Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Immunol ; 196(8): 3227-31, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26927798

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a devastating disease with no effective therapies. We investigated the role of the C-type lectin receptor, CLEC5A, in macrophage activation and pulmonary pathogenesis in a mouse model of COPD. We demonstrate that CLEC5A is expressed on alveolar macrophages in mice exposed long-term to cigarette smoke (CS), as well as in human smokers. We also show that CLEC5A-mediated activation of macrophages enhanced cytokine elaboration alone, as well as in combination with LPS or GM-CSF in CS-exposed mice. Furthermore, usingClec5a-deficient mice, we demonstrate that CS-induced macrophage responsiveness is mediated by CLEC5A, and CLEC5A is required for the development of inflammation, proinflammatory cytokine expression, and airspace enlargement. These findings suggest a novel mechanism that promotes airway inflammation and pathologies in response to CS exposure and identifies CLEC5A as a novel target for the therapeutic control of COPD pathogenesis.


Asunto(s)
Lectinas Tipo C/inmunología , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Receptores de Superficie Celular/inmunología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Líquido del Lavado Bronquioalveolar/citología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inflamación/inmunología , Lectinas Tipo C/genética , Lipopolisacáridos/efectos adversos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad Pulmonar Obstructiva Crónica/patología , Receptores de Superficie Celular/genética
2.
J Immunol ; 190(3): 1331-40, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23264660

RESUMEN

Cigarette smoke (CS) exposure is the primary risk factor for the development of chronic obstructive pulmonary disease (COPD). COPD is characterized by chronic peribronchial, perivascular, and alveolar inflammation. The inflammatory cells consist primarily of macrophage, neutrophils, and lymphocytes. Although myeloid cells are well studied, the role of lymphocyte populations in pathogenesis of COPD remains unclear. Using a mouse model of CS-induced emphysema, our laboratory has previously demonstrated that CS exposure causes changes in the TCR repertoire suggestive of an Ag-specific response and triggers a pathogenic T cell response sufficient to cause alveolar destruction and inflammation. We extend these findings to demonstrate that T cells from CS-exposed mice of the BALB/cJ or C57B6 strain are sufficient to transfer pulmonary pathology to CS-naive, immunosufficient mice. CS exposure causes a proinflammatory phenotype among pulmonary T cells consistent with those from COPD patients. We provide evidence that donor T cells from CS-exposed mice depend on Ag recognition to transfer alveolar destruction using MHC class I-deficient recipient mice. Neither CD4(+) nor CD8(+) T cells from donor mice exposed to CS alone are sufficient to cause inflammation or pathology in recipient mice. We found no evidence of impaired suppression of T cell proliferation among regulatory T cells from CS-exposed mice. These results suggest that CS exposure initiates an Ag-specific response that leads to pulmonary destruction and inflammation that involves both CD8(+) and CD4(+) T cells. These results are direct evidence for an autoimmune response initiated by CS exposure.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Subgrupos de Linfocitos T/inmunología , Contaminación por Humo de Tabaco/efectos adversos , Traslado Adoptivo , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Femenino , Adyuvante de Freund , Reordenamiento Génico de Linfocito T , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/patología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/patología , Especificidad del Receptor de Antígeno de Linfocitos T , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células TH1/inmunología , Células TH1/patología , Células Th17/inmunología , Células Th17/patología
3.
J Immunol ; 188(9): 4468-75, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22467655

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by peribronchial and perivascular inflammation and largely irreversible airflow obstruction. Acute disease exacerbations, due frequently to viral infections, lead to enhanced disease symptoms and contribute to long-term progression of COPD pathology. Previously, we demonstrated that NK cells from cigarette smoke (CS)-exposed mice exhibit enhanced effector functions in response to stimulating cytokines or TLR ligands. In this article, we show that the activating receptor NKG2D is a key mediator for CS-stimulated NK cell hyperresponsiveness, because CS-exposed NKG2D-deficient mice (Klrk1(-/-)) did not exhibit enhanced effector functions as assessed by cytokine responsiveness. NK cell cytotoxicity against MHC class I-deficient targets was not affected in a COPD model. However, NK cells from CS-exposed mice exhibit greater cytotoxic activity toward cells that express the NKG2D ligand RAET1ε. We also demonstrate that NKG2D-deficient mice exhibit diminished airway damage and reduced inflammation in a model of viral COPD exacerbation, which do not affect viral clearance. Furthermore, adoptive transfer of NKG2D(+) NK cells into CS-exposed, influenza-infected NKG2D-deficient mice recapitulated the phenotypes observed in CS-exposed, influenza-infected wild-type mice. Our findings indicate that NKG2D stimulation during long-term CS exposure is a central pathway in the development of NK cell hyperresponsiveness and influenza-mediated exacerbations of COPD.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/inmunología , Células Asesinas Naturales/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Infecciones por Orthomyxoviridae/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Células Asesinas Naturales/patología , Ratones , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/virología
4.
J Immunol ; 184(8): 4460-9, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20228194

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a debilitating, progressive lung disease punctuated by exacerbations of symptoms. COPD exacerbations are most often associated with viral infections, and exposure to cigarette smoke (CS) followed by viral infection has been shown experimentally to enhance lung inflammation, tissue destruction, and airway fibrosis. Despite this, however, the cellular mechanisms responsible for this effect are unknown. In this study, we examined NK cell function in a mouse model of COPD given the vital role of NK cells following viral infection. Ex vivo stimulation of lung leukocytes with poly(I:C), ssRNA40, or ODN1826 enhanced production of NK cell-derived IFN-gamma in CS-exposed mice. NK cells from CS-exposed mice exhibited a novel form of priming; highly purified NK cells from CS-exposed mice, relative to NK cells from filtered air-exposed mice, produced more IFN-gamma following stimulation with IL-12, IL-18, or both. Further, NK cell priming was lost following smoking cessation. NKG2D stimulation through overexpression of Raet1 on the lung epithelium primed NK cell responsiveness to poly(I:C), ssRNA40, or ODN1826 stimulation, but not cytokine stimulation. In addition, NK cells from CS-exposed mice expressed more cell surface CD107a upon stimulation, demonstrating that the NK cell degranulation response was also primed. Together, these results reveal a novel mechanism of activation of the innate immune system and highlight NK cells as important cellular targets in controlling COPD exacerbations.


Asunto(s)
Mediadores de Inflamación/toxicidad , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Células Cultivadas , Técnicas de Cocultivo , ADN/toxicidad , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/farmacología , Interferón gamma/biosíntesis , Células Asesinas Naturales/virología , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodesoxirribonucleótidos , Poli I-C/toxicidad , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/virología , ARN Viral/toxicidad , Regulación hacia Arriba/inmunología
5.
Am J Respir Crit Care Med ; 181(11): 1223-33, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20133926

RESUMEN

RATIONALE: Pathogenic T cells drive, or sustain, a number of inflammatory diseases. Chronic obstructive pulmonary disease (COPD) is an inflammatory lung disease associated with the accumulation of activated T cells. We previously demonstrated that chronic cigarette smoke (CS) exposure causes oligoclonal expansion of lung CD4(+) T cells and CD8(+) T cells in a mouse model of COPD, thus implicating these cells in disease pathogenesis. OBJECTIVES: To determine whether T cells are pathogenic in a CS-induced mouse model of COPD. METHODS: We transferred lung CD3(+) T cells from filtered air (FA)- and CS-exposed mice into Rag2(-/-) recipients. Endpoints associated with the COPD phenotype were then measured. MEASUREMENTS AND MAIN RESULTS: Here, we demonstrate that chronic CS exposure generates pathogenic T cells. Transfer of CD3(+) T cells from the lungs of CS-exposed mice into Rag2(-/-) recipients led to substantial pulmonary changes pathognomonic of COPD. These changes included monocyte/macrophage and neutrophil accumulation, increased expression of cytokines and chemokines, activation of proteases, apoptosis of alveolar epithelial cells, matrix degradation, and airspace enlargement reminiscent of emphysema. CONCLUSIONS: These data formally demonstrate, for the first time, that chronic CS exposure leads to the generation of pathogenic T cells capable of inducing COPD-like disease in Rag2(-/-) mice. This report provides novel insights into COPD pathogenesis.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica/inmunología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Apoptosis , Líquido del Lavado Bronquioalveolar/citología , Complejo CD3/inmunología , Catepsinas/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Leucocitos/metabolismo , Pulmón/patología , Macrófagos/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Alveolos Pulmonares/patología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/patología , Linfocitos T/metabolismo
6.
Infect Immun ; 78(5): 2099-107, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20176793

RESUMEN

CCR7 is a chemokine receptor expressed on the surfaces of T cells, B cells, and mature dendritic cells that controls cell migration in response to the cognate ligands CCL19 and CCL21. CCR7 is critical for the generation of an adaptive T cell response. However, the roles of CCR7 in the host defense against pulmonary infection and innate immunity are not well understood. We investigated the role of CCR7 in the host defense against acute pulmonary infection with Pseudomonas aeruginosa. We intranasally infected C57BL/6 mice with P. aeruginosa and characterized the expression of CCR7 ligands and the surface expression of CCR7 on pulmonary leukocytes. In response to infection, expression of CCL19 and expression of CCL21 were oppositely regulated, and myeloid dendritic cells upregulated CCR7 expression. We further examined the effects of CCR7 deficiency on the inflammatory response to P. aeruginosa infection. We infected Ccr7(-/-) and wild-type mice with P. aeruginosa and characterized the accumulation of pulmonary leukocytes, production of proinflammatory mediators, neutrophil activation, and bacterial clearance. CCR7 deficiency led to an accumulation of myeloid dendritic cells and T cells in the lung in response to infection. CCR7 deficiency resulted in higher expression of CD80 and CD86 on dendritic cells; increased production of interleukin-12/23p40 (IL-12/23p40), gamma interferon (IFN-gamma), and IL-1 alpha; increased neutrophil respiratory burst; and, ultimately, increased clearance of acute P. aeruginosa infection. In conclusion, our results suggest that CCR7 deficiency results in a heightened proinflammatory environment in response to acute pulmonary P. aeruginosa infection and contributes to more efficient clearance.


Asunto(s)
Leucocitos/inmunología , Neumonía Bacteriana/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/inmunología , Receptores CCR7/inmunología , Animales , Quimiocina CCL19/biosíntesis , Quimiocina CCL21/biosíntesis , Recuento de Colonia Microbiana , Perfilación de la Expresión Génica , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/microbiología , Infecciones por Pseudomonas/patología , Receptores CCR7/biosíntesis , Receptores CCR7/deficiencia
7.
J Immunol ; 181(8): 5481-9, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18832705

RESUMEN

Pseudomonas aeruginosa is a major cause of nosocomial respiratory infections. The eradication of P. aeruginosa from the lung involves the orchestrated actions of the pulmonary epithelium and both resident and recruited immune cells. The NKG2D receptor is constitutively expressed on the surface of circulating and tissue-resident NK cells (and other cytotoxic lymphocytes), and is capable of controlling NK cell activation and production of cytokines, such as IFN-gamma via interactions with ligands expressed on the surface of stressed cells. Previously, we demonstrated that NKG2D mediates pulmonary clearance of P. aeruginosa. In the present study, we investigated the cellular and molecular mechanisms of NKG2D-mediated clearance of P. aeruginosa using a novel transgenic mouse model of doxycycline-inducible conditional expression of NKG2D ligands (retinoic acid early transcript 1, alpha) in pulmonary epithelial cells. NKG2D ligand expression in this model increased pulmonary clearance, cellular phagocytosis, and survival following P. aeruginosa respiratory infection. Additionally, NK cell sensitivity to ex vivo LPS stimulation was greater in lung cells isolated from naive transgenic mice administered doxycycline. We also showed that NK cells are the primary source of lymphocyte-derived IFN-gamma in response to P. aeruginosa respiratory infection. Significantly, we demonstrated that NKG2D is critical to the nonredundant IFN-gamma production by pulmonary NK cells following acute P. aeruginosa infection. These results represent the principal report of NKG2D-mediated activation of lung NK cells following respiratory infection with an opportunistic pathogen and further establish the importance of NKG2D in the host response against P. aeruginosa respiratory infection.


Asunto(s)
Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Receptores Inmunológicos/inmunología , Infecciones del Sistema Respiratorio/inmunología , Animales , Expresión Génica/genética , Expresión Génica/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Lipopolisacáridos/farmacología , Pulmón/inmunología , Activación de Linfocitos/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK , Infecciones Oportunistas/genética , Infecciones Oportunistas/inmunología , Infecciones por Pseudomonas/genética , Receptores Inmunológicos/genética , Receptores de Células Asesinas Naturales , Mucosa Respiratoria/inmunología , Infecciones del Sistema Respiratorio/genética
8.
J Immunol ; 181(11): 8036-43, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19017996

RESUMEN

The role of adaptive immunity in the development or progression of chronic obstructive pulmonary disease (COPD) remains undefined. Recently, the presence of autoantibodies and autoreactive T cells has been demonstrated in COPD patients. In addition, oligoclonal expansions of lung T cells have been observed in COPD patients, but the overlapping incidence of infections, tumors, and cigarette smoke exposure obscures the antigenic stimulus. We analyzed the TCR Vbeta repertoire of CD4 and CD8 T cells purified from the lungs and spleens of mice chronically exposed to cigarette smoke. In a mouse model of COPD, we demonstrate that chronic cigarette smoke exposure causes oligoclonal expansions of T cells isolated from the lungs, but not spleens. TCR Vbeta repertoire analyses revealed oligoclonal expansions predominantly occurred in lung CD8 T cells, with preferential usage of Vbeta7, Vbeta9, Vbeta13, and Vbeta14. Using nucleotide sequence analysis based on Jbeta analyses, we demonstrate selection of CDR3 amino acid motifs, which strongly suggests Ag-driven oligoclonal T cell expansion. Analysis of the lung TCR Vbeta repertoire of mice with cigarette smoke-induced emphysema, which had undergone smoking cessation for 6 mo, revealed that oligoclonal expansions persisted. This study formally demonstrates that chronic cigarette smoke exposure, alone, causes a persistent adaptive T cell immune response. These findings have important implications for therapeutic approaches in the treatment of COPD, and provide insight into potential mechanisms involved in disease pathogenesis.


Asunto(s)
Enfermedades Autoinmunes/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfisema Pulmonar/genética , Fumar/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Secuencias de Aminoácidos/genética , Secuencias de Aminoácidos/inmunología , Animales , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/inmunología , Modelos Animales de Enfermedad , Femenino , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/inmunología , Humanos , Pulmón/inmunología , Ratones , Ratones Endogámicos BALB C , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/inmunología
9.
Neurotoxicol Teratol ; 51: 68-76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26340819

RESUMEN

Use of silver nanoparticles (AgNPs) for their antimicrobial properties is widespread. Much of the previous work on the toxicity of AgNPs has been conducted in vitro or following oral or intravenous administration in vivo. Intranasal (IN) instillation of AgNPs mimics inhalation exposure and allows further exploration of the toxicity of these particles via respiratory tract exposure. The present study involved 1) single-dose exposures to assess tissue distribution and toxicity and 2) repeated exposures to assess behavioral effects of IN AgNP exposure (nominally uncoated 25 nm AgNP). AgNP deposition was localized in the liver, gut-associated lymphoid tissue, and brain. Decrease cellularity in spleen follicles was observed in treated mice, along with changes in cell number and populations in the spleen. The splenic GSH:GSSG ratio was also reduced following AgNP exposure. Expression of the oxidative stress-responsive gene Hmox1 was elevated in the hippocampus, but not cortex of treated mice, as was the level of HMOX1 protein. Mice receiving 7 days of IN exposure to 50 mg/kg AgNPs exhibited similar learning- and memory-related behaviors to control mice, except that treated mice spent significantly less time in the target quadrant of the Morris Water Maze during the acquisition phase probe trial. These findings indicate systemic distribution and toxicity following IN administration of AgNPs.


Asunto(s)
Conducta Animal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Reconocimiento en Psicología/efectos de los fármacos , Compuestos de Plata/toxicidad , Administración Intranasal , Animales , Diferenciación Celular/efectos de los fármacos , Plexo Coroideo/efectos de los fármacos , Plexo Coroideo/metabolismo , Relación Dosis-Respuesta a Droga , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Reconocimiento en Psicología/fisiología , Bazo/citología , Bazo/efectos de los fármacos , Distribución Tisular/efectos de los fármacos
10.
PLoS One ; 8(10): e78735, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130907

RESUMEN

Long-term exposure to cigarette smoke (CS) can have deleterious effects on lung epithelial cells including cell death and the initiation of inflammatory responses. CS-induced cell injury can elaborate cell surface signals and cellular byproducts that stimulate immune system surveillance. Our previous work has shown that the expression of ligands for the cytotoxic lymphocyte activating receptor NKG2D is enhanced in patients with COPD and that the induction of these ligands in a mouse model can replicate COPD pathologies. Here, we extend these findings to demonstrate a role for the NKG2D receptor in CS-induced pathophysiology and provide evidence linking nucleic acid-sensing endosomal toll-like receptor (TLR) signaling to COPD pathology through NKG2D activation. Specifically, we show that mice deficient in NKG2D exhibit attenuated pulmonary inflammation and airspace enlargement in a model of CS-induced emphysema. Additionally, we show that CS exposure induces the release of free nucleic acids in the bronchoalveolar lavage and that direct exposure of mouse lung epithelial cells to cigarette smoke extract similarly induces functional nucleic acids as assessed by TLR3, 7, and 9 reporter cell lines. We demonstrate that exposure of mouse lung epithelial cells to TLR ligands stimulates the surface expression of RAET1, a ligand for NKG2D, and that mice deficient in TLR3/7/9 receptor signaling do not exhibit CS-induced NK cell hyperresponsiveness and airspace enlargement. The findings indicate that CS-induced airway injury stimulates TLR signaling by endogenous nucleic acids leading to elevated NKG2D ligand expression. Activation of these pathways plays a major role in the altered NK cell function, pulmonary inflammation and remodeling related to long-term CS exposure.


Asunto(s)
Pulmón/patología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Nicotiana/efectos adversos , Neumonía/etiología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Humo/efectos adversos , Receptores Toll-Like/inmunología , Animales , Línea Celular , Femenino , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Neumonía/inmunología , Neumonía/patología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología
11.
PLoS One ; 7(5): e36055, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22563477

RESUMEN

Previously this laboratory characterized Slc39a8-encoded ZIP8 as a Zn(2+)/(HCO(3)(-))(2) symporter; yet, the overall physiological importance of ZIP8 at the whole-organism level remains unclear. Herein we describe the phenotype of the hypomorphic Slc39a8(neo/neo) mouse which has retained the neomycin-resistance gene in intron 3, hence causing significantly decreased ZIP8 mRNA and protein levels in embryo, fetus, placenta, yolk sac, and several tissues of neonates. The Slc39a8(neo) allele is associated with diminished zinc and iron uptake in mouse fetal fibroblast and liver-derived cultures; consequently, Slc39a8(neo/neo) newborns exhibit diminished zinc and iron levels in several tissues. Slc39a8(neo/neo) homozygotes from gestational day(GD)-11.5 onward are pale, growth-stunted, and die between GD18.5 and 48 h postnatally. Defects include: severely hypoplastic spleen; hypoplasia of liver, kidney, lung, and lower limbs. Histologically, Slc39a8(neo/neo) neonates show decreased numbers of hematopoietic islands in yolk sac and liver. Low hemoglobin, hematocrit, red cell count, serum iron, and total iron-binding capacity confirmed severe anemia. Flow cytometry of fetal liver cells revealed the erythroid series strikingly affected in the hypomorph. Zinc-dependent 5-aminolevulinic acid dehydratase, required for heme synthesis, was not different between Slc39a8(+/+) and Slc39a8(neo/neo) offspring. To demonstrate further that the mouse phenotype is due to ZIP8 deficiency, we bred Slc39a8(+/neo) with BAC-transgenic BTZIP8-3 line (carrying three extra copies of the Slc39a8 allele); this cross generated viable Slc39a8(neo/neo)_BTZIP8-3(+/+) pups showing none of the above-mentioned congenital defects-proving Slc39a8(neo/neo) causes the described phenotype. Our study demonstrates that ZIP8-mediated zinc transport plays an unappreciated critical role during in utero and neonatal growth, organ morphogenesis, and hematopoiesis.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Embrión de Mamíferos/embriología , Hematopoyesis/fisiología , Organogénesis/fisiología , Animales , Animales Recién Nacidos , Transporte Biológico , Western Blotting , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Hígado/citología , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Organogénesis/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Saco Vitelino/embriología , Saco Vitelino/metabolismo , Zinc/metabolismo
12.
J Clin Invest ; 119(3): 636-49, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19197141

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a lethal progressive lung disease culminating in permanent airway obstruction and alveolar enlargement. Previous studies suggest CTL involvement in COPD progression; however, their precise role remains unknown. Here, we investigated whether the CTL activation receptor NK cell group 2D (NKG2D) contributes to the development of COPD. Using primary murine lung epithelium isolated from mice chronically exposed to cigarette smoke and cultured epithelial cells exposed to cigarette smoke extract in vitro, we demonstrated induced expression of the NKG2D ligand retinoic acid early transcript 1 (RAET1) as well as NKG2D-mediated cytotoxicity. Furthermore, a genetic model of inducible RAET1 expression on mouse pulmonary epithelial cells yielded a severe emphysematous phenotype characterized by epithelial apoptosis and increased CTL activation, which was reversed by blocking NKG2D activation. We also assessed whether NKG2D ligand expression corresponded with pulmonary disease in human patients by staining airway and peripheral lung tissues from never smokers, smokers with normal lung function, and current and former smokers with COPD. NKG2D ligand expression was independent of NKG2D receptor expression in COPD patients, demonstrating that ligand expression is the limiting factor in CTL activation. These results demonstrate that aberrant, persistent NKG2D ligand expression in the pulmonary epithelium contributes to the development of COPD pathologies.


Asunto(s)
Pulmón/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Mucosa Respiratoria/fisiopatología , Humo/efectos adversos , Fumar/efectos adversos , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Enfisema/etiología , Enfisema/inmunología , Regulación de la Expresión Génica , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Proteínas de la Membrana/genética , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología
13.
Toxicol Sci ; 105(1): 188-99, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18515264

RESUMEN

Acrolein exposure represents a significant human health hazard. Repeated acrolein exposure causes the accumulation of monocytes/macrophages and lymphocytes, mucous cell metaplasia, and epithelial injury. Currently, the mechanisms that control these events are unclear, and the relative contribution of T-cell subsets to pulmonary pathologies following repeated exposures to irritants is unknown. To examine whether lymphocyte subpopulations regulate inflammation and epithelial cell pathology, we utilized a mouse model of pulmonary pathology induced by repeated acrolein exposures. The role of lymphocyte subsets was examined by utilizing transgenic mice genetically deficient in either alphabeta T cells or gammadelta T cells, and changes in cellular, molecular, and pathologic outcomes associated with repeated inhalation exposure to 2.0 and 0.5 ppm acrolein were measured. To examine the potential functions of lymphocyte subsets, we purified these cells from the lungs of mice repeatedly exposed to 2.0 ppm acrolein, isolated and amplified messenger RNA, and performed microarray analysis. Our data demonstrate that alphabeta T cells are required for macrophage accumulation, whereas gammadelta T cells are critical regulators of epithelial cell homeostasis, as identified by epithelial cell injury and apoptosis, following repeated acrolein exposure. This is supported by microarray analyses that indicated the T-cell subsets are unique in their gene expression profiles following acrolein exposures. Microarray analyses identified several genes that may contribute to phenotypes mediated by T-cell subpopulations including those involved in cytokine receptor signaling, chemotaxis, growth factor production, lymphocyte activation, and apoptosis. These data provide strong evidence that T-cell subpopulations in the lung are major determinants of pulmonary pathology and highlight the advantages of dissecting their effector functions in response to toxicant exposures.


Asunto(s)
Acroleína/toxicidad , Pulmón/efectos de los fármacos , Receptores de Antígenos de Linfocitos T alfa-beta/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/fisiología , Linfocitos T/fisiología , Animales , Apoptosis/efectos de los fármacos , Separación Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Perfilación de la Expresión Génica , Interleucina-18/fisiología , Pulmón/inmunología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Mucina 5AC , Mucinas/análisis , Análisis de Secuencia por Matrices de Oligonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA