Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 148(3): 447-57, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22304915

RESUMEN

Although it is believed that neural activation can affect immune responses, very little is known about the neuroimmune interactions involved, especially the regulators of immune traffic across the blood-brain barrier which occurs in neuroimmune diseases such as multiple sclerosis (MS). Using a mouse model of MS, experimental autoimmune encephalomyelitis, we show that autoreactive T cells access the central nervous system via the fifth lumbar spinal cord. This location is defined by IL-6 amplifier-dependent upregulation of the chemokine CCL20 in associated dorsal blood vessels, which in turn depends on gravity-induced activation of sensory neurons by the soleus muscle in the leg. Impairing soleus muscle contraction by tail suspension is sufficient to reduce localized chemokine expression and block entry of pathogenic T cells at the fifth lumbar cord, suggesting that regional neuroimmune interactions may offer therapeutic targets for a variety of neurological diseases.


Asunto(s)
Barrera Hematoencefálica , Linfocitos T CD4-Positivos/citología , Encefalomielitis Autoinmune Experimental/inmunología , Animales , Movimiento Celular , Quimiocina CCL20/inmunología , Encefalomielitis Autoinmune Experimental/patología , Gravitación , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Músculo Esquelético/inervación , Neuroinmunomodulación , Médula Espinal/irrigación sanguínea
2.
Cell ; 138(5): 961-75, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19737522

RESUMEN

Obesity is associated with chronic low-grade inflammation that negatively impacts insulin sensitivity. Here, we show that high-fat diet can increase NF-kappaB activation in mice, which leads to a sustained elevation in level of IkappaB kinase epsilon (IKKepsilon) in liver, adipocytes, and adipose tissue macrophages. IKKepsilon knockout mice are protected from high-fat diet-induced obesity, chronic inflammation in liver and fat, hepatic steatosis, and whole-body insulin resistance. These mice show increased energy expenditure and thermogenesis via enhanced expression of the uncoupling protein UCP1. They maintain insulin sensitivity in liver and fat, without activation of the proinflammatory JNK pathway. Gene expression analyses indicate that IKKepsilon knockout reduces expression of inflammatory cytokines, and changes expression of certain regulatory proteins and enzymes involved in glucose and lipid metabolism. Thus, IKKepsilon may represent an attractive therapeutic target for obesity, insulin resistance, diabetes, and other complications associated with these disorders.


Asunto(s)
Metabolismo Energético , Quinasa I-kappa B/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Hígado Graso , Quinasa I-kappa B/genética , Insulina/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/metabolismo , Obesidad/inmunología
3.
BMC Cancer ; 22(1): 497, 2022 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-35513776

RESUMEN

BACKGROUND: Current cancer immunotherapies have made tremendous impacts but generally lack high response rates, especially in ovarian cancer. New therapies are needed to provide increased benefits. One understudied approach is to target the large population of immunosuppressive tumor-associated macrophages (TAMs). Using inducible transgenic mice, we recently reported that upregulating nuclear factor-kappaB (NF-κB) signaling in TAMs promotes the M1, anti-tumor phenotype and limits ovarian cancer progression. We also developed a mannose-decorated polymeric nanoparticle system (MnNPs) to preferentially deliver siRNA payloads to M2, pro-tumor macrophages in vitro. In this study, we tested a translational strategy to repolarize ovarian TAMs via MnNPs loaded with siRNA targeting the inhibitor of NF-κB alpha (IκBα) using mouse models of ovarian cancer. METHODS: We evaluated treatment with MnNPs loaded with IκBα siRNA (IκBα-MnNPs) or scrambled siRNA in syngeneic ovarian cancer models. ID8 tumors in C57Bl/6 mice were used to evaluate consecutive-day treatment of late-stage disease while TBR5 tumors in FVB mice were used to evaluate repetitive treatments in a faster-developing disease model. MnNPs were evaluated for biodistribution and therapeutic efficacy in both models. RESULTS: Stimulation of NF-κB activity and repolarization to an M1 phenotype via IκBα-MnNP treatment was confirmed using cultured luciferase-reporter macrophages. Delivery of MnNPs with fluorescent payloads (Cy5-MnNPs) to macrophages in the solid tumors and ascites was confirmed in both tumor models. A three consecutive-day treatment of IκBα-MnNPs in the ID8 model validated a shift towards M1 macrophage polarization in vivo. A clear therapeutic effect was observed with biweekly treatments over 2-3 weeks in the TBR5 model where significantly reduced tumor burden was accompanied by changes in immune cell composition, indicative of reduced immunosuppressive tumor microenvironment. No evidence of toxicity associated with MnNP treatment was observed in either model. CONCLUSIONS: In mouse models of ovarian cancer, MnNPs were preferentially associated with macrophages in ascites fluid and solid tumors. Evidence of macrophage repolarization, increased inflammatory cues, and reduced tumor burden in IκBα-MnNP-treated mice indicate beneficial outcomes in models of established disease. We have provided evidence of a targeted, TAM-directed approach to increase anti-tumor immunity in ovarian cancer with strong translational potential for future clinical studies.


Asunto(s)
Nanopartículas , Neoplasias Ováricas , Animales , Ascitis , Carcinoma Epitelial de Ovario , Modelos Animales de Enfermedad , Femenino , Humanos , Manosa/farmacología , Manosa/uso terapéutico , Ratones , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , ARN Interferente Pequeño/farmacología , Distribución Tisular , Microambiente Tumoral
4.
J Immunol ; 205(4): 1125-1134, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32601098

RESUMEN

NF-κB is a reduction-oxidation-sensitive transcription factor that plays a key role in regulating the immune response. In these studies, we intended to investigate the role of mitochondrial-derived reactive oxygen species in regulating NF-κB activation by studying transgenic mice that overexpress mitochondrial-targeted human catalase (mCAT). We treated wild-type (WT) and mCAT mice with intratracheal instillation of Escherichia coli LPS and found that mCAT mice had exaggerated NF-κB activation in the lungs, increased neutrophilic alveolitis, and greater lung inflammation/injury compared with WT mice. Additional studies using bone marrow chimeras revealed that this hyperinflammatory phenotype was mediated by immune/inflammatory cells. Mechanistic studies using bone marrow-derived macrophages (BMDMs) showed that LPS treatment induced a sustained increase in NF-κB activation and expression of NF-κB-dependent inflammatory mediators in mCAT BMDMs compared with WT BMDMs. Further investigations showed that cytoplasmic, but not mitochondrial, hydrogen peroxide levels were reduced in LPS-treated mCAT BMDMs. However, mCAT macrophages exhibited increased glycolytic and oxidative metabolism, coupled with increased ATP production and an increased intracellular NADH/NAD+ ratio compared with BMDMs from WT mice. Treatment of BMDMs with lactate increased the intracellular NADH/NAD+ ratio and upregulated NF-κB activation after LPS treatment, whereas treatment with a potent inhibitor of the mitochondrial pyruvate carrier (UK5099) decreased the NADH/NAD+ ratio and reduced NF-κB activation. Taken together, these findings point to an increased availability of reducing equivalents in the form of NADH as an important mechanism by which metabolic activity modulates inflammatory signaling through the NF-κB pathway.


Asunto(s)
Catalasa/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Neumonía/metabolismo , Animales , Médula Ósea/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NAD/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología
5.
BMC Cancer ; 20(1): 970, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33028251

RESUMEN

BACKGROUND: New treatment options for ovarian cancer are urgently required. Tumor-associated macrophages (TAMs) are an attractive target for therapy; repolarizing TAMs from M2 (pro-tumor) to M1 (anti-tumor) phenotypes represents an important therapeutic goal. We have previously shown that upregulated NF-kappaB (NF-κB) signaling in macrophages promotes M1 polarization, but effects in the context of ovarian cancer are unknown. Therefore, we aimed to investigate the therapeutic potential of increasing macrophage NF-κB activity in immunocompetent mouse models of ovarian cancer. METHODS: We have generated a transgenic mouse model, termed IKFM, which allows doxycycline-inducible overexpression of a constitutively active form of IKK2 (cIKK2) specifically within macrophages. The IKFM model was used to evaluate effects of increasing macrophage NF-κB activity in syngeneic murine TBR5 and ID8-Luc models of ovarian cancer in two temporal windows: 1) in established tumors, and 2) during tumor implantation and early tumor growth. Tumor weight, ascites volume, ascites supernatant and cells, and solid tumor were collected at sacrifice. Populations of macrophages and T cells within solid tumor and/or ascites were analyzed by immunofluorescent staining and qPCR, and soluble factors in ascitic fluid were analyzed by ELISA. Comparisons of control versus IKFM groups were performed by 2-tailed Mann-Whitney test, and a P-value < 0.05 was considered statistically significant. RESULTS: Increased expression of the cIKK2 transgene in TAMs from IKFM mice was confirmed at the mRNA and protein levels. Tumors from IKFM mice, regardless of the timing of doxycycline (dox) administration, demonstrated greater necrosis and immune infiltration than control tumors. Analysis of IKFM ascites and tumors showed sustained shifts in macrophage populations away from the M2 and towards the anti-tumor M1 phenotype. There were also increased tumor-infiltrating CD3+/CD8+ T cells in IKFM mice, accompanied by higher levels of CXCL9, a T cell activating factor secreted by macrophages, in IKFM ascitic fluid. CONCLUSIONS: In syngeneic ovarian cancer models, increased canonical NF-κB signaling in macrophages promoted anti-tumor TAM phenotypes and increased cytotoxic T cell infiltration, which was sufficient to limit tumor progression. This may present a novel translational approach for ovarian cancer treatment, with the potential to increase responses to T cell-directed therapy in future studies.


Asunto(s)
Macrófagos/metabolismo , FN-kappa B/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Ratones Transgénicos , Transducción de Señal
6.
J Immunol ; 196(8): 3411-20, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26951798

RESUMEN

Inflammation in the developing preterm lung leads to disrupted airway morphogenesis and chronic lung disease in human neonates. However, the molecular mechanisms linking inflammation and the pathways controlling airway morphogenesis remain unclear. In this article, we show that IL-1ß released by activated fetal lung macrophages is the key inflammatory mediator that disrupts airway morphogenesis. In mouse lung explants, blocking IL-1ß expression, posttranslational processing, and signaling protected the formation of new airways from the inhibitory effects ofEscherichia coliLPS. Consistent with a critical role for IL-1ß, mice expressing a gain-of-functionNlrp3allele and subsequent overactive inflammasome activity displayed abnormal saccular-stage lung morphogenesis and died soon after birth. Although the early-stage fetal lung appeared capable of mounting an NF-κB-mediated immune response, airway formation became more sensitive to inflammation later in development. This period of susceptibility coincided with higher expression of multiple inflammasome components that could increase the ability to release bioactive IL-1ß. Macrophages fromNlrp3gain-of-function mice also expressed higher levels of more mature cell surface markers, additionally linking inflammasome activation with macrophage maturation. These data identify developmental expression of the inflammasome and IL-1ß release by fetal lung macrophages as key mechanisms and potential therapeutic targets for neonatal lung disease.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Pulmón/embriología , Macrófagos/inmunología , Animales , Displasia Broncopulmonar/embriología , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/biosíntesis , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal/inmunología
7.
J Immunol ; 196(4): 1891-9, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773153

RESUMEN

Although numerous studies have demonstrated a critical role for canonical NF-κB signaling in inflammation and disease, the function of the noncanonical NF-κB pathway remains ill-defined. In lung tissue from patients with acute respiratory distress syndrome, we identified increased expression of the noncanonical pathway component p100/p52. To investigate the effects of p52 expression in vivo, we generated a novel transgenic mouse model with inducible expression of p52 in Clara cell secretory protein-expressing airway epithelial cells. Although p52 overexpression alone did not cause significant inflammation, p52 overexpression caused increased lung inflammation, injury, and mortality following intratracheal delivery of Escherichia coli LPS. No differences in cytokine/chemokine expression were measured between p52-overexpressing mice and controls, but increased apoptosis of Clara cell secretory protein-positive airway epithelial cells was observed in transgenic mice after LPS stimulation. In vitro studies in lung epithelial cells showed that p52 overexpression reduced cell survival and increased the expression of several proapoptotic genes during cellular stress. Collectively, these studies demonstrate a novel role for p52 in cell survival/apoptosis of airway epithelial cells and implicate noncanonical NF-κB signaling in the pathogenesis of acute respiratory distress syndrome.


Asunto(s)
Apoptosis/inmunología , Subunidad p52 de NF-kappa B/inmunología , Síndrome de Dificultad Respiratoria/patología , Mucosa Respiratoria/patología , Animales , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Subunidad p52 de NF-kappa B/biosíntesis , Neumonía/inmunología , Neumonía/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Síndrome de Dificultad Respiratoria/inmunología , Mucosa Respiratoria/inmunología , Transducción de Señal/inmunología , Regulación hacia Arriba
8.
J Immunol ; 193(3): 1184-93, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24981452

RESUMEN

In preterm infants, exposure to inflammation increases the risk of bronchopulmonary dysplasia, a chronic, developmental lung disease. Although macrophages are the key cells that initiate lung inflammation, less is known about lung macrophage phenotype and maturation. We hypothesized that fetal lung macrophages mature into distinct subpopulations during mouse development, and that activation could influence macrophage maturation. Expression of the fetal macrophage markers CD68, CD86, CD206, Ym1, fibrinogen-like protein 2, and indolamine-2, 3-dioxygenase was developmentally regulated, with each marker having different temporal patterns. Flow cytometry analysis showed macrophages within the fetal lung were less diverse than the distinctly separate subpopulations in newborn and adult lungs. Similar to adult alveolar macrophages, fetal lung macrophages responded to the TLR4 agonist LPS and the alternative activation cytokines IL-4 and IL-13. Using a macrophage-specific constitutively active IκB Kinase transgenic model (IKFM), we demonstrated that macrophage activation increased proinflammatory gene expression and reduced the response of fetal lung macrophages to IL-4 and IL-13. Activation also increased fetal lung macrophage proliferation. Fetal IKFM lungs contained increased percentages of more mature, CD11b(low)F4/80(high) cells that also expressed higher levels of the alternative activation markers CD204 and CD206. Development of fetal lung macrophages into mature alveolar macrophages may therefore include features of both proinflammatory and alternative activation paradigms.


Asunto(s)
Diferenciación Celular/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Quinasa I-kappa B/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Peritoneales/inmunología , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Diferenciación Celular/genética , Activación Enzimática/inmunología , Femenino , Regulación Enzimológica de la Expresión Génica/inmunología , Humanos , Quinasa I-kappa B/fisiología , Inmunofenotipificación , Inflamación/enzimología , Inflamación/inmunología , Inflamación/patología , Enfermedades Pulmonares/enzimología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/patología , Activación de Macrófagos/inmunología , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/patología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Am J Respir Cell Mol Biol ; 53(1): 50-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25375039

RESUMEN

Characterization of markers that identify activated macrophages could advance understanding of inflammatory lung diseases and facilitate development of novel methodologies for monitoring disease activity. We investigated whether folate receptor ß (FRß) expression could be used to identify and quantify activated macrophages in the lungs during acute inflammation induced by Escherichia coli LPS. We found that FRß expression was markedly increased in lung macrophages at 48 hours after intratracheal LPS. In vivo molecular imaging with a fluorescent probe (cyanine 5 polyethylene glycol folate) showed that the fluorescence signal over the chest peaked at 48 hours after intratracheal LPS and was markedly attenuated after depletion of macrophages. Using flow cytometry, we identified the cells responsible for uptake of cyanine 5-conjugated folate as FRß(+) interstitial macrophages and pulmonary monocytes, which coexpressed markers associated with an M1 proinflammatory macrophage phenotype. These findings were confirmed using a second model of acute lung inflammation generated by inducible transgenic expression of an NF-κB activator in airway epithelium. Using CC chemokine receptor 2-deficient mice, we found that FRß(+) macrophage/monocyte recruitment was dependent on the monocyte chemotactic protein-1/CC chemokine receptor 2 pathway. Together, our results demonstrate that folate-based molecular imaging can be used as a noninvasive approach to detect classically activated monocytes/macrophages recruited to the lungs during acute inflammation.


Asunto(s)
Receptor 2 de Folato/metabolismo , Regulación de la Expresión Génica , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Imagen Molecular , Neumonía/metabolismo , Enfermedad Aguda , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Escherichia coli/química , Colorantes Fluorescentes/farmacología , Receptor 2 de Folato/genética , Lipopolisacáridos/química , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/patología , Receptores CCR2/genética , Receptores CCR2/metabolismo
10.
Am J Physiol Regul Integr Comp Physiol ; 309(9): R1144-52, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26377563

RESUMEN

Inflammatory lung diseases (e.g., pneumonia and acute respiratory distress syndrome) are associated with hyperglycemia, even in patients without a prior diagnosis of Type 2 diabetes. It is unknown whether the lung inflammation itself or the accompanying comorbidities contribute to the increased risk of hyperglycemia and insulin resistance. To investigate whether inflammatory signaling by airway epithelial cells can induce systemic insulin resistance, we used a line of doxycycline-inducible transgenic mice that express a constitutive activator of the NF-κB in airway epithelial cells. Airway inflammation with accompanying neutrophilic infiltration was induced with doxycycline over 5 days. Then, hyperinsulinemic-euglycemic clamps were performed in chronically catheterized, conscious mice to assess insulin action. Lung inflammation decreased the whole body glucose requirements and was associated with secondary activation of inflammation in multiple tissues. Metabolic changes occurred in the absence of hypoxemia. Lung inflammation markedly attenuated insulin-induced suppression of hepatic glucose production and moderately impaired insulin action in peripheral tissues. The hepatic Akt signaling pathway was intact, while hepatic markers of inflammation and plasma lactate were increased. As insulin signaling was intact, the inability of insulin to suppress glucose production in the liver could have been driven by the increase in lactate, which is a substrate for gluconeogenesis, or due to an inflammation-driven signal that is independent of Akt. Thus, localized airway inflammation that is observed during inflammatory lung diseases can contribute to systemic inflammation and insulin resistance.


Asunto(s)
Glucemia/metabolismo , Resistencia a la Insulina , Insulina/sangre , Pulmón/metabolismo , FN-kappa B/metabolismo , Neumonía/metabolismo , Animales , Asma , Citocinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
11.
BMC Cancer ; 15: 647, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26424146

RESUMEN

BACKGROUND: Approximately 1 in 5 women diagnosed with breast cancer are considered to have in situ disease, most often termed ductal carcinoma in situ (DCIS). Though recognized as a risk factor for the development of more invasive cancer, it remains unclear what factors contribute to DCIS development. It has been shown that inflammation contributes to the progression of a variety of tumor types, and nuclear factor kappa B (NF-κB) is recognized as a master-regulator of inflammatory signaling. However, the contributions of NF-κB signaling to tumor initiation are less well understood. Aberrant up-regulation of NF-κB activity, either systemically or locally within the breast, could occur due to a variety of commonly experienced stimuli such as acute infection, obesity, or psychological stress. In this study, we seek to determine if activation of NF-κB in mammary epithelium could play a role in the formation of hyperplastic ductal lesions. METHODS: Our studies utilize a doxycycline-inducible transgenic mouse model in which constitutively active IKKß is expressed specifically in mammary epithelium. All previously published models of NF-κB modulation in the virgin mammary gland have been constitutive models, with transgene or knock-out present throughout the life and development of the animal. For the first time, we will induce activation at later time points after normal ducts have formed, thus being able to determine if NF-κB activation can promote pre-malignant changes in previously normal mammary epithelium. RESULTS: We found that even a short pulse of NF-κB activation could induce profound remodeling of mammary ductal structures. Short-term activation created hyperproliferative, enlarged ducts with filled lumens. Increased expression of inflammatory markers was concurrent with the down-regulation of hormone receptors and markers of epithelial differentiation. Furthermore, the oncoprotein mucin 1, known to be up-regulated in human and mouse DCIS, was over-expressed and mislocalized in the activated ductal tissue. CONCLUSIONS: These results indicate that aberrant NF-κB activation within mammary epithelium can lead to molecular and morphological changes consistent with the earliest stages of breast cancer. Thus, inhibition of NF-κB signaling following acute inflammation or the initial signs of hyperplastic ductal growth could represent an important opportunity for breast cancer prevention.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma in Situ/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , FN-kappa B/metabolismo , Transducción de Señal , Animales , Biomarcadores , Neoplasias de la Mama/genética , Carcinoma in Situ/genética , Carcinoma Ductal de Mama/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Epitelio/metabolismo , Epitelio/patología , Femenino , Expresión Génica , Humanos , Hiperplasia , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Ratones Transgénicos , FN-kappa B/genética , Clasificación del Tumor , Especificidad de Órganos/genética
12.
J Immunol ; 190(9): 4786-94, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23530143

RESUMEN

Although reactive oxygen species (ROS) produced by NADPH oxidase are known to regulate inflammatory responses, the impact of ROS on intracellular signaling pathways is incompletely understood. In these studies, we treated wild-type (WT) and p47(phox)-deficient mice with LPS to investigate mechanisms by which NADPH oxidase regulates signaling through the NF-κB pathway. After intratracheal instillation of LPS, ROS generation was impaired in p47(phox)(-/-) mice, whereas these mice had increased neutrophilic alveolitis and greater lung injury compared with WT controls. In mice interbred with transgenic NF-κB reporters (HIV-long terminal repeat/luciferase [HLL]), we found exaggerated LPS-induced NF-κB activation and increased expression of proinflammatory cytokines in lungs of p47(phox)(-/-)/HLL mice compared with controls. Both lung macrophages and bone marrow-derived macrophages (BMDMs) isolated from p47(phox)(-/-)/HLL mice showed enhanced LPS-stimulated NF-κB activity compared with controls. Although nuclear translocation of NF-κB proteins was similar between genotypes, EMSAs under nonreducing conditions showed increased DNA binding in p47(phox)(-/-)/HLL BMDMs, suggesting that ROS production reduces NF-κB binding to DNA without affecting nuclear translocation. Increased intracellular reduced glutathione/glutathione disulfide ratio and greater nuclear redox factor 1 (Ref-1) levels were present in p47(phox)(-/-)/HLL compared with WT BMDMs, pointing to NADPH oxidase modulating intracellular redox status in macrophages. Treatment with the Ref-1-specific inhibitor E3330 or hydrogen peroxide inhibited LPS-induced NF-κB activation in p47(phox)(-/-)/HLL BMDMs but not in WT/HLL cells. Consistent with these findings, small interfering RNA against Ref-1 selectively reduced NF-κB activity in LPS-treated p47(phox)(-/-)/HLL BMDMs. Together, these results indicate that NADPH oxidase limits LPS-induced NF-κB transcriptional activity through regulation of intracellular redox state.


Asunto(s)
Lipopolisacáridos/inmunología , Lesión Pulmonar/metabolismo , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Neumonía/metabolismo , Animales , ADN-(Sitio Apurínico o Apirimidínico) Liasa/inmunología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/inmunología , Ratones , NADPH Oxidasas/inmunología , FN-kappa B/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Oxidación-Reducción , Neumonía/inducido químicamente , Neumonía/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo
13.
Lab Invest ; 94(4): 409-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24535260

RESUMEN

Pancreatic cancer occurs in the setting of a profound fibrotic microenvironment that often dwarfs the actual tumor. Although pancreatic fibrosis has been well studied in chronic pancreatitis, its development in pancreatic cancer is much less well understood. This article describes the dynamic remodeling that occurs from pancreatic precursors (pancreatic intraepithelial neoplasias (PanINs)) to pancreatic ductal adenocarcinoma, highlighting similarities and differences between benign and malignant disease. Although collagen matrix is a commonality throughout this process, early stage PanINs are virtually free of periostin while late stage PanIN and pancreatic cancer are surrounded by an increasing abundance of this extracellular matrix protein. Myofibroblasts also become increasingly abundant during progression from PanIN to cancer. From the earliest stages of fibrogenesis, macrophages are associated with this ongoing process. In vitro co-culture indicates there is cross-regulation between macrophages and pancreatic stellate cells (PaSCs), precursors to at least some of the fibrotic cell populations. When quiescent PaSCs were co-cultured with macrophage cell lines, the stellate cells became activated and the macrophages increased cytokine production. In summary, fibrosis in pancreatic cancer involves a complex interplay of cells and matrices that regulate not only the tumor epithelium but the composition of the microenvironment itself.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Macrófagos/fisiología , Páncreas/patología , Neoplasias Pancreáticas/inmunología , Células Estrelladas Pancreáticas/fisiología , Animales , Carcinoma Ductal Pancreático/patología , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Metaplasia , Ratones , Neoplasias Pancreáticas/patología , Receptor Cross-Talk
14.
Proc Natl Acad Sci U S A ; 108(26): 10562-7, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21670280

RESUMEN

Evidence of endoplasmic reticulum (ER) stress has been found in lungs of patients with familial and sporadic idiopathic pulmonary fibrosis. We tested whether ER stress causes or exacerbates lung fibrosis by (i) conditional expression of a mutant form of surfactant protein C (L188Q SFTPC) found in familial interstitial pneumonia and (ii) intratracheal treatment with the protein misfolding agent tunicamycin. We developed transgenic mice expressing L188Q SFTPC exclusively in type II alveolar epithelium by using the Tet-On system. Expression of L188Q SFTPC induced ER stress, as determined by increased expression of heavy-chain Ig binding protein (BiP) and splicing of X-box binding protein 1 (XBP1) mRNA, but no lung fibrosis was identified in the absence of a second profibrotic stimulus. After intratracheal bleomycin, L188Q SFTPC-expressing mice developed exaggerated lung fibrosis and reduced static lung compliance compared with controls. Bleomycin-treated L188Q SFTPC mice also demonstrated increased apoptosis of alveolar epithelial cells and greater numbers of fibroblasts in the lungs. With a complementary model, intratracheal tunicamycin treatment failed to induce lung remodeling yet resulted in augmentation of bleomycin-induced fibrosis. These data support the concept that ER stress produces a dysfunctional epithelial cell phenotype that facilitates fibrotic remodeling. ER stress pathways may serve as important therapeutic targets in idiopathic pulmonary fibrosis.


Asunto(s)
Retículo Endoplásmico/metabolismo , Pulmón/patología , Fibrosis Pulmonar/patología , Animales , Apoptosis/genética , Bleomicina/toxicidad , Péptidos y Proteínas de Señalización Intercelular , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Mutación , Péptidos/genética , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Proteína C Asociada a Surfactante Pulmonar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tunicamicina/toxicidad
15.
Mol Pharm ; 10(3): 975-87, 2013 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-23331322

RESUMEN

Macrophages represent an important therapeutic target, because their activity has been implicated in the progression of debilitating diseases such as cancer and atherosclerosis. In this work, we designed and characterized pH-responsive polymeric micelles that were mannosylated using "click" chemistry to achieve CD206 (mannose receptor)-targeted siRNA delivery. CD206 is primarily expressed on macrophages and dendritic cells and upregulated in tumor-associated macrophages, a potentially useful target for cancer therapy. The mannosylated nanoparticles improved the delivery of siRNA into primary macrophages by 4-fold relative to the delivery of a nontargeted version of the same carrier (p < 0.01). Further, treatment for 24 h with the mannose-targeted siRNA carriers achieved 87 ± 10% knockdown of a model gene in primary macrophages, a cell type that is typically difficult to transfect. Finally, these nanoparticles were also avidly recognized and internalized by human macrophages and facilitated the delivery of 13-fold more siRNA into these cells than into model breast cancer cell lines. We anticipate that these mannose receptor-targeted, endosomolytic siRNA delivery nanoparticles will become an enabling technology for targeting macrophage activity in various diseases, especially those in which CD206 is upregulated in macrophages present within the pathologic site. This work also establishes a generalizable platform that could be applied for "click" functionalization with other targeting ligands to direct siRNA delivery.


Asunto(s)
Micelas , Polímeros/administración & dosificación , Polímeros/química , Animales , Células Cultivadas , Química Clic , Células Dendríticas/metabolismo , Citometría de Flujo , Humanos , Lectinas Tipo C/genética , Macrófagos/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Microscopía Confocal , Nanopartículas/administración & dosificación , Nanopartículas/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/genética
16.
J Immunol ; 187(5): 2740-7, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21775686

RESUMEN

Bronchopulmonary dysplasia is a common pulmonary complication of extreme prematurity. Arrested lung development leads to bronchopulmonary dysplasia, but the molecular pathways that cause this arrest are unclear. Lung injury and inflammation increase disease risk, but the cellular site of the inflammatory response and the potential role of localized inflammatory signaling in inhibiting lung morphogenesis are not known. In this study, we show that tissue macrophages present in the fetal mouse lung mediate the inflammatory response to LPS and that macrophage activation inhibits airway morphogenesis. Macrophage depletion or targeted inactivation of the NF-κB signaling pathway protected airway branching in cultured lung explants from the effects of LPS. Macrophages also appear to be the primary cellular site of IL-1ß production following LPS exposure. Conversely, targeted NF-κB activation in transgenic macrophages was sufficient to inhibit airway morphogenesis. Macrophage activation in vivo inhibited expression of multiple genes critical for normal lung development, leading to thickened lung interstitium, reduced airway branching, and perinatal death. We propose that fetal lung macrophage activation contributes to bronchopulmonary dysplasia by generating a localized inflammatory response that disrupts developmental signals critical for lung formation.


Asunto(s)
Pulmón/crecimiento & desarrollo , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , FN-kappa B/inmunología , Transducción de Señal/inmunología , Animales , Displasia Broncopulmonar/inmunología , Displasia Broncopulmonar/metabolismo , Feto , Humanos , Recién Nacido , Lipopolisacáridos/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Microscopía Confocal , Morfogénesis , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Immunol ; 187(11): 5703-11, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22048774

RESUMEN

Macrophages have established roles in tumor growth and metastasis, but information about their role in lung tumor promotion is limited. To assess the role of macrophages in lung tumorigenesis, we developed a method of minimally invasive, long-term macrophage depletion by repetitive intratracheal instillation of liposomal clodronate. Compared with controls treated with repetitive doses of PBS-containing liposomes, long-term macrophage depletion resulted in a marked reduction in tumor number and size at 4 mo after a single i.p. injection of the carcinogen urethane. After urethane treatment, lung macrophages developed increased M1 macrophage marker expression during the first 2-3 wk, followed by increased M2 marker expression by week 6. Using a strategy to reduce alveolar macrophages during tumor initiation and early promotion stages (weeks 1-2) or during late promotion and progression stages (weeks 4-16), we found significantly fewer and smaller lung tumors in both groups compared with controls. Late-stage macrophage depletion reduced VEGF expression and impaired vascular growth in tumors. In contrast, early-stage depletion of alveolar macrophages impaired urethane-induced NF-κB activation in the lungs and reduced the development of premalignant atypical adenomatous hyperplasia lesions at 6 wk after urethane injection. Together, these studies elucidate an important role for macrophages in lung tumor promotion and indicate that these cells have distinct roles during different stages of lung carcinogenesis.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/inmunología , Neoplasias Pulmonares/inmunología , Macrófagos/inmunología , Uretano/toxicidad , Animales , Separación Celular , Transformación Celular Neoplásica/inducido químicamente , Femenino , Citometría de Flujo , Inmunohistoquímica , Neoplasias Pulmonares/inducido químicamente , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Mediators Inflamm ; 2013: 503213, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24347827

RESUMEN

INTRODUCTION: Since the NF-κB pathway regulates both inflammation and host defense, it is uncertain whether interventions targeting NF-κB would be beneficial in sepsis. Based on the kinetics of the innate immune response, we postulated that selective NF-κB inhibition during a defined time period after the onset of sepsis would reduce acute lung injury without compromising bacterial host defense. METHODS: Mice underwent cecal ligation and puncture (CLP). An NF-κB inhibitor, BMS-345541 (50 µg/g mice), was administered by peroral gavage beginning 2 hours after CLP and repeated at 6 hour intervals for 2 additional doses. RESULTS: Mice treated with BMS-345541 after CLP showed reduced neutrophilic alveolitis and lower levels of KC in bronchoalveolar lavage fluid compared to mice treated with CLP+vehicle. In addition, mice treated with CLP+BMS had minimal histological evidence of lung injury and normal wet-dry ratios, indicating protection from acute lung injury. Treatment with the NF-κB inhibitor did not affect the ability of cultured macrophages to phagocytose bacteria and did not alter bacterial colony counts in blood, lung tissue, or peritoneal fluid at 24 hours after CLP. While BMS-345541 treatment did not alter mortality after CLP, our results showed a trend towards improved survival. CONCLUSION: Transiently blocking NF-κB activity after the onset of CLP-induced sepsis can effectively reduce acute lung injury in mice without compromising bacterial host defense or survival after CLP.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Infecciones Bacterianas/inmunología , Imidazoles/uso terapéutico , FN-kappa B/antagonistas & inhibidores , Quinoxalinas/uso terapéutico , Sepsis/tratamiento farmacológico , Animales , Células Cultivadas , Quinasa I-kappa B/antagonistas & inhibidores , Ligadura , Macrófagos/inmunología , Ratones , Neutrófilos/inmunología , Fagocitosis/efectos de los fármacos , Sepsis/inmunología , Sepsis/mortalidad
19.
Cancers (Basel) ; 15(6)2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36980752

RESUMEN

Kirsten rat sarcoma virus (KRAS)-mutant cancers are frequent, metastatic, lethal, and largely undruggable. While interleukin (IL)-1ß and nuclear factor (NF)-κB inhibition hold promise against cancer, untargeted treatments are not effective. Here, we show that human KRAS-mutant cancers are addicted to IL-1ß via inflammatory versican signaling to macrophage inhibitor of NF-κB kinase (IKK) ß. Human pan-cancer and experimental NF-κB reporter, transcriptome, and proteome screens reveal that KRAS-mutant tumors trigger macrophage IKKß activation and IL-1ß release via secretory versican. Tumor-specific versican silencing and macrophage-restricted IKKß deletion prevents myeloid NF-κB activation and metastasis. Versican and IKKß are mutually addicted and/or overexpressed in human cancers and possess diagnostic and prognostic power. Non-oncogene KRAS/IL-1ß addiction is abolished by IL-1ß and TLR1/2 inhibition, indicating cardinal and actionable roles for versican and IKKß in metastasis.

20.
Carcinogenesis ; 33(4): 859-67, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22287559

RESUMEN

Since recent evidence indicates a requirement for epithelial nuclear factor (NF)-κB signaling in lung tumorigenesis, we investigated the impact of the NF-κB inhibitor bortezomib on lung tumor promotion and growth. We used an experimental model in which wild-type mice or mice expressing an NF-κB reporter received intraperitoneal urethane (1 g/kg) followed by twice weekly bortezomib (1 mg/kg) during distinct periods of tumor initiation/progression. Mice were serially assessed for lung NF-κB activation, inflammation and carcinogenesis. Short-term proteasome inhibition with bortezomib did not impact tumor formation but retarded the growth of established lung tumors in mice via effects on cell proliferation. In contrast, long-term treatment with bortezomib resulted in significantly increased lung tumor number and size. This tumor-promoting effect of prolonged bortezomib treatment was associated with perpetuation of urethane-induced inflammation and chronic upregulation of interleukin-1ß and proinflammatory C-X-C motif chemokine ligands (CXCL) 1 and 2 in the lungs. In addition to airway epithelium, bortezomib inhibited NF-κB in pulmonary macrophages in vivo, presenting a possible mechanism of tumor amplification. In this regard, RAW264.7 macrophages exposed to bortezomib showed increased expression of interleukin-1ß, CXCL1 and CXCL2. In conclusion, although short-term bortezomib may exert some beneficial effects, prolonged NF-κB inhibition accelerates chemical lung carcinogenesis by perpetuating carcinogen-induced inflammation. Inhibition of NF-κB in pulmonary macrophages appears to play an important role in this adverse process.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Neoplasias Pulmonares/patología , FN-kappa B/antagonistas & inhibidores , Pirazinas/farmacología , Animales , Bortezomib , Línea Celular , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA