Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Am J Physiol Cell Physiol ; 302(10): C1436-51, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22301060

RESUMO

Pathophysiological anomalies in autosomal dominant and recessive forms of polycystic kidney disease (PKD) may derive from impaired function/formation of the apical central monocilium of ductal epithelia such as that seen in the Oak Ridge polycystic kidney or orpk (Ift88(Tg737Rpw)) mouse and its immortalized cell models for the renal collecting duct. According to a previous study, Na/H exchanger (NHE) activity may contribute to hyperabsorptive Na(+) movement in cilium-deficient ("mutant") cortical collecting duct principal cell monolayers derived from the orpk mice compared with cilium-competent ("rescued") monolayers. To examine NHE activity, we measured intracellular pH (pH(i)) by fluorescence imaging with the pH-sensitive dye BCECF, and used a custom-designed perfusion chamber to control the apical and basolateral solutions independently. Both mutant and rescued monolayers exhibited basolateral Na(+)-dependent acid-base transporter activity in the nominal absence of CO(2)/HCO(3)(-). However, only the mutant cells displayed appreciable apical Na(+)-induced pH(i) recoveries from NH(4)(+) prepulse-induced acid loads. Similar results were obtained with isolated, perfused collecting ducts from orpk vs. wild-type mice. The pH(i) dependence of basolateral cariporide/HOE-694-sensitive NHE activity under our experimental conditions was similar in both mutant and rescued cells, and 3.5- to 4.5-fold greater than apical HOE-sensitive NHE activity in the mutant cells (pH(i) 6.23-6.68). Increased apical NHE activity correlated with increased apical NHE1 expression in the mutant cells, and increased apical localization in collecting ducts of kidney sections from orpk vs. control mice. A kidney-specific conditional cilium-knockout mouse produced a more acidic urine compared with wild-type littermates and became alkalotic by 28 days of age. This study provides the first description of altered NHE activity, and an associated acid-base anomaly in any form of PKD.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Córtex Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Doenças Renais Policísticas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Regulação para Cima , Animais , Proteínas de Transporte de Cátions/genética , Técnicas de Cultura de Células , Cílios/metabolismo , Cílios/patologia , Modelos Animais de Doenças , Córtex Renal/patologia , Túbulos Renais Coletores/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Regulação para Cima/genética
2.
BMC Physiol ; 12: 12, 2012 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-22999299

RESUMO

BACKGROUND: Rescue or correction of CFTR function in native epithelia is the ultimate goal of CF therapeutics development. Wild-type (WT) CFTR introduction and replacement is also of particular interest. Such therapies may be complicated by possible CFTR self-assembly into an oligomer or multimer. RESULTS: Surprisingly, functional CFTR assays in native airway epithelia showed that the most common CFTR mutant, ΔF508-CFTR (ΔF-CFTR), inhibits WT-CFTR when both forms are co-expressed. To examine more mechanistically, both forms of CFTR were transfected transiently in varying amounts into IB3-1 CF human airway epithelial cells and HEK-293 human embryonic kidney cells null for endogenous CFTR protein expression. Increasing amounts of ΔF-CFTR inhibited WT-CFTR protein processing and function in CF human airway epithelial cells but not in heterologous HEK-293 cells. Stably expressed ΔF-CFTR in clones of the non-CF human airway epithelial cell line, CALU-3, also showed reduction in cAMP-stimulated anion secretion and in WT-CFTR processing. An ultimate test of this dominant negative-like effect of ΔF-CFTR on WT-CFTR was the parallel study of two different CF mouse models: the ΔF-CFTR mouse and the bitransgenic CFTR mouse corrected in the gut but null in the lung and airways. WT/ΔF heterozygotes had an intermediate phenotype with regard to CFTR agonist responses in in vivo nasal potential difference (NPD) recordings and in Ussing chamber recordings of short-circuit current (ISC) in vitro on primary tracheal epithelial cells isolated from the same mice. In contrast, CFTR bitransgenic +/- heterozygotes had no difference in their responses versus +/+ wild-type mice. CONCLUSIONS: Taken altogether, these data suggest that ΔF-CFTR and WT-CFTR co-assemble into an oligomeric macromolecular complex in native epithelia and share protein processing machinery and regulation at the level of the endoplasmic reticulum (ER). As a consequence, ΔF-CFTR slows WT-CFTR protein processing and limits its expression and function in the apical membrane of native airway epithelia. Implications of these data for the relative health of CF heterozygous carriers, for CFTR protein processing in native airway epithelia, and for the relative efficacy of different CF therapeutic approaches is significant and is discussed.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Mutação , Mucosa Nasal/metabolismo , Mucosa Respiratória/metabolismo , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Células HEK293 , Heterozigoto , Humanos , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos CFTR , Camundongos Knockout , Domínios PDZ/genética
3.
Biochem Biophys Res Commun ; 387(1): 70-6, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19577582

RESUMO

Diminished proteolytic functionality in the lens may cause cataracts. We have reported that O-GlcNAc is an endogenous inhibitor of the proteasome. We hypothesize that in the lens there is a cause-and-effect relationship between proteasome inhibition by O-GlcNAc, and cataract formation. To demonstrate this, we established novel transgenic mouse models to over-express a dominant-negative form of O-GlcNAcase, GK-NCOAT, in the lens. Expression of GK-NCOAT suppresses removal of O-GlcNAc from proteins, resulting in increased levels of O-GlcNAc in the lenses of our transgenic mice, along with decreased proteasome function. We observed that transgenic mice developed markedly larger cataracts than controls and lens fiber cell denucleation was inhibited. Our study suggests that increased O-GlcNAc in the lens could lead to cataract formation and attenuation of lens fiber cell denucleation by inhibition of proteasome function. These findings may explain why cataract formation is a common complication of diabetes since O-GlcNAc is derived from glucose.


Assuntos
Acetilglucosamina/metabolismo , Catarata/patologia , Diferenciação Celular , Cristalino/citologia , Cristalino/patologia , gama-Cristalinas/metabolismo , Animais , Histona Acetiltransferases/genética , Hialuronoglucosaminidase/genética , Camundongos , Camundongos Transgênicos
4.
Hum Gene Ther ; 16(8): 921-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16076250

RESUMO

Recombinant adeno-associated virus serotype 2 (rAAV2)-based human gene therapy for cystic fibrosis has progressed through a series of preclinical studies and phase I and II clinical trials. This agent has shown an encouraging safety profile, consistent levels of DNA transfer, and positive evidence of short-term clinical improvement in lung function in a prospective, placebo-controlled phase II trial of aerosol administration. Nonetheless, it has been difficult to assess the relationship between its molecular action and the observed clinical improvements, because of the lack of positive results from a highly specific assay for vector mRNA. This issue is further complicated by the fact that the clinical vector utilizes a small cryptic rAAV2 promoter sequence that is less robust for mRNA expression than typical viral promoters. In this paper, we report the results of more sensitive assays performed on primary nasal cells harvested from rAAV2-CFTR gene therapy recipients. These studies demonstrate a correlation between the presence of rAAV2-CFTR vector genomes, CFTR mRNA expression, and cAMP-activated chloride channel function in these cells. The observation of sizeable physiological correction in the face of low mRNA levels may reflect the regulatory role of low levels of CFTR protein as an activator of other chloride channels.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Fibrose Cística/terapia , DNA Viral/análise , Células Epiteliais/fisiologia , Técnicas de Transferência de Genes , Terapia Genética , Adenoviridae , Administração Intranasal , Southern Blotting , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/análise , Perfilação da Expressão Gênica , Vetores Genéticos , Humanos , Cavidade Nasal/citologia , Reação em Cadeia da Polimerase , RNA Mensageiro , Sensibilidade e Especificidade
6.
Biochim Biophys Acta ; 1615(1-2): 7-32, 2003 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12948585

RESUMO

The charge of this invited review is to present a convincing case for the fact that cells release their ATP for physiological reasons. Many of our "purinergic" colleagues as well as ourselves have experienced resistance to this concept, because it is teleologically counter-intuitive. This review serves to integrate the three main tenets of extracellular ATP signaling: ATP release from cells, ATP receptors on cells, and ATP receptor-driven signaling within cells to affect cell or tissue physiology. First principles will be discussed in the Introduction concerning extracellular ATP signaling. All possible cellular mechanisms of ATP release will then be presented. Use of nucleotide and nucleoside scavengers as well as broad-specificity purinergic receptor antagonists will be presented as a method of detecting endogenous ATP release affecting a biological endpoint. Innovative methods of detecting released ATP by adapting luciferase detection reagents or by using "biosensors" will be presented. Because our laboratory has been primarily interested in epithelial cell physiology and pathophysiology for several years, the role of extracellular ATP in regulation of epithelial cell function will be the focus of this review. For ATP release to be physiologically relevant, receptors for ATP are required at the cell surface. The families of P2Y G protein-coupled receptors and ATP-gated P2X receptor channels will be introduced. Particular attention will be paid to P2X receptor channels that mediate the fast actions of extracellular ATP signaling, much like neurotransmitter-gated channels versus metabotropic heptahelical neurotransmitter receptors that couple to G proteins. Finally, fascinating biological paradigms in which extracellular ATP signaling has been implicated will be highlighted. It is the goal of this review to convert and attract new scientists into the exploding field of extracellular nucleotide signaling and to convince the reader that extracellular ATP is indeed a signaling molecule.


Assuntos
Trifosfato de Adenosina/metabolismo , Epitélio/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Canais Iônicos/metabolismo , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X
8.
J Cyst Fibros ; 3(2): 99-117, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15463893

RESUMO

BACKGROUND: Loss of cystic fibrosis transmembrane conductance regulator (CFTR) function in cystic fibrosis (CF) causes dysregulation of multiple ion channels, water channels, and acid-base transporters in epithelia. As such, we hypothesized that dysregulation of many critical ion channels and transporters may cause defects in human airway epithelial cell volume regulation. METHODS: Cell volume, regulatory volume decrease, and its regulation was assessed in real-time via Coulter Counter Multisizer III-driven electronic cell sizing in non-CF, CF, and CFTR-complemented CF human airway epithelial cells. SPQ halide fluorescence assay of hypotonicity-induced chloride efflux provided indirect validation of the cell volume assays. RESULTS: CFTR, via autocrine ATP signaling, governs human airway epithelial cell volume regulation. Non-CF cells and wild-type (WT)-CFTR-transfected CF cells had normal regulatory volume decrease (RVD) responses that were attenuated by blockade of autocrine and paracrine purinergic signaling. In contrast, parental IB3-1 CF cells or IB3-1 cells expressing CFTR mutants (DeltaF508, G551D, and S1455X) failed to RVD. CF cell RVD was rescued by agonists to P2Y G protein-coupled receptors and, more robustly, by agonists to P2X purinergic receptor channels. CONCLUSIONS: Loss of CFTR and CFTR-driven autocrine ATP signaling may underlie defective cell volume regulation and dysregulated ion, water, and acid-base transport in CF airway epithelia.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Receptores Purinérgicos P2/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Trifosfato de Adenosina/metabolismo , Comunicação Autócrina/fisiologia , Brônquios/citologia , Cálcio/metabolismo , Linhagem Celular , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Expressão Gênica , Humanos , Soluções Hipotônicas/farmacologia , Soluções Isotônicas/farmacologia , Mucosa Respiratória/citologia , Transfecção
9.
J Cyst Fibros ; 3 Suppl 2: 203-12, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15463959

RESUMO

Gene delivery systems (GDS) play a central role in the development of gene therapy strategies for Cystic Fibrosis (CF). Further, these systems are important tools in studies with cultured cells and in animal models. In this review, we describe the properties of several viral and synthetic gene delivery systems, and evaluate their possible application in gene therapy of CF. While many gene delivery systems give satisfactory results in cultured or animal studies, none of these systems has been shown to fulfil all the requirements of safety and efficacy for use in CF patients. The intact airway epithelium, the most important target in CF gene therapy, proves to be well protected against invading vector systems.


Assuntos
Fibrose Cística/terapia , Terapia Genética/métodos , Mucosa Respiratória/fisiologia , Adenoviridae , Animais , Comunicação Celular/fisiologia , Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Dependovirus , Vetores Genéticos , Humanos , Lentivirus , Modelos Animais , Vírus Sendai , Transfecção
10.
Eur J Pharm Sci ; 20(1): 27-34, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-13678790

RESUMO

The permeability of human bronchial epithelial cells (16HBE14o(-)) to radiolabelled insulin ([125I]insulin) formulated in the absence or presence of two different saccharide-containing permeability enhancers was investigated. In the absence of either enhancer, mannitol permeability and transepithelial electrical resistance (R(TE)) remained essentially unaffected for the duration of a 2-h experiment. Addition of either 0.125% tetradecylmaltoside (TDM) or 1% dimethyl-beta-cyclodextrin (DMBCD) to the apical surface of cells resulted in increased mannitol permeability and decreased R(TE), suggesting a loosening of cellular tight junctions and a concomitant increase in paracellular movement. Addition of [125I]insulin to the apical side of 16HBE14o(-) cells in the absence or presence of 1% DMBCD resulted in little or no [125I]insulin movement to the basolateral chamber or degradation in the apical chamber. However, in the presence of 0.125% TDM, the amount of intact [125I]insulin remaining in the apical chamber was substantially decreased, while [125I]insulin and 125I-labeled fragments were recovered on the basolateral side of the cells after 2 h. These findings provide evidence that the loosening of the tight junctions between cells achieved with DMBCD is not sufficient to stimulate transepithelial insulin movement, whereas exposure to 0.125% TDM causes an increase in [125I]insulin permeation and degradation.


Assuntos
Adjuvantes Farmacêuticos/farmacologia , Ciclodextrinas/farmacologia , Insulina/farmacocinética , Maltose/análogos & derivados , Maltose/farmacologia , Mucosa Respiratória/metabolismo , beta-Ciclodextrinas , Transporte Biológico , Brônquios/citologia , Brônquios/metabolismo , Células Cultivadas , Humanos , Manitol/metabolismo , Permeabilidade , Mucosa Respiratória/citologia , Fatores de Tempo
11.
Am J Physiol Cell Physiol ; 296(1): C131-41, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987251

RESUMO

Cystic fibrosis (CF) is caused by mutations in the gene producing the cystic fibrosis transmembrane conductance regulator (CFTR). CFTR functions as a Cl(-) channel. Its dysfunction limits Cl(-) secretion and enhances Na+ absorption, leading to viscous mucus in the airway. Ca2+-activated Cl(-) channels (CaCCs) are coexpressed with CFTR in the airway surface epithelia. Increases in cytosolic Ca(2+) activate the epithelial CaCCs, which provides an alternative Cl(-) secretory pathway in CF. We developed a screening assay and screened a library for compounds that could enhance cytoplasmic Ca2+, activate the CaCC, and increase Cl(-) secretion. We found that spiperone, a known antipsychotic drug, is a potent intracellular Ca2+ enhancer and demonstrated that it stimulates intracellular Ca2+, not by acting in its well-known role as an antagonist of serotonin 5-HT2 or dopamine D2 receptors, but through a protein tyrosine kinase-coupled phospholipase C-dependent pathway. Spiperone activates CaCCs, which stimulates Cl(-) secretion in polarized human non-CF and CF airway epithelial cell monolayers in vitro and in CFTR-knockout mice in vivo. In conclusion, we have identified spiperone as a new therapeutic platform for correction of defective Cl(-) secretion in CF via a pathway independent of CFTR.


Assuntos
Brônquios/efeitos dos fármacos , Cálcio/metabolismo , Canais de Cloreto/efeitos dos fármacos , Cloretos/metabolismo , Células Epiteliais/efeitos dos fármacos , Medicamentos para o Sistema Respiratório/farmacologia , Espiperona/farmacologia , Traqueia/efeitos dos fármacos , Animais , Bioensaio , Brônquios/enzimologia , Brônquios/metabolismo , Linhagem Celular , Polaridade Celular , Canais de Cloreto/metabolismo , Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Descoberta de Drogas , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos CFTR , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas , Fatores de Tempo , Traqueia/enzimologia , Traqueia/metabolismo , Fosfolipases Tipo C/metabolismo
12.
Purinergic Signal ; 4(2): 89-92, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18368529

RESUMO

The common theme of this introductory article and the minireviews that follow in this special issue is the concept of microenvironments within tissues and surrounding cells that would be ideal signaling venues for a biologically active purinergic ligand. Collectively, the editors/authors and the other contributing authors agree that nucleotides and nucleosides would be most potent within a confined system. A talented cadre of purinergics has been solicited to discuss purinergic signaling in his or her favorite microenvironment within a given organ or tissue. We are gratified by the large number of original articles that also have successfully navigated the peer review process and are part of this special issue. These concepts are not simply purinergic, but the idea of maximal potency in a tissue microenvironment and surrounding specialized cells within a tissue pertains to any autacoid or paracrine agonist.

13.
Purinergic Signal ; 4(2): 109-24, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18438719

RESUMO

The nephron is the functional unit of the kidney. Blood and plasma are continually filtered within the glomeruli that begin each nephron. Adenosine 5' triphosphate (ATP) and its metabolites are freely filtered by each glomerulus and enter the lumen of each nephron beginning at the proximal convoluted tubule (PCT). Flow rate, osmolality, and other mechanical or chemical stimuli for ATP secretion are present in each nephron segment. These ATP-release stimuli are also different in each nephron segment due to water or salt permeability or impermeability along different luminal membranes of the cells that line each nephron segment. Each of the above stimuli can trigger additional ATP release into the lumen of a nephron segment. Each nephron-lining epithelial cell is a potential source of secreted ATP. Together with filtered ATP and its metabolites derived from the glomerulus, secreted ATP and adenosine derived from cells along the nephron are likely the principal two of several nucleotide and nucleoside candidates for renal autocrine and paracrine ligands within the tubular fluid of the nephron. This minireview discusses the first principles of purinergic signaling as they relate to the nephron and the urinary bladder. The review discusses how the lumen of a renal tubule presents an ideal purinergic signaling microenvironment. The review also illustrates how remodeled and encapsulated cysts in autosomal dominant polycystic kidney disease (ADPKD) and remodeled pseudocysts in autosomal recessive PKD (ARPKD) of the renal collecting duct likely create an even more ideal microenvironment for purinergic signaling. Once trapped in these closed microenvironments, purinergic signaling becomes chronic and likely plays a significant epigenetic and detrimental role in the secondary progression of PKD, once the remodeling of the renal tissue has begun. In PKD cystic microenvironments, we argue that normal purinergic signaling within the lumen of the nephron provides detrimental acceleration of ADPKD once remodeling is complete.

14.
Purinergic Signal ; 4(4): 393-405, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18946723

RESUMO

It is well established that ATP is co-secreted with insulin and zinc from pancreatic beta-cells (beta-cells) in response to elevations in extracellular glucose concentration. Despite this knowledge, the physiological roles of extracellular secreted ATP and zinc are ill-defined. We hypothesized that secreted ATP and zinc are autocrine purinergic signaling molecules that activate P2X purinergic receptor (P2XR) channels expressed by beta-cells to enhance glucose-stimulated insulin secretion (GSIS). To test this postulate, we performed ELISA assays for secreted insulin at fixed time points within a "real-time" assay and confirmed that the physiological insulin secretagogue glucose stimulates secretion of ATP and zinc into the extracellular milieu along with insulin from primary rat islets. Exogenous ATP and zinc alone or together also induced insulin secretion in this model system. Most importantly, the presence of an extracellular ATP scavenger, a zinc chelator, and P2 receptor antagonists attenuated GSIS. Furthermore, mRNA and protein were expressed in immortalized beta-cells and primary islets for a unique subset of P2XR channel subtypes, P2X(2), P2X(3), P2X(4), and P2X(6), which are each gated by extracellular ATP and modulated positively by extracellular zinc. On the basis of these results, we propose that, within endocrine pancreatic islets, secreted ATP and zinc have profound autocrine regulatory influence on insulin secretion via ATP-gated and zinc-modulated P2XR channels.

15.
Purinergic Signal ; 4(2): 155-70, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18368523

RESUMO

Renal epithelial cells release ATP constitutively under basal conditions and release higher quantities of purine nucleotide in response to stimuli. ATP filtered at the glomerulus, secreted by epithelial cells along the nephron, and released serosally by macula densa cells for feedback signaling to afferent arterioles within the glomerulus has important physiological signaling roles within kidneys. In autosomal recessive polycystic kidney disease (ARPKD) mice and humans, collecting duct epithelial cells lack an apical central cilium or express dysfunctional proteins within that monocilium. Collecting duct principal cells derived from an Oak Ridge polycystic kidney (orpk ( Tg737 ) ) mouse model of ARPKD lack a well-formed apical central cilium, thought to be a sensory organelle. We compared these cells grown as polarized cell monolayers on permeable supports to the same cells where the apical monocilium was genetically rescued with the wild-type Tg737 gene that encodes Polaris, a protein essential to cilia formation. Constitutive ATP release under basal conditions was low and not different in mutant versus rescued monolayers. However, genetically rescued principal cell monolayers released ATP three- to fivefold more robustly in response to ionomycin. Principal cell monolayers with fully formed apical monocilia responded three- to fivefold greater to hypotonicity than mutant monolayers lacking monocilia. In support of the idea that monocilia are sensory organelles, intentionally harsh pipetting of medium directly onto the center of the monolayer induced ATP release in genetically rescued monolayers that possessed apical monocilia. Mechanical stimulation was much less effective, however, on mutant orpk collecting duct principal cell monolayers that lacked apical central monocilia. Our data also show that an increase in cytosolic free Ca(2+) primes the ATP pool that is released in response to mechanical stimuli. It also appears that hypotonic cell swelling and mechanical pipetting stimuli trigger release of a common ATP pool. Cilium-competent monolayers responded to flow with an increase in cell Ca(2+) derived from both extracellular and intracellular stores. This flow-induced Ca(2+) signal was less robust in cilium-deficient monolayers. Flow-induced Ca(2+) signals in both preparations were attenuated by extracellular gadolinium and by extracellular apyrase, an ATPase/ADPase. Taken together, these data suggest that apical monocilia are sensory organelles and that their presence in the apical membrane facilitates the formation of a mature ATP secretion apparatus responsive to chemical, osmotic, and mechanical stimuli. The cilium and autocrine ATP signaling appear to work in concert to control cell Ca(2+). Loss of a cilium-dedicated autocrine purinergic signaling system may be a critical underlying etiology for ARPKD and may lead to disinhibition and/or upregulation of multiple sodium (Na(+)) absorptive mechanisms and a resultant severe hypertensive phenotype in ARPKD and, possibly, other diseases.

16.
Am J Physiol Cell Physiol ; 292(4): C1409-16, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17182727

RESUMO

Tg737(orpk) mice have defects in cilia assembly and develop hydrocephalus in the perinatal period of life. Hydrocephalus is progressive and is thought to be initiated by abnormal ion and water transport across the choroid plexus epithelium. The pathology is further aggravated by the slow and disorganized beating of motile cilia on ependymal cells that contribute to decreased cerebrospinal fluid movement through the ventricles. Previously, we demonstrated that the hydrocephalus phenotype is associated with a marked increase in intracellular cAMP levels in choroid plexus epithelium, which is known to have regulatory effects on ion and fluid movement in many secretory epithelia. To evaluate whether the hydrocephalus in Tg737(orpk) mutants is associated with defects in ion transport, we compared the steady-state pH(i) and Na(+)-dependent transport activities of isolated choroid plexus epithelium tissue from Tg737(orpk) mutant and wild-type mice. The data indicate that Tg737(orpk) mutant choroid plexus epithelium have lower pH(i) and higher Na(+)-dependent HCO(3)(-) transport activity compared with wild-type choroid plexus epithelium. In addition, wild-type choroid plexus epithelium could be converted to a mutant phenotype with regard to the activity of Na(+)-dependent HCO(3)(-) transport by addition of dibutyryl-cAMP and mutant choroid plexus epithelium toward the wild-type phenotype by inhibiting PKA activity with H-89. Together, these data suggest that cilia have an important role in regulating normal physiology of choroid plexus epithelium and that ciliary dysfunction in Tg737(orpk) mutants disrupts a signaling pathway leading to elevated intracellular cAMP levels and aberrant regulation of pH(i) and ion transport activity.


Assuntos
Plexo Corióideo/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Animais , Cílios/genética , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Epitélio/metabolismo , Hidrocefalia/genética , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Transporte de Íons , Isoquinolinas/farmacologia , Camundongos , Camundongos Mutantes , Sulfonamidas/farmacologia
17.
Am J Physiol Renal Physiol ; 290(6): F1320-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16396941

RESUMO

Recent genetic analysis has identified a pivotal role of primary cilia in the pathogenesis of polycystic kidney disease (PKD). However, little is known regarding how cilia loss/dysfunction contributes to cyst development. In epithelial cells, changes in apical fluid flow induce cilia-mediated Ca2+ entry via polycystin-2 (PC2), a cation channel. The Oak Ridge Polycystic Kidney (orpk) mouse contains a mutated Tg737 gene that disrupts expression of polaris, a protein required for ciliogenesis. These studies examine the effect of cilia malformation on Ca2+ entry in orpk cilia(-) collecting duct principal cells, and in orpk cells in which wild-type Tg737 was reintroduced, orpk cilia(+). [Ca2+]i was monitored in confluent cell monolayers using fluorescence microscopy. Intrinsic apical Ca2+ entry was measured by Mn2+ quenching and Ca2+ depletion/readdition under flow conditions below the threshold for stimulation. We found that unstimulated apical Ca2+ entry was markedly increased in cilia(-) cells and was sensitive to Gd3+, an inhibitor of PC2. Electrophysiological measurements demonstrate increased abundance of an apical channel, consistent with PC2, in cilia(-) cells. Immunofluorescence studies revealed that PC2, normally expressed on and at the base of cilia in orpk cilia(+) cells, was observed throughout the apical membrane in cilia(-) cells. Furthermore, cilia(-) cells displayed elevated subapical Ca2+ levels measured with the near-membrane Ca2+ indicator FFP-18. We propose that cilia exert a tonic regulatory influence on apical Ca2+ entry, and absence of cilia results in loss of spatial organization of PC2, causing unregulated Ca2+ entry and elevations in subapical [Ca2+], a factor which may contribute to cyst formation.


Assuntos
Cálcio/metabolismo , Cílios/patologia , Túbulos Renais Coletores/ultraestrutura , Rim Policístico Autossômico Dominante/patologia , Animais , Permeabilidade da Membrana Celular , Células Cultivadas , Cílios/fisiologia , Imunofluorescência , Corantes Fluorescentes , Fura-2/análogos & derivados , Gadolínio/farmacologia , Túbulos Renais Coletores/metabolismo , Manganês/metabolismo , Camundongos , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/metabolismo , Proteínas Supressoras de Tumor/genética
18.
Am J Physiol Cell Physiol ; 290(4): C952-63, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16207792

RESUMO

The Tg737 degrees (rpk) autosomal recessive polycystic kidney disease (ARPKD) mouse carries a hypomorphic mutation in the Tg737 gene. Because of the absence of its protein product Polaris, the nonmotile primary monocilium central to the luminal membrane of ductal epithelia, such as the cortical collecting duct (CCD) principal cell (PC), is malformed. Although the functions of the renal monocilium remain elusive, primary monocilia or flagella on neurons act as sensory organelles. Thus we hypothesized that the PC monocilium functions as a cellular sensor. To test this hypothesis, we assessed the contribution of Polaris and cilium structure and function to renal epithelial ion transport electrophysiology. Properties of Tg737 degrees (rpk) mutant CCD PC clones were compared with clones genetically rescued with wild-type Tg737 cDNA. All cells were grown as polarized cell monolayers with similarly high transepithelial resistance on permeable filter supports. Three- to fourfold elevated transepithelial voltage (V(te)) and short-circuit current (I(sc)) were measured in mutant orpk monolayers vs. rescued controls. Pharmacological and cell biological examination of this enhanced electrical end point in mutant monolayers revealed that epithelial Na(+) channels (ENaCs) were upregulated. Amiloride, ENaC-selective amiloride analogs (benzamil and phenamil), and protease inhibitors (aprotinin and leupeptin) attenuated heightened V(te) and I(sc). Higher concentrations of additional amiloride analogs (ethylisopropylamiloride and dimethylamiloride) also revealed inhibition of V(te). Cell culture requirements and manipulations were also consistent with heightened ENaC expression and function. Together, these data suggest that ENaC expression and/or function are upregulated in the luminal membrane of mutant, cilium-deficient orpk CCD PC monolayers vs. cilium-competent controls. When the genetic lesion causes loss or malformation of the monocilium, ENaC-driven Na(+) hyperabsorption may explain the rapid emergence of severe hypertension in a majority of patients with ARPKD.


Assuntos
Cílios/metabolismo , Células Epiteliais , Rim , Rim Policístico Autossômico Recessivo/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Amilorida/análogos & derivados , Amilorida/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Eletrofisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Rim/citologia , Rim/metabolismo , Camundongos , Peptídeo Hidrolases/metabolismo , Rim Policístico Autossômico Recessivo/genética , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
19.
Am J Physiol Cell Physiol ; 289(2): C388-96, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15800050

RESUMO

A postulated therapeutic avenue in cystic fibrosis (CF) is activation of Ca(2+)-dependent Cl(-) channels via stimulation of Ca(2+) entry from extracellular solutions independent of CFTR functional status. We have shown that extracellular zinc and ATP induce a sustained increase in cytosolic Ca(2+) in human airway epithelial cells that translates into stimulation of sustained secretory Cl(-) transport in non-CF and CF human and mouse airway epithelial cells, cell monolayers, and nasal mucosa. On the basis of these studies, the Ca(2+) entry channels most likely involved were P2X purinergic receptor channels. In the present study, molecular and biochemical data show coexpression of P2X(4), P2X(5), and P2X(6) subtypes in non-CF (16HBE14o(-)) and CF (IB3-1) human bronchial epithelial cells. Other P2X receptor Ca(2+) entry channel subtypes are expressed rarely or not at all in airway epithelia, epithelial cell models from other CF-relevant tissues, or vascular endothelia. Novel transient lipid transfection-mediated delivery of small interference RNA fragments specific to P2X(4) and P2X(6) (but not P2X(5)) into IB3-1 CF human airway epithelial cells inhibited extracellular zinc- and ATP-induced Ca(2+) entry markedly in fura-2 Ca(2+) measurements and "knocked down" protein by >65%. These data suggest that multiple P2X receptor Ca(2+) entry channel subtypes are expressed in airway epithelia. P2X(4) and P2X(6) may coassemble on the airway surface as targets for possible therapeutics for CF independent of CFTR genotype.


Assuntos
Cálcio/metabolismo , Interferência de RNA , Receptores Purinérgicos P2/fisiologia , Mucosa Respiratória/metabolismo , Zinco/metabolismo , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Fibrose Cística/metabolismo , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística , Humanos , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/fisiologia , RNA Mensageiro/análise , Receptores Purinérgicos P2X , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Purinergic Signal ; 1(4): 299-310, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18404515

RESUMO

In this review, we focus on two attributes of P2X receptor channel function, one essential and one novel. First, we propose that P2X receptors are extracellular sensors as well as receptors and ion channels. In particular, the large extracellular domain (that comprises 70% of the molecular mass of the receptor channel protein) lends itself to be a cellular sensor. Moreover, its exquisite sensitivity to extracellular pH, ionic strength, and multiple ligands evokes the function of a sensor. Second, we propose that P2X receptors are extracellular zinc receptors as well as receptors for nucleotides. We provide novel data in multiple publications and illustrative data in this invited review to suggest that zinc triggers ATP-independent activation of P2X receptor channel function. In this light, P2X receptors are the cellular site of integration between autocrine and paracrine zinc signaling and autocrine and paracrine purinergic signaling. P2X receptors may sense changes in these ligands as well as in extracellular pH and ionic strength and transduce these sensations via calcium and/or sodium entry and changes in membrane potential.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA