colony-stimulating factor/CCL17 pathway in lymphocyte-independent models and thus not as a T-cell chemokine. The review also discusses the potential for CCL17 to be a biomarker and a therapeutic target in human disorders.


Asunto(s)
Autoinmunidad , Receptores de Quimiocina , Humanos , Ligandos , Receptores de Quimiocina/metabolismo , Quimiocina CCL17/metabolismo , Quimiocinas , Inflamación
4.
Osteoarthritis Cartilage ; 31(10): 1327-1341, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37225052

RESUMEN

OBJECTIVES: We have previously identified a granulocyte macrophage-colony stimulating factor (GM-CSF)/C-C motif ligand 17 (CCL17) pathway in monocytes/macrophages, in which GM-CSF regulates the formation of CCL17, and it is important for an experimental osteoarthritis (OA) model. We explore here additional OA models, including in the presence of obesity, such as a requirement for this pathway. DESIGN: The roles of GM-CSF, CCL17, CCR4, and CCL22 in various experimental OA models, including those incorporating obesity (eight-week high-fat diet), were investigated using gene-deficient male mice. Pain-like behavior and arthritis were assessed by relative static weight distribution and histology, respectively. Cell populations (flow cytometry) and cytokine messenger RNA (mRNA) expression (qPCR) in knee infrapatellar fat pad were analyzed. Human OA sera were collected for circulating CCL17 levels (ELISA) and OA knee synovial tissue for gene expression (qPCR). RESULTS: We present evidence that: i) GM-CSF, CCL17, and CCR4, but not CCL22, are required for the development of pain-like behavior and optimal disease in three experimental OA models, as well as for exacerbated OA development due to obesity, ii) obesity alone leads to spontaneous knee joint damage in a GM-CSF- and CCL17-dependent manner, and iii) in knee OA patients, early indications are that BMI correlates with a lower Oxford Knee Score (r = -0.458 and p = 0.0096), with elevated circulating CCL17 levels (r = 0.2108 and p = 0.0153) and with elevated GM-CSF and CCL17 gene expression in OA synovial tissue. CONCLUSIONS: The above findings indicate that GM-CSF, CCL17, and CCR4 are involved in obesity-associated OA development, broadening their potential as targets for possible treatments for OA.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Osteoartritis de la Rodilla , Humanos , Masculino , Animales , Ratones , Citocinas , Dolor , Osteoartritis de la Rodilla/etiología , Membrana Sinovial/metabolismo , Quimiocina CCL17
5.
Int J Mol Sci ; 24(4)2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36834778

RESUMEN

Glioblastoma cells adapt to changes in glucose availability through metabolic plasticity allowing for cell survival and continued progression in low-glucose concentrations. However, the regulatory cytokine networks that govern the ability to survive in glucose-starved conditions are not fully defined. In the present study, we define a critical role for the IL-11/IL-11Rα signalling axis in glioblastoma survival, proliferation and invasion when cells are starved of glucose. We identified enhanced IL-11/IL-11Rα expression correlated with reduced overall survival in glioblastoma patients. Glioblastoma cell lines over-expressing IL-11Rα displayed greater survival, proliferation, migration and invasion in glucose-free conditions compared to their low-IL-11Rα-expressing counterparts, while knockdown of IL-11Rα reversed these pro-tumorigenic characteristics. In addition, these IL-11Rα-over-expressing cells displayed enhanced glutamine oxidation and glutamate production compared to their low-IL-11Rα-expressing counterparts, while knockdown of IL-11Rα or the pharmacological inhibition of several members of the glutaminolysis pathway resulted in reduced survival (enhanced apoptosis) and reduced migration and invasion. Furthermore, IL-11Rα expression in glioblastoma patient samples correlated with enhanced gene expression of the glutaminolysis pathway genes GLUD1, GSS and c-Myc. Overall, our study identified that the IL-11/IL-11Rα pathway promotes glioblastoma cell survival and enhances cell migration and invasion in environments of glucose starvation via glutaminolysis.


Asunto(s)
Glioblastoma , Humanos , Línea Celular , Línea Celular Tumoral , Glioblastoma/metabolismo , Glucosa/metabolismo , Interleucina-11/metabolismo , Receptores de Interleucina-11
6.
J Immunol ; 205(1): 213-222, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32461237

RESUMEN

It has been reported that a GM-CSF→CCL17 pathway, originally identified in vitro in macrophage lineage populations, is implicated in the control of inflammatory pain, as well as arthritic pain and disease. We explore, in this study and in various inflammation models, the cellular CCL17 expression and its GM-CSF dependence as well as the function of CCL17 in inflammation and pain. This study used models allowing the convenient cell isolation from Ccl17E/+ reporter mice; it also exploited both CCL17-dependent and unique CCL17-driven inflammatory pain and arthritis models, the latter permitting a radiation chimera approach to help identify the CCL17 responding cell type(s) and the mediators downstream of CCL17 in the control of inflammation and pain. We present evidence that 1) in the particular inflammation models studied, CCL17 expression is predominantly in macrophage lineage populations and is GM-CSF dependent, 2) for its action in arthritic pain and disease development, CCL17 acts on CCR4+ non-bone marrow-derived cells, and 3) for inflammatory pain development in which a GM-CSF→CCL17 pathway appears critical, nerve growth factor, CGRP, and substance P all appear to be required.


Asunto(s)
Artritis Experimental/inmunología , Quimiocina CCL17/metabolismo , Dolor/inmunología , Peritonitis/inmunología , Neumonía/inmunología , Animales , Artritis Experimental/complicaciones , Artritis Experimental/patología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Quimiocina CCL17/genética , Genes Reporteros/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Ratones , Ratones Transgénicos , Factor de Crecimiento Nervioso/metabolismo , Dolor/diagnóstico , Dolor/patología , Dimensión del Dolor , Peritonitis/complicaciones , Peritonitis/patología , Neumonía/complicaciones , Neumonía/patología , Transducción de Señal/inmunología , Sustancia P/metabolismo
7.
J Immunol ; 202(10): 3033-3040, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988114

RESUMEN

Studies have demonstrated the importance of a GM-CSF→IFN regulatory factor 4 (IRF4)→CCL17 pathway, first identified in monocytes/macrophages, for arthritic pain and disease development. In this study, we further investigated the involvement of this new pathway in shaping the inflammatory response using the zymosan-induced peritonitis (ZIP) model. ZIP (8 mg of zymosan, i.p., day 0) was induced in C57BL/6 wild-type (WT), GM-CSF-/- , Irf4-/- , and Ccl17E/E mice. In comparison with WT mice, GM-CSF-/- and Irf4-/- mice had a reduced ZIP response, as judged by a reduced number of neutrophils and macrophages in the peritoneal cavity. Moreover, the phenotype of the ZIP macrophages was altered by a lack of GM-CSF or IRF4 (increased IL-10 secretion and Arg1 mRNA expression), with IRF4 levels being lower in GM-CSF-/- ZIP macrophages than in the WT cells. In addition, GM-CSF ̶IRF4 signaling upregulated MHC class II expression in ZIP macrophages and bone marrow-derived macrophages. Although Ccl17 mRNA expression was reduced in ZIP macrophages in the absence of either GM-CSF or IRF4, thus supporting the presence of the new pathway in inflammatory macrophages, CCL17 did not modulate the inflammatory response, both in terms of number of myeloid cells or the macrophage phenotype. Thus, during an inflammatory response, both macrophage numbers and their phenotype can depend on GM-CSF- and IRF4-dependent signaling independently of CCL17.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factores Reguladores del Interferón/inmunología , Macrófagos/inmunología , Transducción de Señal/inmunología , Animales , Quimiocina CCL17/genética , Quimiocina CCL17/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Factores Reguladores del Interferón/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Transducción de Señal/genética , Regulación hacia Arriba/inmunología
8.
J Immunol ; 201(7): 2042-2053, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30120124

RESUMEN

Pain is one of the most debilitating symptoms in many diseases for which there is inadequate management and understanding. CSF-1, also known as M-CSF, acts via its receptor (CSF-1R, c-Fms) to regulate the development of the monocyte/macrophage lineage and to act locally in tissues to control macrophage numbers and function. It has been implicated in the control of neuropathic pain via a central action on microglia. We report in this study that systemic administration of a neutralizing anti-CSF-1R or CSF-1 mAb inhibits the development of inflammatory pain induced by zymosan, GM-CSF, and TNF in mice. This approach also prevented but did not ameliorate the development of arthritic pain and optimal disease driven by the three stimuli in mice, suggesting that CSF-1 may only be relevant when the driving inflammatory insults in tissues are acute and/or periodic. Systemic CSF-1 administration rapidly induced pain and enhanced the arthritis in an inflamed mouse joint, albeit via a different pathway(s) from that used by systemic GM-CSF and TNF. It is concluded that CSF-1 can function peripherally during the generation of inflammatory pain and hence may be a target for such pain and associated disease, including when the clinically important cytokines, TNF and GM-CSF, are involved. Our findings have ramifications for the selection and design of anti-CSF-1R/CSF-1 trials.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Inflamación/inmunología , Articulaciones/inmunología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/fisiología , Monocitos/fisiología , Animales , Anticuerpos Neutralizantes/administración & dosificación , Diferenciación Celular , Linaje de la Célula , Humanos , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Dolor , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal
10.
J Biol Chem ; 293(29): 11415-11423, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-29871928

RESUMEN

Interleukin 4 (IL4) is generally viewed as a Th2 cytokine capable of polarizing macrophages into an anti-inflammatory phenotype, whereas granulocyte macrophage-colony-stimulating factor (GM-CSF) is often viewed as a proinflammatory cytokine with part of this function due to its action on monocytes/macrophages. Paradoxically, these two cytokines act additively to enhance the in vitro differentiation of dendritic cells from precursors such as monocytes. One up-regulated marker of an IL4-polarized M2 macrophage is the chemokine (C-C motif) ligand 17 (CCL17), which we have recently reported to be induced by GM-CSF in monocytes/macrophages in an interferon regulatory factor 4 (IRF4)-dependent manner. In this study, we report that IL4 also induces CCL17 production by acting through IRF4 in human monocytes and murine macrophages. Furthermore, evidence is presented that IL4 up-regulates IRF4 expression at the epigenetic level by enhancing the expression and activity of jumonji domain-containing protein 3 (JMJD3) demethylase. Intriguingly, silencing the signal transducer and activator of transcription 6 (STAT6) gene led to a decrease in not only CCL17 formation, but also in that of its upstream regulators, JMJD3 and IRF4. Moreover, IL4 treatment of human monocytes resulted in an increased association of STAT6 to the promoter regions of the CCL17, IRF4, and JMJD3 genes. Thus, despite their vastly different functions, IL4 and GM-CSF appear to share elements of a common signaling pathway in regulating CCL17 production in human monocytes and murine macrophages.


Asunto(s)
Quimiocina CCL17/genética , Epigénesis Genética , Interleucina-4/genética , Macrófagos/metabolismo , Monocitos/metabolismo , Activación Transcripcional , Animales , Células Cultivadas , Humanos , Factores Reguladores del Interferón/genética , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba
11.
J Immunol ; 198(9): 3565-3575, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28320832

RESUMEN

G-CSF or CSF-3, originally defined as a regulator of granulocyte lineage development via its cell surface receptor (G-CSFR), can play a role in inflammation, and hence in many pathologies, due to its effects on mature lineage populations. Given this, and because pain is an extremely important arthritis symptom, the efficacy of an anti-G-CSFR mAb for arthritic pain and disease was compared with that of a neutrophil-depleting mAb, anti-Ly6G, in both adaptive and innate immune-mediated murine models. Pain and disease were ameliorated in Ag-induced arthritis, zymosan-induced arthritis, and methylated BSA/IL-1 arthritis by both prophylactic and therapeutic anti-G-CSFR mAb treatment, whereas only prophylactic anti-Ly6G mAb treatment was effective. Efficacy for pain and disease correlated with reduced joint neutrophil numbers and, importantly, benefits were noted without necessarily the concomitant reduction in circulating neutrophils. Anti-G-CSFR mAb also suppressed zymosan-induced inflammatory pain. A new G-CSF-driven (methylated BSA/G-CSF) arthritis model was established enabling us to demonstrate that pain was blocked by a cyclooxygenase-2 inhibitor, suggesting an indirect effect on neurons. Correspondingly, dorsal root ganglion neurons cultured in G-CSF failed to respond to G-CSF in vitro, and Csf3r gene expression could not be detected in dorsal root ganglion neurons by single-cell RT-PCR. These data suggest that G-CSFR/G-CSF targeting may be a safe therapeutic strategy for arthritis and other inflammatory conditions, particularly those in which pain is important, as well as for inflammatory pain per se.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Artritis Experimental/terapia , Artritis Reumatoide/terapia , Inmunoterapia/métodos , Neuronas/efectos de los fármacos , Neutrófilos/inmunología , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Animales , Antígenos Ly/inmunología , Artritis Experimental/inducido químicamente , Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Procedimientos de Reducción del Leucocitos , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Manejo del Dolor , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Receptores de Factor Estimulante de Colonias de Granulocito/inmunología
12.
J Immunol ; 196(5): 2230-8, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26819203

RESUMEN

IFN regulatory factors (IRFs) help to shape the immune response to pathogens by imparting signaling specificity to individual TLRs. We recently demonstrated that IRF6 provides specificity to TLR2 signaling in oral epithelial cells. TLR2 plays an important role in eliciting inflammation to Porphyromonas gingivalis, a keystone pathogen in periodontitis. Therefore, we investigated a role for IRF6 in mediating the inflammatory cytokine response of oral epithelial cells to P. gingivalis. IRF6 expression was strongly upregulated when human oral epithelial cells were challenged with P. gingivalis. Moreover, gene silencing and gene promoter experiments indicated that IRF6 acts downstream of IL-1R-associated kinase 1 to stimulate the expression of the IL-1 family cytokine IL-36γ in response to P. gingivalis. IRF6 and IL-1R-associated kinase 1 also regulated the stimulation of IL-36γ expression by a TLR2 agonist. IL-36γ was shown to elicit inflammatory responses by human monocyte-derived dendritic cells and macrophages, including the expression of the neutrophil chemokines IL-8 and CXCL1, as well as the Th17 chemokine CCL20. IL-36γ similarly stimulated their expression by human oral epithelial cells. Significantly, the Th17 cytokine IL-17 not only stimulated the expression of important regulators of neutrophil recruitment and survival by oral epithelial cells, but IL-17 also stimulated them to express IL-36γ. Thus, our findings suggest that IRF6 is likely to promote inflammation to P. gingivalis through its regulation of IL-36γ.


Asunto(s)
Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Interleucina-1/genética , Mucosa Bucal/metabolismo , Mucosa Bucal/virología , Porphyromonas gingivalis/inmunología , Infecciones por Bacteroidaceae/genética , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Epiteliales , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-17/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Modelos Biológicos , Mucosa Bucal/inmunología , Receptor Toll-Like 2/metabolismo , Regulación hacia Arriba
13.
J Biol Chem ; 290(26): 16031-42, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25979345

RESUMEN

Urokinase plasminogen activator (uPA) converts plasminogen to plasmin, resulting in a proteolytic cascade that has been implicated in tissue destruction during inflammation. Periodontitis is a highly prevalent chronic inflammatory disease characterized by destruction of the tissue and bone that support the teeth. We demonstrate that stimulation of macrophages with the arginine- and lysine-specific cysteine protease complex (RgpA-Kgp complex), produced by the keystone pathogen Porphyromonas gingivalis, dramatically increased their ability to degrade matrix in a uPA-dependent manner. We show that the RgpA-Kgp complex cleaves the inactive zymogens, pro-uPA (at consensus sites Lys(158)-Ile(159) and Lys(135)-Lys(136)) and plasminogen, yielding active uPA and plasmin, respectively. These findings are consistent with activation of the uPA proteolytic cascade by P. gingivalis being required for the pathogen to induce alveolar bone loss in a model of periodontitis and reveal a new host-pathogen interaction in which P. gingivalis activates a critical host proteolytic pathway to promote tissue destruction and pathogen virulence.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Macrófagos/enzimología , Periodontitis/enzimología , Porphyromonas gingivalis/enzimología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Adhesinas Bacterianas/genética , Animales , Células Cultivadas , Cisteína Endopeptidasas/genética , Activación Enzimática , Femenino , Cisteína-Endopeptidasas Gingipaínas , Interacciones Huésped-Patógeno , Humanos , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Periodontitis/genética , Periodontitis/microbiología , Porphyromonas gingivalis/genética , Unión Proteica , Activador de Plasminógeno de Tipo Uroquinasa/genética
14.
Trends Immunol ; 34(2): 81-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23000011

RESUMEN

The colony stimulating factors (CSFs), granulocyte macrophage-CSF (GM-CSF), macrophage-CSF (M-CSF or CSF-1) and granulocyte-CSF (G-CSF) were first identified as in vitro hematopoietic growth factors. They have since been shown to regulate myeloid cell numbers and function at steady state and during inflammation. Preclinical data suggest that targeting CSFs might be beneficial in autoimmune and inflammatory disease, and manipulation of CSF biology is now being tested in clinical trials. Here, we examine recent insights into CSF function, at steady state and during pathology, as provided by CSF or CSF receptor neutralization/deletion studies or from CSF administration. We discuss controversies regarding the role of CSFs in controlling specific myeloid cell populations and highlight how the newly identified M-CSF receptor ligand, interleukin (IL)-34, is necessitating a reassessment of the field.


Asunto(s)
Enfermedades Autoinmunes , Diferenciación Celular , Factores Estimulantes de Colonias/metabolismo , Inflamación , Células Mieloides/citología , Células Mieloides/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/terapia , Ensayos Clínicos como Asunto , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Ratones , Células Mieloides/fisiología
15.
J Immunol ; 192(8): 3540-7, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24616477

RESUMEN

Urokinase plasminogen activator (uPA) and its receptor (uPAR) coordinate a plasmin-mediated proteolytic cascade that has been implicated in cell adhesion, cell motility, and matrix breakdown, for example, during inflammation. As part of their function during inflammatory responses, macrophages move through tissues and encounter both two-dimensional (2D) surfaces and more complex three-dimensional (3D) interstitial matrices. Based on approaches employing uPA gene-deficient macrophages, plasminogen supplementation, and neutralization with specific protease inhibitors, it is reported in this study that uPA activity is a central component of the invasion of macrophages through a 3D Matrigel barrier; it also has a nonredundant role in macrophage-mediated matrix degradation. For murine macrophages, matrix metalloproteinase-9 activity was found to be required for these uPA-mediated effects. Evidence for a unique role for uPA in the inverse relationship between macrophage adhesion and 2D migration was also noted: macrophage adhesion to vitronectin was enhanced by uPA and blocked by plasminogen activator inhibitor-1, the latter approach also able to enhance in turn the 2D migration on this matrix protein. It is therefore proposed that uPA can have a key role in the inflammatory response at several levels as a central regulator of macrophage 3D invasion, matrix remodeling, and adhesion.


Asunto(s)
Movimiento Celular , Matriz Extracelular/metabolismo , Macrófagos/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Adhesión Celular/genética , Movimiento Celular/genética , Activación Enzimática , Femenino , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Proteolisis , Activador de Plasminógeno de Tipo Uroquinasa/genética
16.
J Immunol ; 188(11): 5752-65, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22547697

RESUMEN

GM-CSF and M-CSF (CSF-1) induce different phenotypic changes in macrophage lineage populations. The nature, extent, and generality of these differences were assessed by comparing the responses to these CSFs, either alone or in combination, in various human and murine macrophage lineage populations. The differences between the respective global gene expression profiles of macrophages, derived from human monocytes by GM-CSF or M-CSF, were compared with the differences between the respective profiles for macrophages, derived from murine bone marrow cells by each CSF. Only 17% of genes regulated differently by these CSFs were common across the species. Whether a particular change in relative gene expression is by direct action of a CSF can be confounded by endogenous mediators, such as type I IFN, IL-10, and activin A. Time-dependent differences in cytokine gene expression were noted in human monocytes treated with the CSFs; in this system, GM-CSF induced a more dramatic expression of IFN-regulated factor 4 (IRF4) than of IRF5, whereas M-CSF induced IRF5 but not IRF4. In the presence of both CSFs, some evidence of "competition" at the level of gene expression was observed. Care needs to be exercised when drawing definitive conclusions from a particular in vitro system about the roles of GM-CSF and M-CSF in macrophage lineage biology.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Animales , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Inmunofenotipificación , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Monocitos/inmunología
17.
Sci Rep ; 14(1): 2949, 2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38316918

RESUMEN

Plasmodium falciparum infection causes the most severe form of malaria, where excessive production of proinflammatory cytokines can drive the pathogenesis of the disease. Monocytes play key roles in host defense against malaria through cytokine production and phagocytosis; however, they are also implicated in pathogenesis through excessive proinflammatory cytokine production. Understanding the underlying molecular mechanisms that contribute to inflammatory cytokine production in P. falciparum-exposed monocytes is key towards developing better treatments. Here, we provide molecular evidence that histone 3 lysine 4 (H3K4) methylation is key for inflammatory cytokine production in P. falciparum-exposed monocytes. In an established in vitro system that mimics blood stage infection, elevated proinflammatory TNF and IL-6 cytokine production is correlated with increased mono- and tri-methylated H3K4 levels. Significantly, we demonstrate through utilizing a pharmacological inhibitor of H3K4 methylation that TNF and IL-6 expression can be suppressed in P. falciparum-exposed monocytes. This elucidated epigenetic regulatory mechanism, controlling inflammatory cytokine production, potentially provides new therapeutic options for future malaria treatment.


Asunto(s)
Malaria Falciparum , Malaria , Humanos , Plasmodium falciparum/metabolismo , Monocitos/metabolismo , Interleucina-6/metabolismo , Citocinas/metabolismo , Malaria/metabolismo , Epigénesis Genética
18.
Brain Sci ; 14(1)2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38248304

RESUMEN

Glioblastoma is highly proliferative and invasive. However, the regulatory cytokine networks that promote glioblastoma cell proliferation and invasion into other areas of the brain are not fully defined. In the present study, we define a critical role for the IL-11/IL-11Rα signalling axis in glioblastoma proliferation, epithelial to mesenchymal transition, and invasion. We identified enhanced IL-11/IL-11Rα expression correlated with reduced overall survival in glioblastoma patients using TCGA datasets. Proteomic analysis of glioblastoma cell lines overexpressing IL-11Rα displayed a proteome that favoured enhanced proliferation and invasion. These cells also displayed greater proliferation and migration, while the knockdown of IL-11Rα reversed these tumourigenic characteristics. In addition, these IL-11Rα overexpressing cells displayed enhanced invasion in transwell invasion assays and in 3D spheroid invasion assays, while knockdown of IL-11Rα resulted in reduced invasion. Furthermore, IL-11Rα-overexpressing cells displayed a more mesenchymal-like phenotype compared to parental cells and expressed greater levels of the mesenchymal marker Vimentin. Overall, our study identified that the IL-11/IL-11Rα pathway promotes glioblastoma cell proliferation, EMT, and invasion.

19.
Mol Immunol ; 166: 101-109, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38278031

RESUMEN

Transforming growth factor-ß (TGF-ß) is a pleiotropic cytokine essential for multiple biological processes, including the regulation of inflammatory and immune responses. One of the important functions of TGF-ß is the suppression of the proinflammatory cytokine interleukin-12 (IL-12), which is crucial for mounting an anti-tumorigenic response. Although the regulation of the IL-12p40 subunit (encoded by the IL-12B gene) of IL-12 has been extensively investigated, the knowledge of IL-12p35 (encoded by IL-12A gene) subunit regulation is relatively limited. This study investigates the molecular regulation of IL-12A by TGF-ß-activated signaling pathways in THP-1 monocytes. Our study identifies a complex regulation of IL-12A gene expression by TGF-ß, which involves multiple cellular signaling pathways, such as Smad2/3, NF-κB, p38 and JNK1/2. Pharmacological inhibition of NF-κB signaling decreased IL-12A expression, while blocking the Smad2/3 signaling pathway by overexpression of Smad7 and inhibiting JNK1/2 signaling with a pharmacological inhibitor, SP600125, increased its expression. The elucidated signaling pathways that regulate IL-12A gene expression potentially provide new therapeutic targets to increase IL-12 levels in the tumor microenvironment.


Asunto(s)
Subunidad p35 de la Interleucina-12 , Factor de Crecimiento Transformador beta , Citocinas , Expresión Génica , Interleucina-12 , Subunidad p35 de la Interleucina-12/metabolismo , Monocitos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Humanos
20.
Front Immunol ; 14: 1196931, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37457726

RESUMEN

Rheumatoid arthritis (RA) is a destructive inflammatory autoimmune disease that causes pain and disability. Many of the currently available drugs for treating RA patients are aimed at halting the progression of the disease and alleviating inflammation. Further, some of these treatment options have drawbacks, including disease recurrence and adverse effects due to long-term use. These inefficiencies have created a need for a different approach to treating RA. Recently, the focus has shifted to direct targeting of transcription factors (TFs), as they play a vital role in the pathogenesis of RA, activating key cytokines, chemokines, adhesion molecules, and enzymes. In light of this, synthetic drugs and natural compounds are being explored to target key TFs or their signaling pathways in RA. This review discusses the role of four key TFs in inflammation, namely NF-κB, STATs, AP-1 and IRFs, and their potential for being targeted to treat RA.


Asunto(s)
Artritis Reumatoide , Factores de Transcripción , Humanos , FN-kappa B/metabolismo , Transducción de Señal , Inflamación/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA