Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Hepatology ; 63(1): 185-96, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26044703

RESUMO

UNLABELLED: Primary sclerosing cholangitis (PSC) is a chronic, idiopathic, fibroinflammatory cholangiopathy. The role of the microbiota in PSC etiopathogenesis may be fundamentally important, yet remains obscure. We tested the hypothesis that germ-free (GF) mutltidrug resistance 2 knockout (mdr2(-/-) ) mice develop a distinct PSC phenotype, compared to conventionally housed (CV) mdr2(-/-) mice. Mdr2(-/-) mice (n = 12) were rederived as GF by embryo transfer, maintained in isolators, and sacrificed at 60 days in parallel with age-matched CV mdr2(-/-) mice. Serum biochemistries, gallbladder bile acids, and liver sections were examined. Histological findings were validated morphometrically, biochemically, and by immunofluorescence microscopy (IFM). Cholangiocyte senescence was assessed by p16(INK4a) in situ hybridization in liver tissue and by senescence-associated ß-galactosidase staining in a culture-based model of insult-induced senescence. Serum biochemistries, including alkaline phosphatase, aspartate aminotransferase, and bilirubin, were significantly higher in GF mdr2(-/-) (P < 0.01). Primary bile acids were similar, whereas secondary bile acids were absent, in GF mdr2(-/-) mice. Fibrosis, ductular reaction, and ductopenia were significantly more severe histopathologically in GF mdr2(-/-) mice (P < 0.01) and were confirmed by hepatic morphometry, hydroxyproline assay, and IFM. Cholangiocyte senescence was significantly increased in GF mdr2(-/-) mice and abrogated in vitro by ursodeoxycholic acid (UDCA) treatment. CONCLUSIONS: GF mdr2(-/-) mice exhibit exacerbated biochemical and histological features of PSC and increased cholangiocyte senescence, a characteristic and potential mediator of progressive biliary disease. UDCA, a commensal microbial metabolite, abrogates senescence in vitro. These findings demonstrate the importance of the commensal microbiota and its metabolites in protecting against biliary injury and suggest avenues for future studies of biomarkers and therapeutic interventions in PSC.


Assuntos
Colangite Esclerosante/etiologia , Microbioma Gastrointestinal/fisiologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Feminino , Masculino , Camundongos , Camundongos Knockout
2.
Lab Invest ; 93(6): 733-43, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23588707

RESUMO

The cholangiopathies are a diverse group of biliary tract disorders, many of which lack effective treatment. Murine models are an important tool for studying their pathogenesis, but existing noninvasive methods for assessing biliary disease in vivo are not optimal. Here we report our experience with using micro-computed tomography (microCT) and nuclear magnetic resonance (MR) imaging to develop a technique for live-mouse cholangiography. Using mdr2 knockout (mdr2KO, a model for primary sclerosing cholangitis (PSC)), bile duct-ligated (BDL), and normal mice, we performed in vivo: (1) microCT on a Siemens Inveon PET/CT scanner and (2) MR on a Bruker Avance 16.4 T spectrometer, using Turbo Rapid Acquisition with Relaxation Enhancement, IntraGate Fast Low Angle Shot, and Half-Fourier Acquisition Single-shot Turbo Spin Echo methods. Anesthesia was with 1.5-2.5% isoflurane. Scans were performed with and without contrast agents (iodipamide meglumine (microCT), gadoxetate disodium (MR)). Dissection and liver histology were performed for validation. With microCT, only the gallbladder and extrahepatic bile ducts were visualized despite attempts to optimize timing, route, and dose of contrast. With MR, the gallbladder, extra-, and intrahepatic bile ducts were well-visualized in mdr2KO mice; the cholangiographic appearance was similar to that of PSC (eg, multifocal strictures) and could be improved with contrast administration. In BDL mice, MR revealed cholangiographically distinct progressive dilation of the biliary tree without ductal irregularity. In normal mice, MR allowed visualization of the gallbladder and extrahepatic ducts, but only marginal visualization of the diminutive intrahepatic ducts. One mouse died during microCT and MR imaging, respectively. Both microCT and MR scans could be obtained in ≤20 min. We, therefore, demonstrate that MR cholangiography can be a useful tool for longitudinal studies of the biliary tree in live mice, whereas microCT yields suboptimal duct visualization despite requiring contrast administration. These findings support further development and application of MR cholangiography to the study of mouse models of PSC and other cholangiopathies.


Assuntos
Doenças dos Ductos Biliares/diagnóstico por imagem , Colangiografia , Animais , Meios de Contraste , Modelos Animais de Doenças , Feminino , Gadolínio DTPA , Imageamento por Ressonância Magnética , Masculino , Camundongos , Microtomografia por Raio-X
3.
Proc Natl Acad Sci U S A ; 104(48): 19138-43, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18024594

RESUMO

Cholangiocytes, epithelial cells lining the biliary tree, have primary cilia extending from their apical membrane into the ductal lumen. Although important in disease, cilia also play a vital role in normal cellular functions. We reported that cholangiocyte cilia are sensory organelles responding to mechanical stimuli (i.e., luminal fluid flow) by alterations in intracellular Ca(2+) and cAMP. Because cholangiocyte cilia are also ideally positioned to detect changes in composition and tonicity of bile, we hypothesized that cilia also function as osmosensors. TRPV4, a Ca(2+)-permeable ion channel, has been implicated in signal transduction of osmotic stimuli. Using purified rat cholangiocytes and perfused intrahepatic bile duct units (IBDUs), we found that TRPV4 is expressed on cholangiocyte cilia, and that hypotonicity induces an increase in intracellular Ca(2+) in a TRPV4-, ciliary-, and extracellular calcium-dependent manner. The osmosensation of luminal tonicity by ciliary TRPV4 induces bicarbonate secretion, the main determinant of ductal bile formation, by a mechanism involving apical ATP release. Furthermore, the activation of TRPV4 in vivo, by its specific agonist, 4alphaPDD, induces an increase in bile flow as well as ATP release and bicarbonate secretion. Our results suggest that cholangiocyte primary cilia play an important role in ductal bile formation by acting as osmosensors.


Assuntos
Bicarbonatos/metabolismo , Ductos Biliares Intra-Hepáticos/citologia , Cílios/metabolismo , Canais de Cátion TRPV/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Bile/metabolismo , Ductos Biliares Intra-Hepáticos/metabolismo , Sinalização do Cálcio , Concentração de Íons de Hidrogênio , Soluções Hipotônicas/farmacologia , Masculino , Concentração Osmolar , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/biossíntese , Canais de Cátion TRPV/genética , Água/metabolismo
4.
BMC Physiol ; 5: 13, 2005 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-16109175

RESUMO

BACKGROUND: Recent data suggest that canalicular bile secretion involves selective expression and coordinated regulation of aquaporins (AQPs), a family of water channels proteins. In order to further characterize the role of AQPs in this process, an in vitro cell system with retained polarity and expression of AQPs and relevant solute transporters involved in bile formation is highly desirable. The WIF-B cell line is a highly differentiated and polarized rat hepatoma/human fibroblast hybrid, which forms abundant bile canalicular structures. This cell line has been reported to be a good in vitro model for studying hepatocyte polarity. RESULTS: Using RT-PCR, immunoblotting and confocal immunofluorescence, we showed that WIF-B cells express the aquaporin water channels that facilitate the osmotically driven water movements in the liver, i.e. AQP8, AQP9, and AQP0; as well as the key solute transporters involved in the generation of canalicular osmotic gradients, i.e., the bile salt export pump Bsep, the organic anion transporter Mrp2 and the chloride bicarbonate exchanger AE2. The subcellular localization of the AQPs and the solute transporters in WIF-B cells was similar to that in freshly isolated rat hepatocytes and in intact liver. Immunofluorescent costaining studies showed intracellular colocalization of AQP8 and AE2, suggesting the possibility that these transporters are expressed in the same population of pericanalicular vesicles. CONCLUSION: The hepatocyte cell line WIF-B retains the expression and subcellular localization of aquaporin water channels as well as key solute transporters for canalicular bile secretion. Thus, these cells can work as a valuable tool for regulatory and mechanistic studies of the biology of bile formation.


Assuntos
Aquaporinas/metabolismo , Polaridade Celular , Hepatócitos/fisiologia , Frações Subcelulares/metabolismo , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Proteínas de Transporte de Ânions/metabolismo , Antiporters/metabolismo , Canalículos Biliares/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Proteínas do Olho/metabolismo , Imunofluorescência , Hepatócitos/metabolismo , Humanos , Células Híbridas , Canais Iônicos/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas SLC4A , Distribuição Tecidual
5.
World J Gastroenterol ; 8(1): 1-4, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11833061

RESUMO

Cholangiocytes-the epithelial cells which line the bile ducts-are increasingly recognized as important transporting epithelia actively involved in the absorption and secretion of water, ions, and solutes. This recognition is due in part to the recent development of new experimental models. New biologic concepts have emerged including the identification and topography of receptors and flux proteins on the apical and/or basolateral membrane which are involved in the molecular mechanisms of ductal bile secretion. Individually isolated and/or perfused bile duct units from livers of rats and mice serve as new,physiologically relevant in vitro models to study cholangiocyte transport. Biliary tree dimensions and novel insights into anatomic remodeling of proliferating bile ducts have emerged from three-dimensional reconstruction using CT scanning and sophisticated software. Moreover, new pathologic concepts have arisen regarding the interaction of cholangiocytes with pathogens such as Cryptosporidium parvum. These concepts and associated methodologies may provide the framework to develop new therapies for the cholangiopathies, a group of important hepatobiliary diseases in which cholangiocytes are the target cell.


Assuntos
Ductos Biliares/citologia , Células Epiteliais/fisiologia , Animais , Transporte Biológico/fisiologia
6.
Curr Opin Gastroenterol ; 22(3): 279-87, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16550043

RESUMO

PURPOSE OF REVIEW: Cholangiocytes are increasingly recognized as biologically important epithelia because of the diverse array of cellular processes in which they participate. Collectively, these processes define normal function and, when disturbed, account for abnormalities that cause disease. Advances in animal models and sophisticated technology in imaging and gene silencing have led to substantial progress in defining the roles that cholangiocytes play in signaling, transport of water, ions and solutes, and alterations that result in cholestasis. The pace of advances in technology justifies a yearly summary to identify trends, and inform the readership of the most significant developments in cholangiocyte biology. RECENT FINDINGS: The main areas of recent progress include insights into the molecular mechanisms of bile secretion and the development of new experimental models and technologies. Major advances have also included the identification of novel roles for receptors and better understanding of mechanistic pathways and biologic processes. SUMMARY: Understanding the key mechanistic and biologic processes in cholangiocytes is required to generate hypotheses and therapies relevant to disease. This compendium of current activities in cholangiocyte biology may promote collegial sharing and exchange of novel concepts, ideas, reagents and probes, thereby promoting positive advances in the field.


Assuntos
Doenças dos Ductos Biliares/patologia , Ductos Biliares/citologia , Animais , Bile/metabolismo , Doenças dos Ductos Biliares/fisiopatologia , Proliferação de Células , Citoproteção , Humanos
7.
Biol Cell ; 98(1): 43-52, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16354161

RESUMO

BACKGROUND INFORMATION: We have previously showed that: (i) cholangiocytes contain AQP1 (aquaporin 1) water channels sequestered in intracellular vesicles; and (ii) upon stimulation with choleretic agonists such as secretin or dibutyryl-cAMP (dbcAMP), the AQP1 vesicles move via microtubules to the apical cholangiocyte membrane to facilitate osmotically driven, passive water movement (i.e. ductal bile secretion). The aim of the present study was to determine which proteins and mechanisms regulate AQP1 trafficking in cholangiocytes. RESULTS: Using polarized cultured NMCs (normal mouse cholangiocytes) or NRCs (normal rat cholangiocytes) and affinity-purified antibodies, we performed immunofluorescent confocal microscopy on fixed cells or immunoblotting on cell lysates for actin, tubulin, kinesin and dynein, proteins known to regulate intracellular vesicle trafficking. By immunostaining, the appropriate orientation of the actin (i.e. sub-apical) and tubulin (i.e. generalized) cytoskeleton was apparent; kinesin and dynein displayed a homogeneous punctate distribution. Immunoblotting showed kinesin and dynein to be present in both cholangiocyte lysates and in isolated AQP1-containing vesicles. We utilized real-time fluorescence confocal microscopy of NMCs transfected with a GFP (green fluorescent protein)-AQP1 fusion construct in the presence and absence of dbcAMP. CONCLUSIONS: Our results provide additional insights into the potential molecular mechanisms of ductal bile secretion.


Assuntos
Aquaporina 1/metabolismo , Ductos Biliares/metabolismo , Proteínas do Citoesqueleto/fisiologia , Animais , Ductos Biliares/citologia , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Dineínas/metabolismo , Cinesinas/metabolismo , Camundongos , Microscopia Confocal , Microtúbulos/fisiologia , Ratos , Transfecção , Vesículas Transportadoras/metabolismo
8.
Am J Physiol Gastrointest Liver Physiol ; 291(3): G500-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16899714

RESUMO

Primary cilia are distinct organelles expressed by many vertebrate cells, including cholangiocytes; however, their functions remain obscure. To begin to explore the physiological role of these organelles in the liver, we described the morphology and structure of cholangiocyte cilia and developed new approaches for their isolation. Primary cilia were present only in bile ducts and were not observed in hepatocytes or in hepatic arterial or portal venous endothelial cells. Each cholangiocyte possesses a single cilium that extends from the apical membrane into the bile duct lumen. In addition, the length of the cilia was proportional to the bile duct diameter. We reproducibly isolated enriched fractions of cilia from normal rat and mouse cholangiocytes by two different approaches as assessed by scanning electron, transmission electron, and confocal microscopy. The purity of isolated ciliary fractions was further analyzed by Western blot analysis using acetylated tubulin as a ciliary marker and P2Y(2) as a nonciliary cell membrane marker. These novel techniques produced enriched ciliary fractions of sufficient purity and quantity for light and electron microscopy and for biochemical analyses. They will permit further assessment of the role of primary cilia in normal and pathological conditions.


Assuntos
Ductos Biliares/ultraestrutura , Cílios/ultraestrutura , Hepatócitos/ultraestrutura , Fígado/ultraestrutura , Animais , Células Cultivadas , Camundongos , Ratos
9.
Cell Microbiol ; 8(12): 1932-45, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16848787

RESUMO

Cryptosporidium parvum attaches to intestinal and biliary epithelial cells via specific molecules on host-cell surface membranes including Gal/GalNAc-associated glycoproteins. Subsequent cellular entry of this parasite depends on host-cell membrane alterations to form a parasitophorous vacuole via activation of phosphatidylinositol 3-kinase (PI-3K)/Cdc42-associated actin remodelling. How C. parvum hijacks these host-cell processes to facilitate its infection of target epithelia is unclear. Using specific probes to known components of sphingolipid-enriched membrane microdomains (SEMs), we detected aggregation of host-cell SEM components at infection sites during C. parvum infection of cultured human biliary epithelial cells (i.e. cholangiocytes). Activation and membrane translocation of acid-sphingomyelinase (ASM), an enzyme involved in SEM membrane aggregation, were also observed in infected cells. Pharmacological disruption of SEMs and knockdown of ASM via a specific small interfering RNA (siRNA) significantly decreased C. parvum attachment (by approximately 84%) and cellular invasion (by approximately 88%). Importantly, knockdown of ASM and disruption of SEMs significantly blocked C. parvum-induced accumulation of Gal/GalNAc-associated glycoproteins at infection sites by approximately 90%. Disruption of SEMs and knockdown of ASM also significantly blocked C. parvum-induced activation of host-cell PI-3K and subsequent accumulation of Cdc42 and actin by up to 75%. Our results suggest an important role of SEMs for C. parvum attachment to and entry of host cells, likely via clustering of membrane-binding molecules and facilitating of C. parvum-induced actin remodelling at infection sites through activation of the PI-3K/Cdc42 signalling pathway.


Assuntos
Ductos Biliares Intra-Hepáticos/parasitologia , Criptosporidiose/metabolismo , Cryptosporidium parvum/patogenicidade , Células Epiteliais/parasitologia , Microdomínios da Membrana/metabolismo , Esfingolipídeos/metabolismo , Actinas/metabolismo , Animais , Ductos Biliares Intra-Hepáticos/citologia , Linhagem Celular , Humanos , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Esfingolipídeos/química , Proteína cdc42 de Ligação ao GTP/metabolismo
10.
Curr Opin Gastroenterol ; 21(3): 337-43, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15818155

RESUMO

PURPOSE OF REVIEW: Cholangiocytes are increasingly recognized as biologically important because of the diverse array of cellular processes in which they participate. Collectively, these processes define normal function and, when disturbed, account for abnormalities that cause disease. Advances in animal models and sophisticated technology in imaging and gene silencing have allowed progress in defining the roles that cholangiocytes play in signaling, transport of water, ions and solutes, and alterations that result in cholestasis. The pace of advances in technology justifies a yearly summary to identify the most important developments in cholangiocyte biology. RECENT FINDINGS: The main areas of recent progress include insights into the molecular mechanisms of bile secretion and the development of new experimental models and technologies. SUMMARY: Understanding the critical components and key biologic processes in cholangiocytes responsible for regulation of ductal bile secretion is an initial and required step in generating hypotheses relevant to disease. With regard to the pathologic relevance of this work, cholestatic liver diseases represent a broad group of hepatobiliary disorders with which hepatologists must deal. In addition to genetic defects, the study of the normal and altered trafficking of cholangiocyte transport systems involved in bile secretion may provide a molecular correlate for the functional changes that occur in disease. Critical to this understanding is the ongoing development of experimental models and techniques to interpret data to answer key hypothesis-driven questions. Second, the collegial sharing and exchange of novel concepts, ideas, reagents, and probes promotes positive advances in the field.


Assuntos
Doenças Biliares/etiologia , Sistema Biliar/citologia , Fenômenos Fisiológicos Celulares , Ácidos e Sais Biliares/fisiologia , Doenças Biliares/fisiopatologia , Humanos
11.
J Immunol ; 175(11): 7447-56, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16301652

RESUMO

Infection of epithelial cells by Cryptosporidium parvum triggers a variety of host-cell innate and adaptive immune responses including release of cytokines/chemokines and up-regulation of antimicrobial peptides. The mechanisms that trigger these host-cell responses are unclear. Thus, we evaluated the role of TLRs in host-cell responses during C. parvum infection of cultured human biliary epithelia (i.e., cholangiocytes). We found that normal human cholangiocytes express all known TLRs. C. parvum infection of cultured cholangiocytes induces the selective recruitment of TLR2 and TLR4 to the infection sites. Activation of several downstream effectors of TLRs including IL-1R-associated kinase, p-38, and NF-kappaB was detected in infected cells. Transfection of cholangiocytes with dominant-negative mutants of TLR2 and TLR4, as well as the adaptor molecule myeloid differentiation protein 88 (MyD88), inhibited C. parvum-induced activation of IL-1R-associated kinase, p-38, and NF-kappaB. Short-interfering RNA to TLR2, TLR4, and MyD88 also blocked C. parvum-induced NF-kappaB activation. Moreover, C. parvum selectively up-regulated human beta-defensin-2 in directly infected cells, and inhibition of TLR2 and TLR4 signals or NF-kappaB activation were each associated with a reduction of C. parvum-induced human beta-defensin-2 expression. A significantly higher number of parasites were detected in cells transfected with a MyD88 dominant-negative mutant than in the control cells at 48-96 h after initial exposure to parasites, suggesting MyD88-deficient cells were more susceptible to infection. These findings demonstrate that cholangiocytes express a variety of TLRs, and suggest that TLR2 and TLR4 mediate cholangiocyte defense responses to C. parvum via activation of NF-kappaB.


Assuntos
Ductos Biliares/imunologia , Cryptosporidium parvum/imunologia , Células Epiteliais/microbiologia , NF-kappa B/metabolismo , Receptores Toll-Like/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos de Diferenciação/imunologia , Antígenos de Diferenciação/metabolismo , Ductos Biliares/citologia , Western Blotting , Células Cultivadas , Ativação Enzimática/imunologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica , Quinases Associadas a Receptores de Interleucina-1 , Microscopia de Fluorescência , Fator 88 de Diferenciação Mieloide , NF-kappa B/imunologia , Proteínas Quinases/metabolismo , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Toll-Like/biossíntese , Transfecção , beta-Defensinas/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Curr Opin Gastroenterol ; 20(3): 270-4, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15703652

RESUMO

PURPOSE OF REVIEW: Cholangiocytes are increasingly recognized as biologically important because of the diversity of cellular processes in which they participate. Collectively, these processes define normal function and, when disturbed, account for abnormalities that cause disease. Advances in animal models of disease, sophistication of technology in imaging, and gene silencing have allowed progress in defining the roles that cholangiocytes play in signaling; transport of water, ions, and solutes; and alterations that result in cholestasis. The pace of advances in technology justifies a yearly summary to identify the most important developments in cholangiocyte biology. RECENT FINDINGS: The main areas of recent progress include insights into the molecular mechanisms of hormone-induced bile secretion, development of new experimental models, and a better understanding of the mechanisms of cholestasis. SUMMARY: Understanding the normal components and key biologic processes in cholangiocytes responsible for the regulation of ductal bile secretion is an initial and required step in generating hypotheses relevant to disease. With regard to the pathologic relevance of this work, cholestatic liver diseases represent a broad group of hepatobiliary disorders with which hepatologists must deal. In addition to genetic defects, the study of the normal and altered trafficking of cholangiocyte transport systems involved in bile secretion may provide a molecular correlate for the functional changes that occur in cholestasis. Crucial to this understanding is the ongoing development of experimental models and techniques to answer key hypothesis-driven questions. Additionally, the collegial sharing and exchange of novel concepts, ideas, reagents, and probes promotes positive advances in the field.

13.
Am J Physiol Cell Physiol ; 283(1): C338-46, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12055103

RESUMO

We previously reported the development of reproducible techniques for isolating and perfusing intact intrahepatic bile duct units (IBDUs) from rats. Given the advantages of transgenic and knockout mice for exploring ductal bile formation, we report here the adaptation of those techniques to mice and their initial application to the study of water transport across mouse intrahepatic biliary epithelia. IBDUs were isolated from livers of normal mice by microdissection combined with enzymatic digestion. After culture, isolated IBDUs sealed to form intact, polarized compartments, and a microperfusion system employing those isolated IBDUs developed. A quantitative image analysis technique was used to observe a rapid increase of luminal area when sealed IBDUs were exposed to a series of inward osmotic gradients reflecting net water secretion; the choleretic agonists secretin and forskolin also induced water secretion into IBDUs. The increase of IBDU luminal area induced by inward osmotic gradients and choleretic agonists was reversibly inhibited by HgCl2, a water channel inhibitor. With the use of a quantitative epifluorescence technique in perfused mouse IBDUs, a high osmotic water permeability (P(f) = 2.5-5.6 x 10(-2) cm/s) was found in response to osmotic gradients, further supporting the presence of water channels. These findings suggest that, as in the rat, water transport across intrahepatic biliary epithelia in mice is water channel mediated.


Assuntos
Aquaporinas/metabolismo , Ductos Biliares Intra-Hepáticos/metabolismo , Água/metabolismo , Animais , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Colagogos e Coleréticos/farmacologia , Colforsina/farmacologia , Técnicas de Cultura , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Cloreto de Mercúrio/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Osmose , Perfusão , Secretina/farmacologia
14.
J Biol Chem ; 279(30): 31671-8, 2004 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-15133042

RESUMO

Cryptosporidium parvum invades target epithelia via a mechanism that involves host cell actin reorganization. We previously demonstrated that C. parvum activates the Cdc42/neural Wiskott-Aldrich syndrome protein network in host cells resulting in actin remodeling at the host cell-parasite interface, thus facilitating C. parvum cellular invasion. Here, we tested the role of phosphatidylinositol 3-kinase (PI3K) and frabin, a guanine nucleotide exchange factor specific for Cdc42 in the activation of Cdc42 during C. parvum infection of biliary epithelial cells. We found that C. parvum infection of cultured human biliary epithelial cells induced the accumulation of PI3K at the host cell-parasite interface and resulted in the activation of PI3K in infected cells. Frabin also was recruited to the host cell-parasite interface, a process inhibited by two PI3K inhibitors, wortmannin and LY294002. The cellular expression of either a dominant negative mutant of PI3K (PI3K-Deltap85) or functionally deficient mutants of frabin inhibited C. parvum-induced Cdc42 accumulation at the host cell-parasite interface. Moreover, LY294002 abolished C. parvum-induced Cdc42 activation in infected cells. Inhibition of PI3K by cellular overexpression of PI3K-Deltap85 or by wortmannin or LY294002, as well as inhibition of frabin by various functionally deficient mutants, decreased C. parvum-induced actin accumulation and inhibited C. parvum cellular invasion. In contrast, the overexpression of the p85 subunit of PI3K promoted C. parvum invasion. Our data suggest that an important component of the complex process of C. parvum invasion of target epithelia results from the ability of the organism to trigger host cell PI3K/frabin signaling to activate the Cdc42 pathway, resulting in host cell actin remodeling at the host cell-parasite interface.


Assuntos
Cryptosporidium parvum/patogenicidade , Proteínas dos Microfilamentos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Androstadienos/farmacologia , Animais , Ductos Biliares/metabolismo , Ductos Biliares/parasitologia , Linhagem Celular , Cromonas/farmacologia , Criptosporidiose/metabolismo , Criptosporidiose/parasitologia , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Morfolinas/farmacologia , Mutação , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais , Wortmanina
15.
J Biol Chem ; 278(44): 43157-62, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12939275

RESUMO

Previous work from our laboratory supports an important role for aquaporins (AQPs), a family of water channel proteins, in bile secretion by hepatocytes. To further define the pathways and molecular mechanisms for water movement across hepatocytes, we directly assessed osmotic water permeability (Pf) and activation energy (Ea) in highly purified, rat hepatocytes basolateral membrane vesicles (BLMV) and canalicular membrane (CMV) vesicles by measuring scattered light intensity using stopped-flow spectrophotometry. The time course of scattered light for BLMV and CMV fit well to a single-exponential function. In BLMV, Pf was 108 +/- 4 mum.s-1 (25 degrees C) with an Ea of 7.7 kcal/mol; in CMV, Pf was 86 +/- 5 mum.s-1 (25 degrees C) with an Ea of 8.0 kcal/mol. The AQP blocker, dimethyl sulfoxide, significantly inhibited the Pf of both basolateral (81 +/- 4 mum.s-1; -25%) and canalicular (59 +/- 4 mum.s-1; -30%) membrane vesicles. When CMV were isolated from hepatocytes treated with dibutyryl cAMP, a double-exponential fit was needed, implying two functionally different vesicle populations; one population had Pf and Ea values similar to those of CMV from untreated hepatocytes, but the other population had a very high Pf (655 +/- 135 mum.s-1, 25 degrees C) and very low Ea (2.8 kcal/mol). Dimethyl sulfoxide completely inhibited the high Pf value in this second vesicle population. In contrast, Pf and Ea of BLMV were unaltered by cAMP treatment of hepatocytes. Our results are consistent with the presence of both lipid- and AQP-mediated pathways for basolateral and canalicular water movement across the hepatocyte plasma membrane barrier. Our data also suggest that the hepatocyte canalicular membrane domain is rate-limiting for transcellular water transport and that this domain becomes more permeable to water when hepatocytes are exposed to a choleretic agonist, presumably by insertion of AQP molecules. These data suggest a molecular mechanism for the efficient coupling of osmotically active solutes and water transport during canalicular bile formation.


Assuntos
Membrana Celular/metabolismo , Hepatócitos/metabolismo , Hepatócitos/fisiologia , Água/metabolismo , Animais , Canalículos Biliares/metabolismo , Transporte Biológico , Permeabilidade da Membrana Celular , Células Cultivadas , Dimetil Sulfóxido/farmacologia , Cinética , Luz , Masculino , Microscopia Eletrônica , Osmose , Estrutura Terciária de Proteína , Ratos , Ratos Endogâmicos F344 , Espalhamento de Radiação , Temperatura , Fatores de Tempo
16.
Am J Physiol Cell Physiol ; 283(3): C785-91, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12176735

RESUMO

The physiological relevance of the absorption of glucose from bile by cholangiocytes remains unclear. The aim of this study was to test the hypothesis that absorbed glucose drives aquaporin (AQP)-mediated water transport by biliary epithelia and is thus involved in ductal bile formation. Glucose absorption and water transport by biliary epithelia were studied in vitro by microperfusing intrahepatic bile duct units (IBDUs) isolated from rat liver. In a separate set of in vivo experiments, bile flow and absorption of biliary glucose were measured after intraportal infusion of D-glucose or phlorizin. IBDUs absorbed D-glucose in a dose- and phlorizin-dependent manner with an absorption maximum of 92.8 +/- 6.2 pmol. min(-1). mm(-1). Absorption of D-glucose by microperfused IBDUs resulted in an increase of water absorption (J(v) = 3-10 nl. min(-1). mm(-1), P(f) = 40 x 10(-3) cm/sec). Glucose-driven water absorption by IBDUs was inhibited by HgCl(2), suggesting that water passively follows absorbed D-glucose mainly transcellularly via mercury-sensitive AQPs. In vivo studies showed that as the amount of absorbed biliary glucose increased after intraportal infusion of D-glucose, bile flow decreased. In contrast, as the absorption of biliary glucose decreased after phlorizin, bile flow increased. Results support the hypothesis that the physiological significance of the absorption of biliary glucose by cholangiocytes is likely related to regulation of ductal bile formation.


Assuntos
Ductos Biliares Intra-Hepáticos/metabolismo , Glucose/metabolismo , Água/metabolismo , Animais , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Relação Dose-Resposta a Droga , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Glucose/farmacologia , Técnicas In Vitro , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Monossacarídeos/antagonistas & inibidores , Proteínas de Transporte de Monossacarídeos/metabolismo , Perfusão , Florizina/farmacologia , Ratos , Transportador 1 de Glucose-Sódio
17.
Am J Physiol Cell Physiol ; 284(5): C1205-14, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12676656

RESUMO

With an in vitro model using enclosed intrahepatic bile duct units (IBDUs) isolated from wild-type and somatostatin receptor (SSTR) subtype 2 knockout mice, we tested the effects of somatostatin, secretin, and a selective SSTR2 agonist (L-779976) on fluid movement across the bile duct epithelial cell layer. By RT-PCR, four of five known subtypes of SSTRs (SSTR1, SSTR2A/2B, SSTR3, and SSTR4, but not SSTR5) were detected in cholangiocytes in wild-type mice. In contrast, SSTR2A/2B were completely depleted in the SSTR2 knockout mice whereas SSTR1, SSTR3 and SSTR4 were expressed in these cholangiocytes. Somatostatin induced a decrease of luminal area of IBDUs isolated from wild-type mice, reflecting net fluid absorption; L-779976 also induced a comparable decrease of luminal area. No significant decrease of luminal area by either somatostatin or L-779976 was observed in IBDUs from SSTR2 knockout mice. Secretin, a choleretic hormone, induced a significant increase of luminal area of IBDUs of wild-type mice, reflecting net fluid secretion; somatostatin and L-779976 inhibited (P < 0.01) secretin-induced fluid secretion. The inhibitory effect of both somatostatin and L-779976 on secretin-induced IBDU secretion was absent in IBDUs of SSTR2 knockout mice. Somatostatin induced an increase of intracellular cGMP and inhibited secretin-stimulated cAMP synthesis in cholangiocytes; depletion of SSTR2 blocked these effects of somatostatin. These data suggest that somatostatin regulates ductal bile formation in mice not only by inhibition of ductal fluid secretion but also by stimulation of ductal fluid absorption via interacting with SSTR2 on cholangiocytes, a process involving the intracellular cAMP/cGMP second messengers.


Assuntos
Ductos Biliares Intra-Hepáticos/metabolismo , Bile/metabolismo , Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Absorção , Amidas/farmacologia , Animais , Ductos Biliares Intra-Hepáticos/citologia , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Líquidos Corporais/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Indóis/farmacologia , Membranas Intracelulares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Receptores de Somatostatina/genética , Secretina/farmacologia , Somatostatina/farmacologia
18.
Hepatology ; 37(6): 1435-41, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12774023

RESUMO

Although glucagon is known to stimulate the cyclic adenosine monophosphate (cAMP)-mediated hepatocyte bile secretion, the precise mechanisms accounting for this choleretic effect are unknown. We recently reported that hepatocytes express the water channel aquaporin-8 (AQP8), which is located primarily in intracellular vesicles, and its relocalization to plasma membranes can be induced with dibutyryl cAMP. In this study, we tested the hypothesis that glucagon induces the trafficking of AQP8 to the hepatocyte plasma membrane and thus increases membrane water permeability. Immunoblotting analysis in subcellular fractions from isolated rat hepatocytes indicated that glucagon caused a significant, dose-dependent increase in the amount of AQP8 in plasma membranes (e.g., 102% with 1 micromol/L glucagon) and a simultaneous decrease in intracellular membranes (e.g., 38% with 1 micromol/L glucagon). Confocal immunofluorescence microscopy in cultured hepatocytes confirmed the glucagon-induced redistribution of AQP8 from intracellular vesicles to plasma membrane. Polarized hepatocyte couplets showed that this redistribution was specifically to the canalicular domain. Glucagon also significantly increased hepatocyte membrane water permeability by about 70%, which was inhibited by the water channel blocker dimethyl sulfoxide (DMSO). The inhibitors of protein kinase A, H-89, and PKI, as well as the microtubule blocker colchicine, prevented the glucagon effect on both AQP8 redistribution to hepatocyte surface and cell membrane water permeability. In conclusion, our data suggest that glucagon induces the protein kinase A and microtubule-dependent translocation of AQP8 water channels to the hepatocyte canalicular plasma membrane, which in turn leads to an increase in membrane water permeability. These findings provide evidence supporting the molecular mechanisms of glucagon-induced hepatocyte bile secretion.


Assuntos
Aquaporinas/metabolismo , Glucagon/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Canais Iônicos , Animais , Transporte Biológico/efeitos dos fármacos , Membrana Celular/metabolismo , Separação Celular , Colchicina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Masculino , Osmose/efeitos dos fármacos , Ratos , Ratos Wistar , Frações Subcelulares/metabolismo , Distribuição Tecidual , Água/metabolismo
19.
J Biol Chem ; 278(22): 20413-9, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12660234

RESUMO

We previously proposed that ductal bile formation is regulated by secretin-responsive relocation of aquaporin 1 (AQP1), a water-selective channel protein, from an intracellular vesicular compartment to the apical membrane of cholangiocytes. In this study, we immunoisolated AQP1-containing vesicles from cholangiocytes prepared from rat liver; quantitative immunoblotting revealed enrichment in these vesicles of not only AQP1 but also cystic fibrosis transmembrane regulator (CFTR) and AE2, a Cl- channel and a Cl-/HCO3- exchanger, respectively. Dual labeled immunogold electron microscopy of cultured polarized mouse cholangiocytes showed significant colocalization of AQP1, CFTR, and AE2 in an intracellular vesicular compartment; exposure of cholangiocytes to dibutyryl-cAMP (100 microm) resulted in co-redistribution of all three proteins to the apical cholangiocyte plasma membrane. After administration of secretin to rats in vivo, bile flow increased, and AQP1, CFTR, and AE2 co-redistributed to the apical cholangiocyte membrane; both events were blocked by pharmacologic disassembly of microtubules. Based on these in vitro and in vivo observations utilizing independent and complementary approaches, we propose that cholangiocytes contain an organelle that sequesters functionally related proteins that can account for ion-driven water transport, that this organelle moves to the apical cholangiocyte membrane in response to secretory agonists, and that these events account for ductal bile secretion at a molecular level.


Assuntos
Proteínas de Transporte de Ânions , Antiporters , Ductos Biliares Intra-Hepáticos/metabolismo , Secretina/farmacologia , Água/metabolismo , Animais , Aquaporina 1 , Aquaporinas/metabolismo , Ductos Biliares Intra-Hepáticos/citologia , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Ductos Biliares Intra-Hepáticos/ultraestrutura , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Transporte de Íons , Masculino , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Transporte Proteico , Ratos , Ratos Endogâmicos F344 , Proteínas SLC4A
20.
Hepatology ; 37(5): 1026-33, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12717383

RESUMO

Hepatocytes express the water channel aquaporin-8 (AQP8), which is mainly localized in intracellular vesicles, and its adenosine 3',5'-cyclic monophosphate (cAMP)-induced translocation to the plasma membrane facilitates osmotic water movement during canalicular bile secretion. Thus, defective expression of AQP8 may be associated with secretory dysfunction of hepatocytes caused by extrahepatic cholestasis. We studied the effect of 1, 3, and 7 days of bile duct ligation (BDL) on protein expression, subcellular localization, and messenger RNA (mRNA) levels of AQP8; this was determined in rat livers by immunoblotting in subcellular membranes, light immunohistochemistry, immunogold electron microscopy, and Northern blotting. One day of BDL did not affect expression or subcellular localization of AQP8. Three days of BDL reduced the amount of intracellular AQP8 (75%; P <.001) without affecting its plasma membrane expression. Seven days after BDL, AQP8 was markedly decreased in intracellular (67%; P <.05) and plasma (56%; P <.05) membranes. Dibutyryl cAMP failed to increase AQP8 in plasma membranes from liver slices, suggesting a defective translocation of AQP8 in 7-day BDL rats. Immunohistochemistry and immunoelectron microscopy in liver sections confirmed the BDL-induced decreased expression of hepatocyte AQP8 in intracellular vesicles and canalicular membranes. AQP8 mRNA expression was unaffected by 1-day BDL but was significantly increased by about 200% in 3- and 7-day BDL rats, indicating a posttranscriptional mechanism for protein level reduction. In conclusion, BDL-induced extrahepatic cholestasis caused posttranscriptional down-regulation of hepatocyte AQP8 protein expression. Defective expression of AQP8 water channels may contribute to bile secretory dysfunction of cholestatic hepatocytes.


Assuntos
Aquaporinas/metabolismo , Colestase Extra-Hepática/metabolismo , Hepatócitos/metabolismo , Canais Iônicos , Animais , Aquaporinas/genética , Ductos Biliares , Colestase Extra-Hepática/fisiopatologia , Regulação para Baixo , Expressão Gênica , Ligadura , Fígado/citologia , Fígado/metabolismo , Masculino , RNA Mensageiro/análise , Ratos , Ratos Wistar , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA