Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur Respir J ; 31(1): 43-6, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18166592

RESUMEN

As excess mucin expression can contribute to the exacerbation of asthma, the present authors hypothesised that Mycoplasma pneumoniae significantly induces MUC5AC (the major airway mucin) expression in airway epithelial cells isolated directly from asthmatic subjects. A total of 11 subjects with asthma and six normal controls underwent bronchoscopy with airway brushing. Epithelial cells were cultured at an air-liquid interface and incubated with and without M. pneumoniae for 48 h, and in the presence and absence of nuclear factor (NF)-kappaB and a toll-like receptor (TLR)2 inhibitor. Quantitative PCR was performed for MUC5AC and TLR2 mRNA. MUC5AC protein and total protein were determined by ELISA. M. pneumoniae exposure significantly increased MUC5AC mRNA and protein expression after 48 h in epithelial cells isolated from asthmatic, but not from normal control subjects, at all concentrations as compared to unexposed cells. TLR2 mRNA expression was significantly increased in asthmatic epithelial cells at 4 h compared with unexposed cells. NF-kappaB and TLR2 inhibition reduced MUC5AC expression to the level of the unexposed control in both groups. Mycoplasma pneumoniae exposure significantly increased MUC5AC mRNA and protein expression preferentially in airway epithelial cells isolated from asthmatic subjects. The toll-like receptor 2 pathway may be involved in this process.


Asunto(s)
Asma/metabolismo , Células Epiteliales/microbiología , Regulación de la Expresión Génica , Mucinas/biosíntesis , Mycoplasma pneumoniae/metabolismo , Adulto , Asma/inmunología , Asma/microbiología , Broncoscopía/métodos , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/citología , Femenino , Humanos , Masculino , Modelos Biológicos , Mucina 5AC , Mucinas/metabolismo , Mycoplasma pneumoniae/fisiología , FN-kappa B/metabolismo , Factores de Tiempo , Receptor Toll-Like 2/metabolismo
2.
Sci Rep ; 8(1): 13278, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30185885

RESUMEN

Through the process of neuronal differentiation, newly born neurons change from simple, spherical cells to complex, sprawling cells with many highly branched processes. One of the first stages in this process is neurite initiation, wherein cytoskeletal modifications facilitate membrane protrusion and extension from the cell body. Hundreds of actin modulators and microtubule-binding proteins are known to be involved in this process, but relatively little is known about how upstream regulators bring these complex networks together at discrete locations to produce neurites. Here, we show that Myristoylated alanine-rich C kinase substrate (MARCKS) participates in this process. Marcks-/- cortical neurons extend fewer neurites and have less complex neurite arborization patterns. We use an in vitro proteomics screen to identify MARCKS interactors in developing neurites and characterize an interaction between MARCKS and a CDC42-centered network. While the presence of MARCKS does not affect whole brain levels of activated or total CDC42, we propose that MARCKS is uniquely positioned to regulate CDC42 localization and interactions within specialized cellular compartments, such as nascent neurites.


Asunto(s)
Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/metabolismo , Neuritas/metabolismo , Neuronas/citología , Actinas/metabolismo , Animales , Citoesqueleto/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Neuronas/metabolismo , Fosforilación , Cultivo Primario de Células , Unión Proteica , Seudópodos/metabolismo , Transducción de Señal , Proteína de Unión al GTP cdc42/metabolismo
3.
J Clin Invest ; 85(1): 75-85, 1990 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2153154

RESUMEN

Several common pulmonary disorders characterized by mucus hypersecretion and airway obstruction may relate to increased levels of inhaled or endogenously generated oxidants (O2 metabolites) in the respiratory tract. We found that O2 metabolites stimulated release of high-molecular-weight glycoconjugates (HMG) by respiratory epithelial cells in vitro through a mechanism involving cyclooxygenase metabolism of arachidonic acid. Noncytolytic concentrations of chemically generated O2 metabolites (purine + xanthine oxidase) stimulated HMG release by cell and explant cultures of rodent airway epithelium, an effect which is inhibitable by coaddition of specific O2 metabolite scavengers or inhibitors of arachidonic acid metabolism. Addition of O2 metabolites to epithelial cells provoked production of PGF2a, an effect also inhibitable by coaddition of O2 metabolite scavengers or inhibitors of arachidonic acid metabolism. Finally, addition of exogenous PGF2a to cell cultures stimulated HMG release. We conclude that O2 metabolites increase release of respiratory HMG through a mechanism involving cyclooxygenase metabolism of arachidonic acid with production mainly of PGF2a. This mechanism may be fundamental to the pathogenesis of a variety of lung diseases associated with hypersecretion of mucus and/or other epithelial fluids, as well as a basic cellular response to increased oxidants.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Glicoconjugados/metabolismo , Peróxido de Hidrógeno/farmacología , Superóxidos/farmacología , Tráquea/metabolismo , Animales , Ácidos Araquidónicos/aislamiento & purificación , Catalasa/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/ultraestructura , Cobayas , Cinética , L-Lactato Deshidrogenasa/metabolismo , Masculino , Manitol/farmacología , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Técnicas de Cultivo de Órganos , Polietilenglicoles/farmacología , Conejos , Superóxido Dismutasa/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología , Tráquea/citología , Xantina Oxidasa/metabolismo
4.
J Appl Physiol (1985) ; 102(1): 399-405, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16946028

RESUMEN

Allergic asthma is associated with airway epithelial cell mucous metaplasia and mucin hypersecretion, but the consequences of mucin hypersecretion on airway function are unclear. Recently, a peptide derived from the myristoylated alanine-rich C kinase substrate protein NH(2)-terminal sequence (MANS) was shown to inhibit methacholine (MCh)-induced mucin secretion from airway mucous cells by >90%. We studied the effect of intranasal pretreatment with this peptide on specific airway conductance (sGaw) during challenge with MCh in mice with allergen-induced mucous cell metaplasia. sGaw was noninvasively measured in spontaneously breathing restrained mice, using a double-chamber plethysmograph. Pretreatment with MANS peptide, but not a control peptide [random NH(2)-terminal sequence (RNS)], resulted in partial inhibition of the fall in sGaw induced by 60 mM MCh (mean +/- SE; baseline 1.15 +/- 0.06; MANS/MCh 0.82 +/- 0.05; RNS/MCh 0.55 +/- 0.05 cmH(2)O/s). The protective effect of MANS was also seen in mice challenged with allergen for 3 consecutive days to increase airway hyperresponsiveness, although the degree of protection was less (baseline 1.1 +/- 0.08; MANS/MCh, 0.65 +/- 0.06; RNS/MCh 0.47 +/- 0.03 cmH(2)O/s). Because routine sGaw measurement in mice includes nasal airways, the effectiveness of MANS was also confirmed in mice breathing through their mouths after nasal occlusion (baseline 0.92 +/- 0.05; MANS/MCh 0.83 +/- 0.06; RNS/MCh 0.61 +/- 0.03 cmH(2)O/s). In all instances, sGaw in the MANS-pretreated group was approximately 35% higher than in RNS-treated controls, and mucous obstruction accounted for approximately 50% of the MCh-induced fall in sGaw. In summary, mucin secretion has a significant role in airway obstruction in a mouse model of allergic asthma, and strategies to inhibit mucin secretion merit further investigation.


Asunto(s)
Obstrucción de las Vías Aéreas/metabolismo , Asma/metabolismo , Mucinas/metabolismo , Fragmentos de Péptidos/farmacología , Obstrucción de las Vías Aéreas/tratamiento farmacológico , Obstrucción de las Vías Aéreas/fisiopatología , Animales , Asma/tratamiento farmacológico , Asma/fisiopatología , Broncoconstricción/efectos de los fármacos , Broncoconstricción/fisiología , Broncoconstrictores/administración & dosificación , Modelos Animales de Enfermedad , Cloruro de Metacolina/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/uso terapéutico
5.
Cancer Res ; 40(12): 4403-9, 1980 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7192176

RESUMEN

In respiratory carcinogenesis studies using rodents, the tracheal epithelium is the target tissue for the induction of tumors after exposure of animals to chemical carcinogens. In the studies described below, tracheal epithelial cells were isolated to evaluate their biological and biochemical features. Epithelial cells derived from the tracheal mucosa of Syrian golden hamsters were established in culture. Three morphological types of polygonal cells were observed as mixed populations in four clonally derived lines. One type of cell is mucin secreting since membrane-bound vesicles that stain positively using the alcian blue:periodic acid-Schiff reaction are present in the cytoplasm and increased amounts of mucin constituents are demonstrable in the culture medium. Cells of a second type possess both intracytoplasmic and surface cilia, but they lack mucin vesicles. The third type exhibits no differentiating features. Four density-dependent inhibited cloned cell lines were established. After repeated passage, these cells: (a) grew in soft agar; (b) released proteases that were activators of plasminogen; (c) demonstrated measurable basal and inducible aryl hydrocarbon hydroxylase activity; and (d) produced anaplastic carcinomas in syngeneic hamsters. Factors affecting the transformation and differentiation of respiratory epithelial cells have not been elucidated. The availability of these cell lines will permit studies that focus on these questions.


Asunto(s)
Transformación Celular Neoplásica/patología , Tráquea/patología , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Línea Celular , Transformación Celular Neoplásica/metabolismo , Cricetinae , Epitelio/patología , Femenino , Glicoproteínas/metabolismo , Cariotipificación , Mesocricetus , Proteínas de Neoplasias/metabolismo , Activadores Plasminogénicos/metabolismo
6.
Free Radic Biol Med ; 24(5): 863-8, 1998 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9586818

RESUMEN

Intracellularly generated reactive species of both oxygen (ROS) and nitrogen (RNS) have been implicated in signaling responses in airway epithelial cells, but these radicals have not been measured directly in such cells. In this study, intracellular production of both ROS and RNS were measured in the same cell lysates of guinea pig tracheal epithelial (GPTE) cells maintained in primary culture. ROS and RNS were quantified under basal (constitutive) conditions and in response to different stimuli: LPS and TNFalpha [activators of inducible nitric oxide synthase (iNOS)]; several activators of calcium-dependent cNOS (ATP, bradykinin, ionophore A23187, and thapsigargin); and exogenous oxidant stress generated by addition of xanthine oxidase to purine (p + XO). Studies with LPS and TNFalpha also were performed using the murine macrophage cell line, RAW 264.7, as a positive control. Intracellular oxidant production was detected from oxidation of dihydrorhodamine to rhodamine. NOx was quantified by either chemiluminescent or fluorescent detection. NOS activity was measured as citrulline production from arginine. Basal production of oxidants by GPTE cells (0.08 + 0.00 nmol rhodamine) was less than 10% that of RAW.267 cells (0.91 + 0.03 nmol rhodamine). TNFalpha and LPS significantly increased intracellular oxidant production in GPTE cells, as did p + XO, but none of the cNOS activators affected production of oxidants in these cells. Concentrations of NO2 after 4 h in unstimulated RAW 264.7 and GPTE cells were similar and comprised 63% of total NOx in GPTE and 62% in RAW cells. TNFalpha and LPS both increased NO2 in GPTE cells, but none of the Ca++-mobilizing agents nor p + XO significantly affected intracellular RNS. The results suggest both ROS and RNS can be measured in the same lysates from airway epithelial cells, and that both ROS and RNS are produced in these cells in response to different stimuli.


Asunto(s)
Nitrógeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tráquea/metabolismo , Animales , Línea Celular , Activación Enzimática , Células Epiteliales/metabolismo , Radicales Libres , Cobayas , Macrófagos/metabolismo , Ratones , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Estrés Oxidativo/fisiología , Tráquea/citología
8.
Environ Health Perspect ; 102 Suppl 10: 85-90, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7705313

RESUMEN

Reactive oxygen species (ROS) have been implicated in the pathogenesis of numerous disease processes. Epithelial cells lining the respiratory airways are uniquely vulnerable regarding potential for oxidative damage due to their potential for exposure to both endogenous (e.g., mitochondrial respiration, phagocytic respiratory burst, cellular oxidases) and exogenous (e.g., air pollutants, xenobiotics, catalase negative organisms) oxidants. Airway epithelial cells use several nonenzymatic and enzymatic antioxidant mechanisms to protect against oxidative insult. Nonenzymatic defenses include certain vitamins and low molecular weight compounds such as thiols. The enzymes superoxide dismutase, catalase, and glutatione peroxidase are major sources of antioxidant protection. Other materials associated with airway epithelium such as mucus, epithelial lining fluid, and even the basement membrane/extracellular matrix may have protective actions as well. When the normal balance between oxidants and antioxidants is upset, oxidant stress ensues and subsequent epithelial cell alterations or damage may be a critical component in the pathogenesis of several respiratory diseases. Oxidant stress may profoundly alter lung physiology including pulmonary function (e.g., forced expiratory volumes, flow rates, and maximal inspiratory capacity), mucociliary activity, and airway reactivity. ROS may induce airway inflammation; the inflammatory process may serve as an additional source of ROS in airways and provoke the pathophysiologic responses described. On a more fundamental level, cellular mechanisms in the pathogenesis of ROS may involve activation of intracellular signaling enzymes including phospholipases and protein kinases stimulating the release of inflammatory lipids and cytokines. Respiratory epithelium may be intimately involved in defense against, and pathophysiologic changes invoked by, ROS.


Asunto(s)
Especies Reactivas de Oxígeno/farmacología , Sistema Respiratorio/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Epitelio/efectos de los fármacos , Epitelio/fisiopatología , Humanos , Oxidorreductasas/metabolismo , Sistema Respiratorio/fisiopatología
9.
Environ Health Perspect ; 106 Suppl 5: 1197-203, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9788898

RESUMEN

The body first encounters deleterious inhaled substances, such as allergens, industrial particles, pollutants, and infectious agents, at the airway epithelium. When this occurs, the epithelium and its resident inflammatory cells respond defensively by increasing production of cytokines, mucus, and reactive oxygen and nitrogen species (ROS/RNS). As inflammation in the airway increases, additional infiltrating cells increase the level of these products. Recent interest has focused on ROS/RNS as potential modulators of the expression of inflammation-associated genes important to the pathogenesis of various respiratory diseases. ROS/RNS appear to play a variety of roles that lead to changes in expression of genes such as interleukin-6 and intercellular adhesion molecule 1. By controlling this regulation, the reactive species can serve as exogenous stimuli, as intercellular signaling molecules, and as modulators of the redox state in epithelial cells. Unraveling the molecular mechanisms affected by ROS/RNS acting in these capacities should aid in the understanding of how stimulated defense mechanisms within the airway can lead to disease.


Asunto(s)
Nitrógeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sistema Respiratorio/metabolismo , Animales , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Inflamación/etiología , Inflamación/genética , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/genética , Interleucina-6/biosíntesis , Interleucina-6/genética , Oxidantes/toxicidad , Sistema Respiratorio/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
10.
Environ Health Perspect ; 105 Suppl 5: 1301-7, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9400742

RESUMEN

Epidemiologic and occupational studies indicate adverse health effects due to inhalation of particulate air pollutants, but precise biologic mechanisms responsible have yet to be fully established. The tracheobronchial epithelium forms the body's first physiologic barrier to such airborne pollutants, where ciliary movement functions to remove the offending substances caught in the overlying mucus layer. Resident and infiltrating phagocytic cells also function in this removal process. In this paper, we examine the role of reactive oxygen and nitrogen species (ROS/RNS) in the response of airway epithelium to particulates. Some particulates themselves can generate ROS, as can the epithelial cells, in response to appropriate stimulation. In addition, resident macrophages in the airways and the alveolar spaces can release ROS/RNS after phagocytosis of inhaled particles. These macrophages also release large amounts of tumor necrosis factor alpha (TNF-alpha), a cytokine that can generate responses within the airway epithelium dependent upon intracellular generation of ROS/RNS. As a result, signal transduction pathways are set in motion that may contribute to inflammation and other pathobiology in the airway. Such effects include increased expression of intercellular adhesion molecule 1, interleukin-6, cytosolic and inducible nitric oxide synthase, manganese superoxide dismutase, cytosolic phospholipase A2, and hypersecretion of mucus. Ultimately, ROS/RNS may play a role in the global response of the airway epithelium to particulate pollutants via activation of kinases and transcription factors common to many response genes. Thus, defense mechanisms involved in responding to offending particulates may result in a complex cascade of events that can contribute to airway pathology.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Nitrógeno/metabolismo , Especies Reactivas de Oxígeno/fisiología , Sistema Respiratorio/efectos de los fármacos , Animales , Epitelio/efectos de los fármacos , Epitelio/patología , Humanos , Sistema Respiratorio/patología
11.
Ann N Y Acad Sci ; 725: 128-45, 1994 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-8030984

RESUMEN

Epithelial cells lining respiratory airways can participate in inflammation in a number of ways. They can act as target cells, responding to exposure to a variety of inflammatory mediators and cytokines by altering one or several of their functions, such as mucin secretion, ion transport, or ciliary beating. Aberrations in any of these functions can affect local inflammatory responses and compromise pulmonary defense. For example, oxidant stress can increase secretion of mucin and depress ciliary beating efficiency, thereby affecting the ability of the mucociliary system to clear potentially pathogenic microbial agents. Recent studies have indicated that airway epithelial cells also can act as "effector" cells, synthesizing and releasing cytokines, lipid mediators, and reactive oxygen species in response to a number of pathologically relevant stimuli, thereby contributing to inflammation. Many of these epithelial-derived substances can act locally, affecting both neighboring cells and tissues, or, via autocrine or paracrine mechanisms, affect structure and function of the epithelial cells themselves. Studies in our laboratories utilized cell cultures of both human and guinea pig tracheobronchial and nasal epithelial cells, and isolated human nasal epithelial cells, to investigate activity of respiratory epithelial cells in vitro as sources of cytokines and inflammatory mediators. Primary cultures of guinea pig and human tracheobronchial and nasal epithelial cells synthesize and secrete low levels of IL-6 and IL-8 constitutively. Production and release of these cytokines increases substantially after exposure to specific inflammatory stimuli, such as TNF or IL-1, and after viral infection.


Asunto(s)
Citocinas/fisiología , Inflamación/fisiopatología , Pulmón/fisiología , Pulmón/fisiopatología , Animales , Asma/fisiopatología , Células Epiteliales , Epitelio/fisiología , Humanos , Inflamación/inmunología , Pulmón/citología , Fenómenos Fisiológicos Respiratorios , Sistema Respiratorio/citología , Sistema Respiratorio/fisiopatología
12.
Ann N Y Acad Sci ; 796: 30-7, 1996 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-8906209

RESUMEN

Within the past several years research on the interaction of cytokines and adhesion molecules with airway epithelium in diseases has allowed us to develop a better understanding of the disease process. The cytokine, TNF alpha and the adhesion molecule ICAM-1 are important mediators in the pathogenesis of airway diseases such as asthma, chronic bronchitis, and adult respiratory distress syndrome. Effects of TNF alpha on ICAM-1 surface expression was investigated in both primary cultures of normal human bronchial epithelial (NHBE) cells and immortalized human bronchial epithelial cell line BEAS-2B. TNF alpha (0.015-150 ng/mL) significantly enhanced ICAM-1 surface expression (measured by flow cytometry) in a dose and time-dependent manner, with peak expression seen at 24 hours. This response was negated by heat inactivation of the TNF alpha prior to incubation. TNF alpha-induced ICAM-1 expression also was inhibited by pre- and coincubation of TNF alpha with 3 micrograms/mL soluble TNF-R1 or by the PKC inhibitor, Calphostin C (0.1 and 0.5 microM). The ROI scavengers, dimethylthiourea (4 mM), and dimethyl sulfoxide (0.001%), enhanced TNF alpha-induced ICAM-1 expression. Collectively, these results indicate that TNF alpha-induced ICAM-1 surface expression is a specific receptor-mediated response (TNF-R1), which is mediated by mechanisms dependent on PKC and intracellular reactive oxygen species.


Asunto(s)
Bronquios/citología , Molécula 1 de Adhesión Intercelular/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología , Adulto , Antígenos CD/metabolismo , Antioxidantes/farmacología , Bronquios/metabolismo , Línea Celular , Epitelio/metabolismo , Citometría de Flujo , Humanos , Proteína Quinasa C/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral
13.
In Vitro Cell Dev Biol Anim ; 29(6): 481-92, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27519750

RESUMEN

In vitro culture conditions enabling rat tracheal epithelial (RTE) cells to differentiate to mucociliary, mucous, or squamous phenotypes are described. Medium composition for rapid cell growth to confluence in membrane insert cultures was determined, and the effects of major modifiers of differentiation were tested. Retinoic acid (RA), collagen gel substratum, and an air-liquid interface at the level of the cell layer were required for expression of a mucociliary phenotype which most closely approximated the morphology of the tracheal epithelium in vivo. Large quantities of high molecular weight, hyaluronidase-resistant glycoconjugates, most likely mucin glycoproteins, were produced in the presence of RA when the cells were grown with or without a collagen gel and in submerged as well as in interface cultures. However, extensive ciliagenesis was dependent on the simultaneous presence of RA, collagen gel, and an air-liquid interface. When RA was omitted from the media, the cells became stratified squamous and developed a cornified apical layer in air-liquid interface cultures. This phenotype was accompanied by loss of transglutaminase (TGase) type II and keratin 18 and expression of the squamous markers TGase type I and keratin 13. The ability to modulate RTE cell phenotypes in culture will facilitate future studies investigating molecular regulation of tracheal cell proliferation, differentiation, and function.

14.
In Vitro Cell Dev Biol Anim ; 29A(6): 481-92, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7687243

RESUMEN

In vitro culture conditions enabling rat tracheal epithelial (RTE) cells to differentiate to mucociliary, mucous, or squamous phenotypes are described. Medium composition for rapid cell growth to confluence in membrane insert cultures was determined, and the effects of major modifiers of differentiation were tested. Retinoic acid (RA), collagen gel substratum, and an air-liquid interface at the level of the cell layer were required for expression of a mucociliary phenotype which most closely approximated the morphology of the tracheal epithelium in vivo. Large quantities of high molecular weight, hyaluronidase-resistant glycoconjugates, most likely mucin glycoproteins, were produced in the presence of RA when the cells were grown with or without a collagen gel and in submerged as well as in interface cultures. However, extensive ciliagenesis was dependent on the simultaneous presence of RA, collagen gel, and an air-liquid interface. When RA was omitted from the media, the cells became stratified squamous and developed a cornified apical layer in air-liquid interface cultures. This phenotype was accompanied by loss of transglutaminase (TGase) type II and keratin 18 and expression of the squamous markers TGase type I and keratin 13. The ability to modulate RTE cell phenotypes in culture will facilitate future studies investigating molecular regulation of tracheal cell proliferation, differentiation, and function.


Asunto(s)
Medios de Cultivo , Tráquea/citología , Animales , Anticuerpos Monoclonales , Diferenciación Celular/fisiología , Colágeno/farmacología , Células Epiteliales , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Inmersión , Inmunohistoquímica , Técnicas In Vitro , Queratinas/metabolismo , Masculino , Fenotipo , Ratas , Ratas Endogámicas F344 , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Transglutaminasas/metabolismo , Tretinoina/farmacología
15.
Hybridoma ; 13(4): 281-7, 1994 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7806248

RESUMEN

Thirty-five hybridomas that secrete mouse monoclonal antibodies (MAb) against guinea pig (G.P.) tracheal mucins were established. The MAbs were characterized immunologically, biochemically, and immunohistochemically at both light and electron microscopic levels. Isotyping of the MAbs revealed 14 to be IgM, 13 IgG1, 3 IgG2, and 5 IgG3. The MAbs demonstrated various patterns of binding in immunoblots against mucins derived from G.P. tracheal explants. This suggested the presence of "subpopulations" of G.P. tracheal mucins with specific MAbs binding to different epitopes on the mucin molecules. Periodate oxidation indicated that 33 of the 35 MAbs recognized carbohydrate epitopes on the mucin molecules. Ten of the MAbs also reacted with both bovine and ferret tracheal mucins, while 7 and 6 MAbs bound only to bovine and ferret tracheal mucins, respectively. The generated MAbs should be useful for immunomeasurement of mucin secretion in vivo (e.g., in bronchoalveolar or airway lavage fluid) and in vitro (e.g., cell and organ cultures) from cells of guinea pig and (with certain MAbs) bovine and ferret origin.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Mucinas/inmunología , Tráquea/inmunología , Animales , Western Blotting , Cromatografía de Afinidad , Ensayo de Inmunoadsorción Enzimática , Cobayas , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Microscopía Inmunoelectrónica , Mucinas/análisis , Ácido Peryódico/inmunología , Especificidad de la Especie , Tráquea/química
16.
Am J Vet Res ; 57(4): 574-9, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8712527

RESUMEN

OBJECTIVE: To test the hypothesis that platelet-activating factor (PAF) induces inositol phosphate turnover through a receptor-linked, pertussis toxin-sensitive guanine nucleotide-binding (G) protein-dependent pathway in porcine alveolar macrophages. DESIGN: Randomized complete block design was used with 2 or 3 replicates/block. ANIMALS: Porcine alveolar macrophages were obtained by lavage of excised lungs from Yorkshire-type pigs (mean +/- SEM, 21 +/- 2 kg). PROCEDURE: Phospholipase C activation was assessed, using anion exchange chromatography to measure accumulation of inositol phosphates in [3H]myo-inositol-labeled alveolar macrophages. Macrophages were incubated with saline solution, pertussis toxin (4.75 nM), or B-oligomer (4.75 nM) for 2 hours. Cells then were washed and incubated for 5 minutes with PAF (0, 0.1, 1.0, or 10 microM; n = 15). Results were expressed as total inositol phosphates (inositol monophosphate, bisphosphate, trisphosphate, and tetrakisphosphate). RESULTS: Concentrations of total inositol phosphates were significantly (P < 0.05) increased to 162 +/- 7, 172 +/- 4, and 194 +/- 9% of control in response to 0.1, 1.0, and 10 microM PAF, respectively. Pertussis toxin attenuated the PAF-induced increase in total inositol phosphates by approximately 50% (P < 0.05). The B-oligomer of pertussis toxin failed to modify PAF-induced increases in total inositol phosphates. The specific PAF receptor antagonist WEB 2086 markedly attenuated PAF-induced. (10 microM) increase in inositol phosphates. CONCLUSIONS: We conclude that PAF stimulates accumulation of inositol phosphates through a specific receptor and that a pertussis toxin-sensitive G protein is involved in the signal transduction process leading to activation of phospholipase C in porcine alveolar macrophages.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Fosfatos de Inositol/metabolismo , Macrófagos Alveolares/fisiología , Toxina del Pertussis , Factor de Activación Plaquetaria/farmacología , Factores de Virulencia de Bordetella/farmacología , Adenosina Difosfato Ribosa/metabolismo , Cloruro de Aluminio , Compuestos de Aluminio/farmacología , Análisis de Varianza , Animales , Azepinas/farmacología , Lavado Broncoalveolar , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cloruros/farmacología , Inositol/metabolismo , L-Lactato Deshidrogenasa , Macrófagos Alveolares/citología , Macrófagos Alveolares/efectos de los fármacos , NAD/metabolismo , Inhibidores de Agregación Plaquetaria/farmacología , Distribución Aleatoria , Fluoruro de Sodio/farmacología , Porcinos , Triazoles/farmacología , Fosfolipasas de Tipo C/metabolismo
17.
Oncogene ; 33(28): 3696-706, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23955080

RESUMEN

Myristoylated Alanine-Rich C Kinase Substrate (MARCKS), a substrate of protein kinase C, is a key regulatory molecule controlling mucus granule secretion by airway epithelial cells as well as directed migration of leukocytes, stem cells and fibroblasts. Phosphorylation of MARKCS may be involved in these responses. However, the functionality of MARCKS and its related phosphorylation in lung cancer malignancy have not been characterized. This study demonstrated elevated levels of MARCKS and phospho-MARCKS in highly invasive lung cancer cell lines and lung cancer specimens from non-small-cell lung cancer patients. siRNA knockdown of MARCKS expression in these highly invasive lung cancer cell lines reduced cell migration and suppressed PI3K (phosphatidylinositol 3'-kinase)/Akt phosphorylation and Slug level. Interestingly, treatment with a peptide identical to the MARCKS N-terminus sequence (the MANS peptide) impaired cell migration in vitro and also the metastatic potential of invasive lung cancer cells in vivo. Mechanistically, MANS peptide treatment resulted in a coordination of increase of E-cadherin expression, suppression of MARCKS phosphorylation and AKT/Slug signalling pathway but not the expression of total MARCKS. These results indicate a crucial role for MARCKS, specifically its phosphorylated form, in potentiating lung cancer cell migration/metastasis and suggest a potential use of MARCKS-related peptides in the treatment of lung cancer metastasis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/química , Neoplasias Pulmonares/patología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Fragmentos de Péptidos/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Invasividad Neoplásica , Metástasis de la Neoplasia , Oncogenes/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA