Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 134(6)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38488009

RESUMEN

Uncontrolled accumulation of extracellular matrix leads to tissue fibrosis and loss of organ function. We previously demonstrated in vitro that the DNA/RNA-binding protein fused in sarcoma (FUS) promotes fibrotic responses by translocating to the nucleus, where it initiates collagen gene transcription. However, it is still not known whether FUS is profibrotic in vivo and whether preventing its nuclear translocation might inhibit development of fibrosis following injury. We now demonstrate that levels of nuclear FUS are significantly increased in mouse models of kidney and liver fibrosis. To evaluate the direct role of FUS nuclear translocation in fibrosis, we used mice that carry a mutation in the FUS nuclear localization sequence (FUSR521G) and the cell-penetrating peptide CP-FUS-NLS that we previously showed inhibits FUS nuclear translocation in vitro. We provide evidence that FUSR521G mice or CP-FUS-NLS-treated mice showed reduced nuclear FUS and fibrosis following injury. Finally, differential gene expression analysis and immunohistochemistry of tissues from individuals with focal segmental glomerulosclerosis or nonalcoholic steatohepatitis revealed significant upregulation of FUS and/or collagen genes and FUS protein nuclear localization in diseased organs. These results demonstrate that injury-induced nuclear translocation of FUS contributes to fibrosis and highlight CP-FUS-NLS as a promising therapeutic option for organ fibrosis.


Asunto(s)
Esclerosis Amiotrófica Lateral , ARN , Animales , Ratones , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Mutación , ADN , Fibrosis , Colágeno/metabolismo , Esclerosis Amiotrófica Lateral/genética
2.
JCI Insight ; 8(19)2023 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-37676731

RESUMEN

A hallmark of idiopathic pulmonary fibrosis (IPF) and other interstitial lung diseases is dysregulated repair of the alveolar epithelium. The Hippo pathway effector transcription factors YAP and TAZ are implicated as essential for type 1 and type 2 alveolar epithelial cell (AT1 and AT2) differentiation in the developing lung, yet aberrant activation of YAP/TAZ is a prominent feature of the dysregulated alveolar epithelium in IPF. In these studies, we sought to define the functional role of YAP/TAZ activity during alveolar regeneration. We demonstrated that Yap and Taz were normally activated in AT2 cells shortly after injury, and deletion of Yap/Taz in AT2 cells led to pathologic alveolar remodeling, failure of AT2-to-AT1 cell differentiation, increased collagen deposition, exaggerated neutrophilic inflammation, and increased mortality following injury induced by a single dose of bleomycin. Loss of Yap/Taz activity prior to an LPS injury prevented AT1 cell regeneration, led to intraalveolar collagen deposition, and resulted in persistent innate inflammation. These findings establish that AT2 cell Yap/Taz activity is essential for functional alveolar epithelial repair and prevention of fibrotic remodeling.


Asunto(s)
Lesión Pulmonar Aguda , Fibrosis Pulmonar Idiopática , Proteínas Señalizadoras YAP , Humanos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Colágeno/metabolismo , Fibrosis Pulmonar Idiopática/patología , Inflamación , Regeneración , Transducción de Señal , Proteínas Señalizadoras YAP/metabolismo
3.
Am J Respir Crit Care Med ; 206(5): 596-607, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35728047

RESUMEN

Rationale: Although persistent fibroblast activation is a hallmark of idiopathic pulmonary fibrosis (IPF), mechanisms regulating persistent fibroblast activation in the lungs have not been fully elucidated. Objectives: On the basis of our observation that lung fibroblasts express TBXA2R (thromboxane-prostanoid receptor) during fibrosis, we investigated the role of TBXA2R signaling in fibrotic remodeling. Methods: We identified TBXA2R expression in lungs of patients with IPF and mice and studied primary mouse and human lung fibroblasts to determine the impact of TBXA2R signaling on fibroblast activation. We used TBXA2R-deficient mice and small-molecule inhibitors to investigate TBXA2R signaling in preclinical lung fibrosis models. Measurements and Main Results: TBXA2R expression was upregulated in fibroblasts in the lungs of patients with IPF and in mouse lungs during experimental lung fibrosis. Genetic deletion of TBXA2R, but not inhibition of thromboxane synthase, protected mice from bleomycin-induced lung fibrosis, thereby suggesting that an alternative ligand activates profibrotic TBXA2R signaling. In contrast to thromboxane, F2-isoprostanes, which are nonenzymatic products of arachidonic acid induced by reactive oxygen species, were persistently elevated during fibrosis. F2-isoprostanes induced TBXA2R signaling in fibroblasts and mediated a myofibroblast activation profile due, at least in part, to potentiation of TGF-ß (transforming growth factor-ß) signaling. In vivo treatment with the TBXA2R antagonist ifetroban reduced profibrotic signaling in the lungs, protected mice from lung fibrosis in three preclinical models (bleomycin, Hermansky-Pudlak mice, and radiation-induced fibrosis), and markedly enhanced fibrotic resolution after bleomycin treatment. Conclusions: TBXA2R links oxidative stress to fibroblast activation during lung fibrosis. TBXA2R antagonists could have utility in treating pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Receptores de Tromboxanos , Animales , Bleomicina/farmacología , F2-Isoprostanos/metabolismo , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/genética , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Prostaglandinas/metabolismo , Receptores de Tromboxanos/metabolismo , Tromboxanos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
4.
Am J Respir Cell Mol Biol ; 67(3): 334-345, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35687143

RESUMEN

Loss of secretory IgA (SIgA) is common in chronic obstructive pulmonary disease (COPD) small airways and likely contributes to disease progression. We hypothesized that loss of SIgA results from reduced expression of pIgR (polymeric immunoglobulin receptor), a chaperone protein needed for SIgA transcytosis, in the COPD small airway epithelium. pIgR-expressing cells were defined and quantified at single-cell resolution in human airways using RNA in situ hybridization, immunostaining, and single-cell RNA sequencing. Complementary studies in mice used immunostaining, primary murine tracheal epithelial cell culture, and transgenic mice with secretory or ciliated cell-specific knockout of pIgR. SIgA degradation by human neutrophil elastase or secreted bacterial proteases from nontypeable Haemophilus influenzae was evaluated in vitro. We found that secretory cells are the predominant cell type responsible for pIgR expression in human and murine airways. Loss of SIgA in small airways was not associated with a reduction in secretory cells but rather a reduction in pIgR protein expression despite intact PIGR mRNA expression. Neutrophil elastase and nontypeable H. influenzae-secreted proteases are both capable of degrading SIgA in vitro and may also contribute to a deficient SIgA immunobarrier in COPD. Loss of the SIgA immunobarrier in small airways of patients with severe COPD is complex and likely results from both pIgR-dependent defects in IgA transcytosis and SIgA degradation.


Asunto(s)
Inmunoglobulina A Secretora , Enfermedad Pulmonar Obstructiva Crónica , Receptores de Inmunoglobulina Polimérica , Animales , Haemophilus influenzae/enzimología , Humanos , Inmunoglobulina A Secretora/metabolismo , Elastasa de Leucocito/metabolismo , Ratones , Proteolisis , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores de Inmunoglobulina Polimérica/genética , Receptores de Inmunoglobulina Polimérica/metabolismo , Sistema Respiratorio/metabolismo
5.
J Vis Exp ; (162)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32831298

RESUMEN

The ability to evaluate lung histology is critical for the fields of lung cancer research and cancer metastasis. It is equally important to perform necropsies rapidly and efficiently from studies without sacrificing the quality of the tissues procured. The goal of this protocol is to present a method to rapidly perfuse, inflate, and fix mouse lungs for downstream histological analysis. This method does not standardize lung inflation; thus, it does not require any special procedures or equipment and instead simply instills fixative directly through the trachea following perfusion through the heart. This allows for sufficient estimation of tumor size, histology, and scoring. This also allows for the collection of frozen tissue prior to lung tissue fixation. This method is limited in that it does not allow for later morphometric quantification of the lung; however, it is more than sufficient for lung tumor analysis from genetically engineered mouse models (GEMMs), syngeneic models, as well as xenograft tumor and metastasis studies.


Asunto(s)
Neoplasias Pulmonares/patología , Pulmón/patología , Perfusión , Animales , Humanos , Ratones , Coloración y Etiquetado , Fijación del Tejido , Ensayos Antitumor por Modelo de Xenoinjerto
6.
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
7.
ERJ Open Res ; 5(2)2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31149621

RESUMEN

This novel mouse model mimics malignant pleural effusion drainage using an indwelling pleural catheter in humans, and provides direct access to the pleural space potentially enabling the testing of intrapleural therapies in the treatment of MPE. bit.ly/2W2kzO0.

8.
Oncotarget ; 10(66): 7031-7042, 2019 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-31903163

RESUMEN

Growth arrest-specific 6 (Gas6) has been implicated in carcinogenesis through activation of its receptors, particularly MerTK. To investigate whether Gas6 plays a role in resistance to NF-κB inhibitors, which have not proven to be effective agents for lung cancer therapy, we studied lung cancer models induced by urethane injection or expression of mutant Kras (KrasG12D). We found that Gas6 is primarily produced by macrophages during tumorigenesis and that Gas6 is negatively regulated by NF-κB. Since Gas6 is a vitamin K dependent protein, we used low-dose warfarin to block Gas6 production and showed that this treatment inhibited tumorigenesis in both the urethane and KrasG12D models, most prominently in mice with targeted deletion of IKKß in myeloid cells (IKKßΔMye mice). In addition, MerTK deficient mice had reduced urethane-induced tumorigenesis. Inhibition of the Gas6-MerTK pathway in all these models reduced macrophages and neutrophils in the lungs of tumor-bearing mice. Analysis of mouse lung tumors revealed MerTK staining on tumor cells and in vitro studies showed that Gas6 increased proliferation of human lung cancer cell lines. To assess the therapeutic potential for combination treatment targeting NF-κB and Gas6-MerTK, we injected Lewis Lung Carcinoma cells subcutaneously and treated mice with Bay 11-70852 (NF-κB inhibitor) and/or Foretinib (MerTK inhibitor). While individual treatments were ineffective, combination therapy markedly reduced tumor growth, blocked tumor cell proliferation, reduced tumor-associated macrophages, and increased CD4+ T cells. Together, our studies unmask a role for Gas6-MerTK signaling in lung carcinogenesis and indicate that up-regulation of Gas6 production in macrophages could be a major mechanism of resistance to NF-κB inhibitors.

9.
JCI Insight ; 3(16)2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30135303

RESUMEN

ER stress in type II alveolar epithelial cells (AECs) is common in idiopathic pulmonary fibrosis (IPF), but the contribution of ER stress to lung fibrosis is poorly understood. We found that mice deficient in C/EBP homologous protein (CHOP), an ER stress-regulated transcription factor, were protected from lung fibrosis and AEC apoptosis in 3 separate models where substantial ER stress was identified. In mice treated with repetitive intratracheal bleomycin, we identified localized hypoxia in type II AECs as a potential mechanism explaining ER stress. To test the role of hypoxia in lung fibrosis, we treated mice with bleomycin, followed by exposure to 14% O2, which exacerbated ER stress and lung fibrosis. Under these experimental conditions, CHOP-/- mice, but not mice with epithelial HIF (HIF1/HIF2) deletion, were protected from AEC apoptosis and fibrosis. In vitro studies revealed that CHOP regulates hypoxia-induced apoptosis in AECs via the inositol-requiring enzyme 1α (IRE1α) and the PKR-like ER kinase (PERK) pathways. In human IPF lungs, CHOP and hypoxia markers were both upregulated in type II AECs, supporting a conclusion that localized hypoxia results in ER stress-induced CHOP expression, thereby augmenting type II AEC apoptosis and potentiating lung fibrosis.


Asunto(s)
Estrés del Retículo Endoplásmico , Fibrosis Pulmonar Idiopática/patología , Alveolos Pulmonares/patología , Factor de Transcripción CHOP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Bleomicina/toxicidad , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Endorribonucleasas/metabolismo , Femenino , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Masculino , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Factor de Transcripción CHOP/genética , eIF-2 Quinasa/metabolismo
10.
J Immunol ; 199(2): 510-519, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28576981

RESUMEN

The appropriate orchestration of different arms of the immune response is critical during viral infection to promote efficient viral clearance while limiting immunopathology. However, the signals and mechanisms that guide this coordination are not fully understood. IFNs are produced at high levels during viral infection and have convergent signaling through STAT1. We hypothesized that STAT1 signaling during viral infection regulates the balance of innate lymphoid cells (ILC), a diverse class of lymphocytes that are poised to respond to environmental insults including viral infections with the potential for both antiviral or immunopathologic functions. During infection with respiratory syncytial virus (RSV), STAT1-deficient mice had reduced numbers of antiviral IFN-γ+ ILC1 and increased numbers of immunopathologic IL-5+ and IL-13+ ILC2 and IL-17A+ ILC3 compared with RSV-infected wild-type mice. Using bone marrow chimeric mice, we found that both ILC-intrinsic and ILC-extrinsic factors were responsible for this ILC dysregulation during viral infection in STAT1-deficient mice. Regarding ILC-extrinsic mechanisms, we found that STAT1-deficient mice had significantly increased expression of IL-33 and IL-23, cytokines that promote ILC2 and ILC3, respectively, compared with wild-type mice during RSV infection. Moreover, disruption of IL-33 or IL-23 signaling attenuated cytokine-producing ILC2 and ILC3 responses in STAT1-deficient mice during RSV infection. Collectively, these data demonstrate that STAT1 is a key orchestrator of cytokine-producing ILC responses during viral infection via ILC-extrinsic regulation of IL-33 and IL-23.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Factor de Transcripción STAT1/metabolismo , Animales , Citocinas/biosíntesis , Regulación de la Expresión Génica , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-23/genética , Interleucina-23/inmunología , Interleucina-33/genética , Interleucina-33/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Linfocitos/clasificación , Ratones , Infecciones por Virus Sincitial Respiratorio/virología , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Transducción de Señal
11.
JCI Insight ; 1(17): e88947, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27777976

RESUMEN

Alveolar epithelial cell (AEC) dysfunction underlies the pathogenesis of pulmonary fibrosis in Hermansky-Pudlak syndrome (HPS) and other genetic syndromes associated with interstitial lung disease; however, mechanisms linking AEC dysfunction and fibrotic remodeling are incompletely understood. Since increased macrophage recruitment precedes pulmonary fibrosis in HPS, we investigated whether crosstalk between AECs and macrophages determines fibrotic susceptibility. We found that AECs from HPS mice produce excessive MCP-1, which was associated with increased macrophages in the lungs of unchallenged HPS mice. Blocking MCP-1/CCR2 signaling in HPS mice with genetic deficiency of CCR2 or targeted deletion of MCP-1 in AECs normalized macrophage recruitment, decreased AEC apoptosis, and reduced lung fibrosis in these mice following treatment with low-dose bleomycin. We observed increased TGF-ß production by HPS macrophages, which was eliminated by CCR2 deletion. Selective deletion of TGF-ß in myeloid cells or of TGF-ß signaling in AECs through deletion of TGFBR2 protected HPS mice from AEC apoptosis and bleomycin-induced fibrosis. Together, these data reveal a feedback loop in which increased MCP-1 production by dysfunctional AECs results in recruitment and activation of lung macrophages that produce TGF-ß, thus amplifying the fibrotic cascade through AEC apoptosis and stimulation of fibrotic remodeling.


Asunto(s)
Células Epiteliales/citología , Síndrome de Hermanski-Pudlak/inmunología , Macrófagos/citología , Fibrosis Pulmonar/inmunología , Animales , Bleomicina , Quimiocina CCL2/metabolismo , Susceptibilidad a Enfermedades , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , Alveolos Pulmonares/citología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores CCR2/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
12.
Oncoimmunology ; 5(6): e1168549, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27471643

RESUMEN

Several studies have demonstrated that NF-κB activation is common in lung cancer; however, the mechanistic links between NF-κB signaling and tumorigenesis remain to be fully elucidated. We investigated the function of NF-κB signaling in epidermal growth factor receptor (EGFR)-mutant lung tumors using a transgenic mouse model with doxycycline (dox)-inducible expression of oncogenic EGFR in the lung epithelium with or without a dominant inhibitor of NF-κB signaling. NF-κB inhibition resulted in a significant reduction in tumor burden in both EGFR tyrosine kinase inhibitor (TKI)-sensitive and resistant tumors. However, NF-κB inhibition did not alter epithelial cell survival in vitro or in vivo, and no changes were detected in activation of EGFR downstream signaling pathways. Instead, we observed an influx of inflammatory cells (macrophages and neutrophils) in the lungs of mice with oncogenic EGFR expression that was blocked in the setting of NF-κB inhibition. To investigate whether inflammatory cells play a role in promoting EGFR-mutant lung tumors, we depleted macrophages and neutrophils during tumorigenesis and found that neutrophil depletion had no effect on tumor formation, but macrophage depletion caused a significant reduction in tumor burden. Together, these data suggest that epithelial NF-κB signaling supports carcinogenesis in a non-cell autonomous manner in EGFR-mutant tumors through recruitment of pro-tumorigenic macrophages.

13.
Cell Rep ; 16(1): 120-132, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27320908

RESUMEN

Although epithelial NF-κB signaling is important for lung carcinogenesis, NF-κB inhibitors are ineffective for cancer treatment. To explain this paradox, we studied mice with genetic deletion of IKKß in myeloid cells and found enhanced tumorigenesis in Kras(G12D) and urethane models of lung cancer. Myeloid-specific inhibition of NF-κB augmented pro-IL-1ß processing by cathepsin G in neutrophils, leading to increased IL-1ß and enhanced epithelial cell proliferation. Combined treatment with bortezomib, a proteasome inhibitor that blocks NF-κB activation, and IL-1 receptor antagonist reduced tumor formation and growth in vivo. In lung cancer patients, plasma IL-1ß levels correlated with poor prognosis, and IL-1ß increased following bortezomib treatment. Together, our studies elucidate an important role for neutrophils and IL-1ß in lung carcinogenesis and resistance to NF-κB inhibitors.


Asunto(s)
Interleucina-1beta/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , FN-kappa B/antagonistas & inhibidores , Neutrófilos/metabolismo , Animales , Bortezomib/farmacología , Bortezomib/uso terapéutico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Quinasa I-kappa B/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , FN-kappa B/metabolismo , Neutrófilos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia
14.
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
15.
Oncotarget ; 7(5): 5470-82, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26756215

RESUMEN

Nuclear Factor (NF)-κB is positioned to provide the interface between COPD and carcinogenesis through regulation of chronic inflammation in the lungs. Using a tetracycline-inducible transgenic mouse model that conditionally expresses activated IκB kinase ß (IKKß) in airway epithelium (IKTA), we found that sustained NF-κB signaling results in chronic inflammation and emphysema by 4 months. By 11 months of transgene activation, IKTA mice develop lung adenomas. Investigation of lung inflammation in IKTA mice revealed a substantial increase in M2-polarized macrophages and CD4+/CD25+/FoxP3+ regulatory T lymphocytes (Tregs). Depletion of alveolar macrophages in IKTA mice reduced Tregs, increased lung CD8+ lymphocytes, and reduced tumor numbers following treatment with the carcinogen urethane. Alveolar macrophages from IKTA mice supported increased generation of inducible Foxp3+ Tregs ex vivo through expression of TGFß and IL-10. Targeting of TGFß and IL-10 reduced the ability of alveolar macrophages from IKTA mice to induce Foxp3 expression on T cells. These studies indicate that sustained activation of NF-κB pathway links COPD and lung cancer through generation and maintenance of a pro-tumorigenic inflammatory environment consisting of alternatively activated macrophages and regulatory T cells.


Asunto(s)
Epitelio/inmunología , Inflamación/inmunología , Neoplasias Pulmonares/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , FN-kappa B/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Animales , Western Blotting , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Femenino , Citometría de Flujo , Humanos , Quinasa I-kappa B/fisiología , Inmunosupresores/inmunología , Interleucina-10/genética , Interleucina-10/metabolismo , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Transgénicos , FN-kappa B/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
16.
Cancer Res ; 75(8): 1624-1634, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25691457

RESUMEN

Although the lung is the most common metastatic site for cancer cells, biologic mechanisms regulating lung metastasis are not fully understood. Using heterotopic and intravenous injection models of lung metastasis in mice, we found that IL5, a cytokine involved in allergic and infectious diseases, facilitates metastatic colonization through recruitment of sentinel eosinophils and regulation of other inflammatory/immune cells in the microenvironment of the distal lung. Genetic IL5 deficiency offered marked protection of the lungs from metastasis of different types of tumor cells, including lung cancer, melanoma, and colon cancer. IL5 neutralization protected subjects from metastasis, whereas IL5 reconstitution or adoptive transfer of eosinophils into IL5-deficient mice exerted prometastatic effects. However, IL5 deficiency did not affect the growth of the primary tumor or the size of metastatic lesions. Mechanistic investigations revealed that eosinophils produce CCL22, which recruits regulatory T cells to the lungs. During early stages of metastasis, Treg created a protumorigenic microenvironment, potentially by suppressing IFNγ-producing natural killer cells and M1-polarized macrophages. Together, our results establish a network of allergic inflammatory circuitry that can be co-opted by metastatic cancer cells to facilitate lung colonization, suggesting interventions to target this pathway may offer therapeutic benefits to prevent or treat lung metastasis.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Interleucina-5/fisiología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Microambiente Tumoral/inmunología , Animales , Carcinoma Pulmonar de Lewis/genética , Línea Celular Tumoral , Eosinófilos/patología , Femenino , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/inmunología , Escape del Tumor/genética , Microambiente Tumoral/genética
17.
Infect Immun ; 82(9): 3723-39, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24958709

RESUMEN

The Th17 cytokines interleukin-17A (IL-17A), IL-17F, and IL-22 are critical for the lung immune response to a variety of bacterial pathogens, including Klebsiella pneumoniae. Th2 cytokine expression in the airways is a characteristic feature of asthma and allergic airway inflammation. The Th2 cytokines IL-4 and IL-13 diminish ex vivo and in vivo IL-17A protein expression by Th17 cells. To determine the effect of IL-4 and IL-13 on IL-17-dependent lung immune responses to acute bacterial infection, we developed a combined model in which allergic airway inflammation and lung IL-4 and IL-13 expression were induced by ovalbumin sensitization and challenge prior to acute lung infection with K. pneumoniae. We hypothesized that preexisting allergic airway inflammation decreases lung IL-17A expression and airway neutrophil recruitment in response to acute K. pneumoniae infection and thereby increases the lung K. pneumoniae burden. As hypothesized, we found that allergic airway inflammation decreased the number of K. pneumoniae-induced airway neutrophils and lung IL-17A, IL-17F, and IL-22 expression. Despite the marked reduction in postinfection airway neutrophilia and lung expression of Th17 cytokines, allergic airway inflammation significantly decreased the lung K. pneumoniae burden and postinfection mortality. We showed that the decreased lung K. pneumoniae burden was independent of IL-4, IL-5, and IL-17A and partially dependent on IL-13 and STAT6. Additionally, we demonstrated that the decreased lung K. pneumoniae burden associated with allergic airway inflammation was both neutrophil and CCL8 dependent. These findings suggest a novel role for CCL8 in lung antibacterial immunity against K. pneumoniae and suggest new mechanisms of orchestrating lung antibacterial immunity.


Asunto(s)
Quimiocina CCL8/inmunología , Hipersensibilidad/inmunología , Inflamación/inmunología , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Pulmón/inmunología , Neutrófilos/inmunología , Animales , Eosinófilos/inmunología , Eosinófilos/microbiología , Femenino , Hipersensibilidad/microbiología , Inflamación/microbiología , Interleucinas/inmunología , Infecciones por Klebsiella/microbiología , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Neutrófilos/microbiología , Ovalbúmina/inmunología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología
18.
Cancer Discov ; 3(11): 1286-301, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23921231

RESUMEN

UNLABELLED: Synovial sarcoma is an aggressive soft-tissue malignancy of children and young adults, with no effective systemic therapies. Its specific oncogene, SYT-SSX (SS18-SSX), drives sarcoma initiation and development. The exact mechanism of SYT-SSX oncogenic function remains unknown. In an SYT-SSX2 transgenic model, we show that a constitutive Wnt/ß-catenin signal is aberrantly activated by SYT-SSX2, and inhibition of Wnt signaling through the genetic loss of ß-catenin blocks synovial sarcoma tumor formation. In a combination of cell-based and synovial sarcoma tumor xenograft models, we show that inhibition of the Wnt cascade through coreceptor blockade and the use of small-molecule CK1α activators arrests synovial sarcoma tumor growth. We find that upregulation of the Wnt/ß-catenin cascade by SYT-SSX2 correlates with its nuclear reprogramming function. These studies reveal the central role of Wnt/ß-catenin signaling in SYT-SSX2-induced sarcoma genesis, and open new venues for the development of effective synovial sarcoma curative agents. SIGNIFICANCE: Synovial sarcoma is an aggressive soft-tissue cancer that afflicts children and young adults, and for which there is no effective treatment. The current studies provide critical insight into our understanding of the pathogenesis of SYT­SSX-dependent synovial sarcoma and pave the way for the development of effective therapeutic agents for the treatment of the disease in humans.


Asunto(s)
Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Sarcoma Sinovial/genética , Sarcoma Sinovial/patología , Vía de Señalización Wnt/efectos de los fármacos , Adolescente , Adulto , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Compuestos de Pirvinio/farmacología , Sarcoma Experimental , Sarcoma Sinovial/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
19.
Am J Respir Cell Mol Biol ; 49(3): 396-402, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23590311

RESUMEN

Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, a multienzyme complex, is the major source for production of reactive oxygen species (ROS). ROS are increased in allergic diseases, such as asthma, but the role of ROS in disease pathogenesis remains uncertain. We hypothesized that mice unable to generate ROS via the NADPH oxidase pathway would have decreased allergic airway inflammation. To test this hypothesis, we studied gp91phox(-/-) mice in a model of allergic airway inflammation after sensitization and challenge with ovalbumin. Serum, bronchoalveolar lavage fluid, and lungs were then examined for evidence of allergic inflammation. We found that mice lacking a functional NADPH oxidase complex had significantly decreased ROS production and allergic airway inflammation, compared with wild-type (WT) control animals. To determine the mechanism by which allergic inflammation was inhibited by gp91phox deficiency, we cultured bone marrow-derived dendritic cells from WT and gp91phox(-/-) mice and activated them with LPS. IL-12 expression was significantly increased in the gp91phox(-/-) bone marrow-derived dendritic cells, suggesting that the cytokine profile produced in the absence of gp91phox enhanced the conditions leading to T helper (Th) type 1 differentiation, while inhibiting Th2 polarization. Splenocytes from sensitized gp91phox(-/-) animals produced significantly less IL-13 in response to ovalbumin challenge in vitro compared with splenocytes from sensitized WT mice, suggesting that NADPH oxidase promotes allergic sensitization. In contrast, inflammatory cytokines produced by T cells cultured from WT and gp91phox(-/-) mice under Th0, Th1, Th2, and Th17 conditions were not significantly different. This study demonstrates the importance of NADPH oxidase activity and ROS production in a murine model of asthma.


Asunto(s)
Asma/inmunología , Interleucina-12/inmunología , Interleucina-13/inmunología , Pulmón/inmunología , Glicoproteínas de Membrana/inmunología , NADPH Oxidasas/inmunología , Especies Reactivas de Oxígeno/inmunología , Animales , Asma/inducido químicamente , Asma/genética , Asma/patología , Líquido del Lavado Bronquioalveolar/química , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Eliminación de Gen , Interleucina-12/biosíntesis , Interleucina-13/biosíntesis , Lipopolisacáridos/farmacología , Pulmón/metabolismo , Pulmón/patología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , NADPH Oxidasa 2 , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células TH1/inmunología , Células TH1/patología , Balance Th1 - Th2 , Células Th17/inmunología , Células Th17/patología , Células Th2/inmunología , Células Th2/patología
20.
Am J Respir Cell Mol Biol ; 49(2): 180-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23492192

RESUMEN

Although the antibody-based recognition of cell-surface markers has been widely used for the identification of immune cells, overlap in the expression of markers by different cell types and the inconsistent use of antibody panels have resulted in a lack of clearly defined signatures for myeloid cell subsets. We developed a 10-fluorochrome flow cytometry panel for the identification and quantitation of myeloid cells in the lungs, including pulmonary monocytes, myeloid dendritic cells, alveolar and interstitial macrophages, and neutrophils. After the initial sorting of viable CD45(+) leukocytes, we detected three leukocyte subpopulations based on CD68 expression: CD68(-), CD68(low), and CD68(hi). Further characterization of the CD68(hi) population revealed CD45(+)/CD68(hi)/F4/80(+)/CD11b(-)/CD11c(+)/Gr1(-) alveolar macrophages and CD45(+)/CD68(hi)/F4/80(-)/CD11c(+)/Gr1(-)/CD103(+)/major histocompatibility complex (MHC) class II(hi) dendritic cells. The CD68(low) population contained primarily CD45(+)/CD68(low)/F4/80(+)/CD11b(+)/CD11c(+)/Gr1(-)/CD14(low) interstitial macrophages and CD45(+)/CD68(low)/F4/80(+)/CD11b(+)/CD11c(-)/Gr1(low)/CD14(hi) monocytes, whereas the CD68(-) population contained neutrophils (CD45(+)/CD68(-)/F4/80(-)/CD11b(+)/Gr1(hi)). The validity of cellular signatures was confirmed by a morphological analysis of FACS-sorted cells, functional studies, and the depletion of specific macrophage subpopulations using liposomal clodronate. We believe our approach provides an accurate and reproducible method for the isolation, quantification, and characterization of myeloid cell subsets in the lungs, which may be useful for studying the roles of myeloid cells during various pathological processes.


Asunto(s)
Células Dendríticas/citología , Citometría de Flujo , Pulmón/citología , Macrófagos Alveolares/citología , Monocitos/citología , Animales , Conservadores de la Densidad Ósea/farmacología , Ácido Clodrónico/farmacología , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Pulmón/metabolismo , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/fisiología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Transgénicos , Monocitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA