Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 296(3): L372-83, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19112100

RESUMEN

Among the multiple organ disorders caused by the severe acute respiratory syndrome coronavirus (SARS-CoV), acute lung failure following atypical pneumonia is the most serious and often fatal event. We hypothesized that two of the hydrophilic structural coronoviral proteins (S and E) would regulate alveolar fluid clearance by decreasing the cell surface expression and activity of amiloride-sensitive epithelial sodium (Na(+)) channels (ENaC), the rate-limiting protein in transepithelial Na(+) vectorial transport across distal lung epithelial cells. Coexpression of either S or E protein with human alpha-, beta-, and gamma-ENaC in Xenopus oocytes led to significant decreases of both amiloride-sensitive Na(+) currents and gamma-ENaC protein levels at their plasma membranes. S and E proteins decreased the rate of ENaC exocytosis and either had no effect (S) or decreased (E) rates of endocytosis. No direct interactions among SARS-CoV E protein with either alpha- or gamma-ENaC were indentified. Instead, the downregulation of ENaC activity by SARS proteins was partially or completely restored by administration of inhibitors of PKCalpha/beta1 and PKCzeta. Consistent with the whole cell data, expression of S and E proteins decreased ENaC single-channel activity in oocytes, and these effects were partially abrogated by PKCalpha/beta1 inhibitors. Finally, transfection of human airway epithelial (H441) cells with SARS E protein decreased whole cell amiloride-sensitive currents. These findings indicate that lung edema in SARS infection may be due at least in part to activation of PKC by SARS proteins, leading to decreasing levels and activity of ENaC at the apical surfaces of lung epithelial cells.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Proteína Quinasa C/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Proteínas Virales/metabolismo , Lesión Pulmonar Aguda/etiología , Amilorida/farmacología , Animales , Línea Celular , Endocitosis , Activación Enzimática , Canales Epiteliales de Sodio/genética , Exocitosis , Femenino , Expresión Génica , Humanos , Técnicas In Vitro , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Oocitos/metabolismo , Técnicas de Placa-Clamp , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Edema Pulmonar/etiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Glicoproteína de la Espiga del Coronavirus , Transfección , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/genética , Proteínas Viroporinas , Xenopus
2.
J Cell Biochem ; 105(4): 1092-108, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18816594

RESUMEN

mrtl (myc-related translation/localization regulatory factor) is a previously uncharacterized protein synthesized from the first open reading frame contained within the human c-myc P0 transcript, approximately 800 nucleotides upstream of the Myc coding sequence. The mrtl protein, 114 amino acids in length, is projected to contain an N-terminal transmembrane domain and a highly charged C-terminal interaction domain with homology to numerous RNA-binding proteins. Using monoclonal antibodies raised against the hydrophilic C-terminal domain, endogenous mrtl was visualized in human breast tumor cell lines and primary mammary epithelial cells at the nuclear envelope and contiguous endoplasmic/nucleoplasmic reticulum. mrtl colocalizes and coimmunoprecipitates with translation initiation factor eIF2alpha and the 40S ribosomal protein RACK1, and appears capable of binding specifically to the c-myc RNA. Inducible ectopic overexpression of wild-type mrtl interferes with the function of endogenous mrtl, which results in loss of Myc from the nucleus. Furthermore, treatment of cells with a peptide derived from the C-terminal domain displaces endogenous mrtl and causes a dramatic reduction in total cellular Myc protein levels. Together with our previous work demonstrating complete loss of tumorigenicity in association with ectopic expression of the c-myc P0 5'-UTR (containing the mrtl coding sequence), these results suggest that mrtl may serve an important function in regulating Myc translation and localization to the nucleus, perhaps ultimately contributing to the role of the c-myc locus in oncogenesis.


Asunto(s)
Proteínas Portadoras , Retículo Endoplásmico/química , Matriz Nuclear/química , Proteínas Proto-Oncogénicas c-myc/genética , Transporte Activo de Núcleo Celular , Neoplasias de la Mama/etiología , Neoplasias de la Mama/patología , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Línea Celular Tumoral , Femenino , Humanos , Glándulas Mamarias Humanas/citología , Sistemas de Lectura Abierta , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Proteínas de Unión al ARN
3.
Nat Immunol ; 9(2): 166-75, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18157131

RESUMEN

Interleukin 17 (IL-17) is a cytokine associated with inflammation, autoimmunity and defense against some bacteria. Here we show that IL-17 can promote autoimmune disease through a mechanism distinct from its proinflammatory effects. As compared with wild-type mice, autoimmune BXD2 mice express more IL-17 and show spontaneous development of germinal centers (GCs) before they increase production of pathogenic autoantibodies. We show that blocking IL-17 signaling disrupts CD4+ T cell and B cell interactions required for the formation of GCs and that mice lacking the IL-17 receptor have reduced GC B cell development and humoral responses. Production of IL-17 correlates with upregulated expression of the genes Rgs13 and Rgs16, which encode regulators of G-protein signaling, and results in suppression of the B cell chemotactic response to the chemokine CXCL12. These findings suggest a mechanism by which IL-17 drives autoimmune responses by promoting the formation of spontaneous GCs.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Centro Germinal/inmunología , Interleucina-17/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Interleucina-17/antagonistas & inhibidores , Ratones , Ratones Mutantes , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo
4.
Cell Cycle ; 6(23): 2944-52, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18000399

RESUMEN

LIM kinases (LIMK1 and LIMK2) are LIM domain containing serine/threonine kinases that modulate reorganization of actin cytoskeleton through inactivating phosphorylation of cofilin. The Rho family of small GTPases regulates the catalytic activity of LIMK1 and LIMK2 through activating phosphorylation by ROCK or by p21 kinase. Recent studies have suggested that LIMK1 could play a role in modulation of cellular growth by alteration of the cell cycle in breast and prostate tumor cells; however, the direct mitogenic effects of LIMK1 in these tumor cells is yet to be elucidated. Via immunofluorescence, in this study, we show that phosphorylated LIM kinases (pLIMK1/2) are colocalized with gamma-tubulin in the centrosomes during the early mitotic phases of human breast and prostate cancer cells (MDA-MB-231 and DU145); apparent colocalization begins in the centrosomes in prophase. As shown by both bright field (MDA-MB-231) and fluorescent immunohistochemistry (MDA-MB-231 and DU145), pLIMK1/2 does not localize to centrosomes during interphase. By bright field immunohistochemistry, the largest area of the centrosome that is stained with pLIMK1/2 occurs at anaphase. In early telophase, reduced staining of pLIMK1/2 at the spindle poles and concomitant accumulation of pLIMK1/2 at the cleavage furrow begins to occur. In late telophase, loss of staining of pLIMK1/2 and of colocalization with gamma-tubulin occurs at the poles and pLIMK1/2 became further concentrated at the junction between the two daughter cells. Co-immunoprecipitation studies indicated that gamma-tubulin associates with phosphorylated LIMK1 and LIMK2 but not with dephosphorylated LIMK1 or LIMK2. The results suggest that activated LIMK1/2 may associate with gamma-tubulin and play a role in mitotic spindle assembly.


Asunto(s)
Centrosoma/química , Quinasas Lim/metabolismo , Mitosis , Tubulina (Proteína)/metabolismo , Línea Celular Tumoral , Humanos , Interfase , Fosforilación , Transporte de Proteínas , Huso Acromático
5.
J Biol Chem ; 282(50): 36481-8, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17913705

RESUMEN

We present the evidence for a direct physical association of cystic fibrosis transmembrane conductance regulator (CFTR) and epithelial sodium channel (ENaC), two major ion channels implicated in the pathophysiology of cystic fibrosis, a devastating inherited disease. We employed fluorescence resonance energy transfer, a distance-dependent imaging technique with capability to detect molecular complexes with near angstrom resolution, to estimate the proximity of CFTR and ENaC, an essential variable for possible physical interaction to occur. Fluorescence resonance energy transfer studies were complemented with a classic biochemical approach: coimmunoprecipitation. Our results place CFTR and ENaC within reach of each other, suggestive of a direct interaction between these two proteins.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Canales Epiteliales de Sodio/metabolismo , Línea Celular , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Canales Epiteliales de Sodio/genética , Transferencia Resonante de Energía de Fluorescencia , Humanos , Unión Proteica/genética
6.
Mol Biol Cell ; 16(10): 4905-17, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16055507

RESUMEN

Nuclear aggregates formed by proteins containing expanded poly-glutamine (poly-Q) tracts have been linked to the pathogenesis of poly-Q neurodegenerative diseases. Here, we show that a protein (GFP170*) lacking poly-Q tracts forms nuclear aggregates that share characteristics of poly-Q aggregates. GFP170* aggregates recruit cellular chaperones and proteasomes, and alter the organization of nuclear domains containing the promyelocytic leukemia (PML) protein. These results suggest that the formation of nuclear aggregates and their effects on nuclear architecture are not specific to poly-Q proteins. Using GFP170* as a model substrate, we explored the mechanistic details of nuclear aggregate formation. Fluorescence recovery after photobleaching and fluorescence loss in photobleaching analyses show that GFP170* molecules exchange rapidly between aggregates and a soluble pool of GFP170*, indicating that the aggregates are dynamic accumulations of GFP170*. The formation of cytoplasmic and nuclear GFP170* aggregates is microtubule-dependent. We show that within the nucleus, GFP170* initially deposits in small aggregates at or adjacent to PML bodies. Time-lapse imaging of live cells shows that small aggregates move toward each other and fuse to form larger aggregates. The coalescence of the aggregates is accompanied by spatial rearrangements of the PML bodies. Significantly, we find that the larger nuclear aggregates have complex internal substructures that reposition extensively during fusion of the aggregates. These studies suggest that nuclear aggregates may be viewed as dynamic multidomain inclusions that continuously remodel their components.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Agrecanos , Animales , Células COS , Núcleo Celular/ultraestructura , Chlorocebus aethiops , Citoplasma/metabolismo , Citoplasma/ultraestructura , Proteínas de la Matriz Extracelular/metabolismo , Proteínas Fluorescentes Verdes/genética , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/genética , Microscopía Electrónica de Transmisión , Péptidos/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Transporte de Proteínas , Proteoglicanos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
7.
Environ Mol Mutagen ; 46(1): 43-52, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15880734

RESUMEN

The environmental estrogen, dichlorodiphenyltrichloroethane (DDT), and its metabolites have been implicated in the development of breast cancer through mechanisms that remain to be elucidated. It has been hypothesized that exposure to DDT and its metabolites, during critical periods of development, can contribute to an elevated risk for breast cancer in adults. In the present study, we have investigated the effect of o,p'-DDT on mammary gland cell proliferation and chromosomal alterations, in a rat mammary cancer model (commonly used to study human cancer), to gain insights into its potential role in the development of breast cancer. Twenty-one-day-old female Sprague-Dawley (SD) rats were administered o,p'-DDT, 7,12-dimethylbenz[a]anthracene (DMBA), genistein, DDT+DMBA, or DDT+DMBA+genistein, over a 14-day period. To determine changes in chromosome number and structure, we used the micronucleus assay as well as multicolor fluorescence in situ hybridization (FISH) region-specific DNA probes for rat chromosomes 4 and 19. Cell proliferation was evaluated using 5-bromo-2'-deoxyuridine (BrdU). Significant increases in BrdU-incorporated cells were seen in the rats treated with DDT+DMBA. Although micronucleus frequencies were somewhat elevated in several of the treatment groups, significant increases were not seen in any of them. Significant increases in numerical chromosomal aberrations were detected in all of the DDT- and DMBA-treated groups. Genistein significantly reduced BrdU incorporation and polyploidy in the DDT+DMBA-treated rats. These initial studies indicate that DDT and DMBA can induce cellular and chromosomal alterations in the rat mammary gland, which is consistent with the hypothesis that these agents can induce early events in mammary carcinogenesis.


Asunto(s)
Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Micronúcleos con Defecto Cromosómico/inducido químicamente , Poliploidía , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , DDT/toxicidad , Sinergismo Farmacológico , Femenino , Genisteína/toxicidad , Hibridación Fluorescente in Situ , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Pruebas de Micronúcleos , Ratas , Ratas Sprague-Dawley
8.
Am J Physiol Renal Physiol ; 287(2): F329-35, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15082450

RESUMEN

Renin release from juxtaglomerular granular cells is considered the rate-limiting step in activation of the renin-angiotensin system that helps to maintain body salt and water balance. Available assays to measure renin release are complex, indirect, and work with significant internal errors. To directly visualize and study the dynamics of both the release and tissue activity of renin, we isolated and perfused afferent arterioles with attached glomeruli dissected from rabbit kidneys and used multiphoton fluorescence imaging. Acidotropic fluorophores, such as quinacrine and LysoTrackers, clearly and selectively labeled renin granules. Immunohistochemistry of mouse kidney with a specific renin antibody and quinacrine staining colocalized renin granules and quinacrine fluorescence. A low-salt diet for 1 wk caused an approximately fivefold increase in the number of both individual granules and renin-positive granular cells. Time-lapse imaging showed no signs of granule trafficking or any movement, only the dimming and disappearance of fluorescence from individual renin granules within 1 s in response to 100 microM isoproterenol. There appeared to be a quantal release of the granular contents; i.e., an all-or-none phenomenon. Using As4.1 cells, a granular cell line, we observed further classic signs of granule exocytosis, the emptying of granule content associated with a flash of quinacrine fluorescence. Using a fluorescence resonance energy transfer-based, 5-(2-aminoethylamino)naphthalene-1-sulfonic acid (EDANS)-conjugated renin substrate in the bath, an increase in EDANS fluorescence (renin activity) was observed around granular cells in response to isoproterenol. Fluorescence microscopy is an excellent tool for the further study of the mechanism, regulation, and dynamics of renin release.


Asunto(s)
Sistemas de Computación , Microscopía Confocal , Microscopía Fluorescente , Renina/metabolismo , Animales , Colorantes Fluorescentes , Técnicas In Vitro , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/metabolismo , Ratones , Conejos
9.
Cancer Res ; 63(23): 8079-84, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678956

RESUMEN

Aneuploidy and chromosomal aberrations are hallmarks of most human epithelial malignancies. Here we show that overexpression of the oncogenic kinase Pim-1 in human prostate epithelial cells induces genomic instability by subverting the mitotic spindle checkpoint. Cells overexpressing Pim-1 have a defect in the mitotic spindle checkpoint, abnormal mitotic spindles, centrosome amplification, and chromosome missegregation. Polyploidy and aneuploidy ensue due to a delay in completing cytokinesis. These results define a novel role for elevated Pim-1 expression in promoting genomic instability in human prostate tumors.


Asunto(s)
Segregación Cromosómica , Poliploidía , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Ciclo Celular/fisiología , Línea Celular Tumoral , Células Epiteliales/enzimología , Células Epiteliales/patología , Humanos , Masculino , Mitosis/genética , Próstata/enzimología , Próstata/fisiología , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-pim-1 , Huso Acromático/genética
10.
J Clin Invest ; 112(9): 1332-41, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14597760

RESUMEN

Previously, we described an APC-adenovirus (APC-Ad) FasL cell gene therapy method which could be used to deplete autoreactive T cells in vivo. FasL was toxic, however, and controlled regulation of FasL was not achieved. Here we describe an improved approach to delivering TNF-related apoptosis-inducing ligand (TRAIL) in vivo in which collagen II-induced (CII-induced) arthritis-susceptible (CIA-susceptible) DBA/1j mice were treated with CII-pulsed DCs that had been transfected with a novel Ad system. The Ad was engineered to exhibit inducible TRAIL under the control of the doxycycline-inducible (DOX-inducible) tetracycline response element (TRE). Four groups of mice were treated with CII-DC-AdTRAIL+DOX, CII-DC-AdTRAIL (no DOX), CII-DC-AdGFP+DOX, or DC-AdTRAIL+DOX (no CII), beginning 2 weeks after priming with CII in CFA. The incidence of arthritis and infiltration of T cells in the joint was significantly decreased in CII-DC-AdTRAIL+DOX-treated mice. The in vitro splenic T cell proliferative response and induction of IFN-gamma to bovine CII stimulation were also significantly reduced in mice treated with CII-DC-AdTRAIL+DOX. AdTRAIL+DOX was not toxic to DCs or mice but could induce activated T cells to undergo apoptosis in the spleen. Our results suggest that CII-DC-AdTRAIL+DOX cell gene therapy is a safe and effective method for inhibiting the development of CIA.


Asunto(s)
Artritis Reumatoide/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , Colágeno Tipo II/inmunología , Células Dendríticas/fisiología , Terapia Genética , Glicoproteínas de Membrana/genética , Linfocitos T/fisiología , Factor de Necrosis Tumoral alfa/genética , Adenoviridae/genética , Animales , Anticuerpos/inmunología , Formación de Anticuerpos , Apoptosis , Proteínas Reguladoras de la Apoptosis , Movimiento Celular , Doxorrubicina/farmacología , Femenino , Activación de Linfocitos , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos DBA , Bazo/patología , Ligando Inductor de Apoptosis Relacionado con TNF , Transfección , Factor de Necrosis Tumoral alfa/fisiología
11.
J Gene Med ; 5(10): 839-851, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14533192

RESUMEN

BACKGROUND: Systemic adenoviral (Ad) gene therapy for renal disorders is largely hampered by the unique architecture of the kidney. Consequently, currently available Ad vectors are of only limited therapeutic utility in the context of glomerular and fibroproliferative renal diseases. METHODS: The Ad vectors studied in the context of blocking renal fibrosis were AdTbeta-ExR and AdCATbeta-TR. AdTbeta-ExR encodes a chimeric soluble molecule comprising the entire ectodomain of the human type II TGF-beta receptor, genetically fused to the Fc fragment of the human IgG1 (sTbetaRII), while AdCATbeta-TR encodes only the dominant-negative truncated ectodomain of the human type II TGF-beta receptor. The biologic activity of the type II TGF-beta receptor was evaluated in vitro by its ability to inhibit cellular proliferation and in vivo in a unilateral ureter obstruction fibrosis model. Renal targeting with sTbetaRII was evaluated immunohistochemically after intramuscular (IM) delivery of AdTbeta-ExR. The renal antifibrotic effect of the Ad vectors was evaluated in a lupus murine model with both light and electron microscopy and urinalysis. RESULTS: sTbetaRII was detected in the glomeruli after remote IM injection of AdTbeta-ExR, but not the control AdCATbeta-TR, indicating renal deposition of the heterologous soluble fusion protein after its expression in the muscle and secretion into the circulation. AdTbeta-ExR, but not AdCATbeta-TR, could transiently inhibit mesangial expansion, glomerular hypercellularity, proteinuria and cortical interstitial fibrosis in a murine lupus model. However, the autoimmune renal disease eventually surpassed the antifibrotic effect. CONCLUSIONS: These results indicate the superiority of a soluble type II TGF-beta receptor over a dominant-negative, non-soluble type II TGF-beta receptor in the context of blocking renal fibrosis in murine models.


Asunto(s)
Terapia Genética , Enfermedades Renales/terapia , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Adenoviridae/genética , Animales , Modelos Animales de Enfermedad , Fibrosis/terapia , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Glomerulonefritis/genética , Glomerulonefritis/terapia , Inyecciones Intramusculares , Riñón/patología , Enfermedades Renales/genética , Ratones , Ratones Endogámicos BALB C , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Proteínas Recombinantes de Fusión/genética
12.
J Exp Med ; 198(1): 123-32, 2003 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-12835480

RESUMEN

Limited frequencies of T cells express IL-2 in primary antigenic responses, despite activation marker expression and proliferation by most clonal members. To define the basis for restricted IL-2 expression, a videomicroscopic system and IL-2 reporter transgenic model were used to characterize dendritic cell (DC)-T cell interactions. T cells destined to produce IL-2 required prolonged interactions with DCs, whereas most T cells established only transient interactions with DCs and were activated, but did not express IL-2. Extended conjugation of T cells with DCs was not always sufficient to initiate IL-2 expression. Thus, there is intrinsic variability in clonal T cell populations that restricts IL-2 commitment, and prolonged engagement with mature DCs is necessary, but not sufficient, for IL-2 gene transcription.


Asunto(s)
Comunicación Celular , Células Dendríticas/fisiología , Interleucina-2/genética , Linfocitos T/fisiología , Animales , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Receptores de Antígenos de Linfocitos T/fisiología , Transcripción Genética
13.
Histochem Cell Biol ; 119(6): 437-46, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12768285

RESUMEN

The acid-sensing ion channels (ASICs) are members of the DEG/ENaC superfamily of Na+ channels. Acid-gated cation currents have been detected in neurons from multiple regions of the brain including the cerebellum, but little is known about their molecular identity and function. Recently, one of ASICs (ASIC1a) was implicated in synaptic plasticity. In this study we examined the subcellular distribution of ASIC2a in rat cerebellum by immunostaining and confocal microscopy. Monoclonal antibodies for labeling of defined brain structures, for example, astroglia, Purkinje cell dendrites, nuclei, and presynaptic terminals were used for colocalization analyses. In the gray matter, the anti-ASIC2a antibody intensively stained dendrite branches of Purkinje cells evenly distributed throughout the entire molecular layer (ML). In the granule cell layer (GL), anti-ASIC2a antibody stained synaptic glomeruli. Neuronal localization of ASIC2a was confirmed by lack of co-staining with glial fibrillary acidic protein. Anti-ASIC2a staining in the ML colocalized with metabotropic glutamate receptor 1alpha (mGluR1alpha) in Purkinje cell dendrites and dendritic spines. Both proteins, mGluR1alpha and ASIC2a, were enriched in a crude synaptic membrane fraction prepared from cerebellum, suggesting synaptic expression of these proteins. Dual staining with anti-syntaxin 1A and anti-ASIC2a antibodies demonstrates characteristic complementary distribution of two proteins in both ML and GL. Because syntaxin 1A localized in presynaptic membranes and synaptic vesicles, complementary distribution with ASIC2a suggests postsynaptic localization of ASIC2a in these structures. This study shows specific localization of ASIC2a in both Purkinje and granule cell dendrites of the cerebellum and enrichment of ASIC2a in a crude cerebellar synaptic membrane fraction. The study is the first report of synaptic localization of ASIC2a in the CNS. The synaptic localization of ASIC2a in the cerebellum makes this channel a candidate for a role in motor coordination and learning.


Asunto(s)
Cerebelo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Sodio/metabolismo , Canales Iónicos Sensibles al Ácido , Animales , Antígenos de Superficie/metabolismo , Cerebelo/citología , Técnica del Anticuerpo Fluorescente Indirecta , Microscopía Confocal , Células de Purkinje/citología , Células de Purkinje/metabolismo , Ratas , Receptores de Glutamato Metabotrópico/metabolismo , Membranas Sinápticas/metabolismo , Sinaptosomas/metabolismo , Sintaxina 1
14.
Proc Natl Acad Sci U S A ; 99(19): 12477-82, 2002 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-12209004

RESUMEN

Soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) mediate membrane fusion reactions in eukaryotic cells by assembling into complexes that link vesicle-associated SNAREs with SNAREs on target membranes (t-SNAREs). Many SNARE complexes contain two t-SNAREs that form a heterodimer, a putative intermediate in SNARE assembly. Individual t-SNAREs (e.g., syntaxin 1A) also regulate synaptic calcium channels and cystic fibrosis transmembrane conductance regulator (CFTR), the epithelial chloride channel that is defective in cystic fibrosis. Whether the regulation of ion channels by individual t-SNAREs is related to SNARE complex assembly and membrane fusion is unknown. Here we show that CFTR channels are coordinately regulated by two cognate t-SNAREs, SNAP-23 (synaptosome-associated protein of 23 kDa) and syntaxin 1A. SNAP-23 physically associates with CFTR by binding to its amino-terminal tail, a region that modulates channel gating. CFTR-mediated chloride currents are inhibited by introducing excess SNAP-23 into HT29-Cl.19A epithelial cells. Conversely, CFTR activity is stimulated by a SNAP-23 antibody that blocks the binding of this t-SNARE to the CFTR amino-terminal tail. The physical and functional interactions between SNAP-23 and CFTR depend on syntaxin 1A, which binds to both proteins. We conclude that CFTR channels are regulated by a t-SNARE complex that may tune CFTR activity to rates of membrane traffic in epithelial cells.


Asunto(s)
Antígenos de Superficie/metabolismo , Proteínas Portadoras/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Transporte Vesicular , Animales , Antígenos de Superficie/química , Antígenos de Superficie/genética , Sitios de Unión , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Membrana Celular/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Activación del Canal Iónico , Células L , Sustancias Macromoleculares , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Proteínas Qb-SNARE , Proteínas Qc-SNARE , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas SNARE , Sintaxina 1
15.
J Mol Med (Berl) ; 80(9): 595-604, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12226741

RESUMEN

Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) protein. Since approximately 5% of all mutant CF alleles are stop mutations, it can be calculated that approximately 10% of CF patients carry a premature stop mutation in at least one copy of the CFTR gene. Certain ethnic groups, such as the Ashkenazi Jewish population, carry a much higher percentage of CF stop mutations. Consequently, a therapeutic strategy aimed at suppressing this class of mutation would be highly desirable for the treatment of this common genetic disease. We have shown previously that aminoglycoside antibiotics can suppress premature stop mutations in the CFTR gene in a bronchial epithelial cell line [Nat Med (1997) 3:1280]. To address whether aminoglycosides can suppress a CFTR premature stop mutation in an animal model, we constructed a transgenic mouse with a null mutation in the endogenous CFTR locus (Cftr-/-) that also expressed a human CFTR-G542X cDNA under control of the intestinal fatty acid binding protein promoter. We then investigated whether the daily administration of the aminoglycoside antibiotics gentamicin or tobramycin could restore the expression of a detectable level of CFTR protein. Immunofluorescence staining of intestinal tissues from Cftr-/- hCFTR-G542X mice revealed that gentamicin treatment resulted in the appearance of hCFTR protein at the apical surface of the glands of treated mice. Weaker staining was also observed in the intestinal glands following tobramycin treatment. Short-circuit current measurements made on intestinal tissues from these mice demonstrated that a significant number of positive cAMP-stimulated transepithelial chloride current measurements could be observed following gentamicin treatment (P=0.008) and a near significant number following tobramycin treatment (P=0.052). When taken together, these results indicate that gentamicin, and to a lesser extent tobramycin, can restore the synthesis of functional hCFTR protein by suppressing the hCFTR-G542X premature stop mutation in vivo.


Asunto(s)
Antibacterianos/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Mutación , Supresión Genética , Transgenes , Potenciales de Acción/efectos de los fármacos , Animales , Colforsina/farmacología , Fibrosis Quística/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Expresión Génica , Gentamicinas/administración & dosificación , Gentamicinas/sangre , Gentamicinas/farmacología , Heterocigoto , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tobramicina/administración & dosificación , Tobramicina/farmacología
16.
Science ; 296(5577): 2391-4, 2002 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-12089442

RESUMEN

Myeloperoxidase (MPO) is an abundant mammalian phagocyte hemoprotein thought to primarily mediate host defense reactions. Although its microbicidal functions are well established in vitro, humans deficient in MPO are not at unusual risk of infection. MPO was observed herein to modulate the vascular signaling and vasodilatory functions of nitric oxide (NO) during acute inflammation. After leukocyte degranulation, MPO localized in and around vascular endothelial cells in a rodent model of acute endotoxemia and impaired endothelium-dependent relaxant responses, to which MPO-deficient mice were resistant. Altered vascular responsiveness was due to catalytic consumption of NO by substrate radicals generated by MPO. Thus MPO can directly modulate vascular inflammatory responses by regulating NO bioavailability.


Asunto(s)
Endotelio Vascular/fisiología , Inflamación/enzimología , Leucocitos/enzimología , Óxido Nítrico/metabolismo , Peroxidasa/metabolismo , Vasodilatación , Animales , Aorta , Catálisis , Bovinos , Células Cultivadas , Cromanos/metabolismo , Cromanos/farmacología , Técnicas de Cocultivo , GMP Cíclico/metabolismo , Endotelio Vascular/enzimología , Endotoxemia/enzimología , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Inflamación/fisiopatología , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Mutación , Oxidación-Reducción , Peroxidasa/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Transfección , Células Tumorales Cultivadas
17.
Anat Rec ; 267(1): 7-16, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11984787

RESUMEN

C2 is a serum glycoprotein that is essential for activation of the classical and lectin pathways of the complement system. We reported previously that in transiently transfected COS cells, C2 accumulates in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC). Transfection with a cDNA corresponding to a variant C2 mRNA in which exon 17 is spliced out, C2Delta(17), resulted in retention of the mutant polypeptide in the ER. We now show that calnexin, a lectin-like chaperone, colocalizes with wild-type (wt) C2 and C2Delta(17). Biosynthetic labeling and sequential immunoprecipitation experiments indicated that colocalization is due to a physical association between calnexin and C2. Immunofluorescence analysis indicated that calnexin was upregulated in cells transfected with either C2 species. Upregulation of calnexin was not affected by castanospermine, which inhibits glucosidases I and II. However, castanospermine inhibited translocation of calnexin to the ERGIC in wt C2 transfected cells. Upregulation of calnexin was also observed in cells transfected with the complement protein factor B, a glycoprotein with extensive structural and functional similarities to C2, but not in cells transfected with complement proteins C3 or factor D, which have no structural similarity to C2, and low or no glycan content, respectively. Calnexin upregulation by transfection with C2 or factor B, but not factor D, was also demonstrated by quantitative analysis of calnexin immunoprecipitates from biosynthetically labeled cells. Increased calnexin expression by overexpressed C2 and factor B appears to be triggered either by the high glycan content of these proteins or, since it also occurs in the presence of castanospermine, by shared features of the structure of these two proteins.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Complemento C2/metabolismo , Chaperonas Moleculares/metabolismo , Animales , Células CHO/metabolismo , Células COS/metabolismo , Proteínas de Unión al Calcio/análisis , Calnexina , Chlorocebus aethiops , Factor B del Complemento/metabolismo , Cricetinae , Técnica del Anticuerpo Fluorescente , Humanos , Chaperonas Moleculares/análisis , Transfección , Regulación hacia Arriba
18.
Am J Physiol Renal Physiol ; 282(3): F541-52, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11832437

RESUMEN

Cilia are organelles that play diverse roles, from fluid movement to sensory reception. Polaris, a protein associated with cystic kidney disease in Tg737(o)(rpk) mice, functions in a ciliogenic pathway. Here, we explore the role of polaris in primary cilia on Madin-Darby canine kidney cells. The results indicate that polaris localization and solubility change dramatically during cilia formation. These changes correlate with the formation of basal bodies and large protein rafts at the apical surface of the epithelia. A cortical collecting duct cell line has been derived from mice with a mutation in the Tg737 gene. These cells do not develop normal cilia, which can be corrected by reexpression of the wild-type Tg737 gene. These data suggest that the primary cilia are important for normal renal function and/or development and that the ciliary defect may be a contributing factor to the cystic disease in Tg737(o)(rpk) mice. Further characterization of these cells will be important in elucidating the physiological role of renal cilia and in determining their relationship to cystic disease.


Asunto(s)
Proteínas de Caenorhabditis elegans , Cilios/patología , Riñón/patología , Proteínas del Tejido Nervioso , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas/genética , Proteínas/metabolismo , Proteínas Supresoras de Tumor , Animales , Antineoplásicos/farmacología , Línea Celular , Femenino , Expresión Génica/fisiología , Masculino , Ratones , Ratones Mutantes , Nocodazol/farmacología , Fenotipo , ARN Mensajero/análisis , Transducción de Señal/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...