Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Cells ; 11(19)2022 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-36230937

RESUMO

Alpha-2-macroglobulin (A2M) is a protease inhibitor that regulates extracellular matrix (ECM) stability and turnover. Here, we show that A2M is expressed by endothelial cells (ECs) from human eye choroid. We demonstrate that retinal pigment epithelium (RPE)-conditioned medium induces A2M expression specifically in ECs. Experiments using chemical inhibitors, blocking antibodies, and recombinant proteins revealed a key role of VEGF-A in RPE-mediated A2M induction in ECs. Furthermore, incubation of ECs with RPE-conditioned medium reduces matrix metalloproteinase-2 gelatinase activity of culture supernatants, which is partially restored after A2M knockdown in ECs. We propose that dysfunctional RPE or choroidal blood vessels, as observed in retinal diseases such as age-related macular degeneration, may disrupt the crosstalk mechanism we describe here leading to alterations in the homeostasis of choroidal ECM, Bruch's membrane and visual function.


Assuntos
alfa 2-Macroglobulinas Associadas à Gravidez , Epitélio Pigmentado da Retina , Anticorpos Bloqueadores , Meios de Cultivo Condicionados , Células Endoteliais , Feminino , Gelatinases , Humanos , Metaloproteinase 2 da Matriz , Gravidez , Inibidores de Proteases , Proteínas Recombinantes , Fatores de Transcrição , Fator A de Crescimento do Endotélio Vascular
2.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34782457

RESUMO

Lipofuscin granules enclose mixtures of cross-linked proteins and lipids in proportions that depend on the tissue analyzed. Retinal lipofuscin is unique in that it contains mostly lipids with very little proteins. However, retinal lipofuscin also presents biological and physicochemical characteristics indistinguishable from conventional granules, including indigestibility, tendency to cause lysosome swelling that results in rupture or defective functions, and ability to trigger NLRP3 inflammation, a symptom of low-level disruption of lysosomes. In addition, like conventional lipofuscins, it appears as an autofluorescent pigment, considered toxic waste, and a biomarker of aging. Ocular lipofuscin accumulates in the retinal pigment epithelium (RPE), whereby it interferes with the support of the neuroretina. RPE cell death is the primary cause of blindness in the most prevalent incurable genetic and age-related human disorders, Stargardt disease and age-related macular degeneration (AMD), respectively. Although retinal lipofuscin is directly linked to the cell death of the RPE in Stargardt, the extent to which it contributes to AMD is a matter of debate. Nonetheless, the number of AMD clinical trials that target lipofuscin formation speaks for the potential relevance for AMD as well. Here, we show that retinal lipofuscin triggers an atypical necroptotic cascade, amenable to pharmacological intervention. This pathway is distinct from canonic necroptosis and is instead dependent on the destabilization of lysosomes. We also provide evidence that necroptosis is activated in aged human retinas with AMD. Overall, this cytotoxicity mechanism may offer therapeutic targets and markers for genetic and age-related diseases associated with lipofuscin buildups.


Assuntos
Membranas Intracelulares/metabolismo , Lipofuscina/farmacologia , Lisossomos/metabolismo , Necroptose/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Envelhecimento , Oxirredutases do Álcool , Animais , Morte Celular , Humanos , Lipofuscina/metabolismo , Degeneração Macular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo
3.
Sci Rep ; 11(1): 16323, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381080

RESUMO

Bulk RNA sequencing of a tissue captures the gene expression profile from all cell types combined. Single-cell RNA sequencing identifies discrete cell-signatures based on transcriptomic identities. Six adult human corneas were processed for single-cell RNAseq and 16 cell clusters were bioinformatically identified. Based on their transcriptomic signatures and RNAscope results using representative cluster marker genes on human cornea cross-sections, these clusters were confirmed to be stromal keratocytes, endothelium, several subtypes of corneal epithelium, conjunctival epithelium, and supportive cells in the limbal stem cell niche. The complexity of the epithelial cell layer was captured by eight distinct corneal clusters and three conjunctival clusters. These were further characterized by enriched biological pathways and molecular characteristics which revealed novel groupings related to development, function, and location within the epithelial layer. Moreover, epithelial subtypes were found to reflect their initial generation in the limbal region, differentiation, and migration through to mature epithelial cells. The single-cell map of the human cornea deepens the knowledge of the cellular subsets of the cornea on a whole genome transcriptional level. This information can be applied to better understand normal corneal biology, serve as a reference to understand corneal disease pathology, and provide potential insights into therapeutic approaches.


Assuntos
Córnea/citologia , Adulto , Diferenciação Celular/fisiologia , Túnica Conjuntiva/citologia , Córnea/patologia , Doenças da Córnea/patologia , Células Epiteliais/citologia , Epitélio Corneano/citologia , Humanos , Limbo da Córnea/citologia , Análise de Sequência de RNA/métodos , Nicho de Células-Tronco/fisiologia , Células-Tronco/citologia , Transcriptoma/fisiologia
4.
FASEB J ; 35(7): e21689, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34085737

RESUMO

Knockout of the chloride channel protein 2 (CLC-2; CLCN2) results in fast progressing blindness in mice. Retinal Pigment Epithelium (RPE) and photoreceptors undergo, in parallel, rapid, and profound morphological changes and degeneration. Immunohistochemistry and electron microscopy of the outer retina and electroretinography of the CLC-2 KO mouse demonstrated normal morphology at postnatal day 2, followed by drastic changes in RPE and photoreceptor morphology and loss of vision during the first postnatal month. To investigate whether the RPE or the photoreceptors are the primary cause of the degeneration, we injected lentiviruses carrying HA-tagged CLC-2 with an RPE-specific promotor in the subretinal space of CLC-2-KO mice at the time of eye opening. As expected, CLC-2-HA was expressed exclusively in RPE; strikingly, this procedure rescued the degeneration of both RPE and photoreceptors. Light response in transduced eyes was also recovered. Only a fraction of RPE was transduced with the lentivirus; however, the entire RPE monolayer appears healthy, even the RPE cells not expressing the CLC-2-HA. Surprisingly, in contrast with previous physiological observations that postulate that CLC-2 has a basolateral localization in RPE, our immunofluorescence experiments demonstrated CLC-2 has an apical distribution, facing the subretinal space and the photoreceptor outer segments. Our findings suggest that CLC-2 does not play the postulated role in fluid transport at the basolateral membrane. Rather, they suggest that CLC-2 performs a critical homeostatic role in the subretinal compartment involving a chloride regulatory mechanism that is critical for the survival of both RPE and photoreceptors.


Assuntos
Canais de Cloreto/fisiologia , Células Fotorreceptoras/citologia , Retina/citologia , Degeneração Retiniana , Epitélio Pigmentado da Retina/metabolismo , Animais , Canais de Cloro CLC-2 , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Fotorreceptoras/metabolismo , Retina/metabolismo
5.
J Exp Med ; 217(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32196081

RESUMO

The activity and survival of retinal photoreceptors depend on support functions performed by the retinal pigment epithelium (RPE) and on oxygen and nutrients delivered by blood vessels in the underlying choroid. By combining single-cell and bulk RNA sequencing, we categorized mouse RPE/choroid cell types and characterized the tissue-specific transcriptomic features of choroidal endothelial cells. We found that choroidal endothelium adjacent to the RPE expresses high levels of Indian Hedgehog and identified its downstream target as stromal GLI1+ mesenchymal stem cell-like cells. In vivo genetic impairment of Hedgehog signaling induced significant loss of choroidal mast cells, as well as an altered inflammatory response and exacerbated visual function defects after retinal damage. Our studies reveal the cellular and molecular landscape of adult RPE/choroid and uncover a Hedgehog-regulated choroidal immunomodulatory signaling circuit. These results open new avenues for the study and treatment of retinal vascular diseases and choroid-related inflammatory blinding disorders.


Assuntos
Corioide/imunologia , Corioide/patologia , Endotélio/imunologia , Imunomodulação , Análise de Célula Única , Animais , Proliferação de Células , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Inflamação/genética , Mastócitos/metabolismo , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteína GLI1 em Dedos de Zinco/metabolismo
6.
Nat Commun ; 8: 15374, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28524846

RESUMO

The outer blood-retina barrier is established through the coordinated terminal maturation of the retinal pigment epithelium (RPE), fenestrated choroid endothelial cells (ECs) and Bruch's membrane, a highly organized basement membrane that lies between both cell types. Here we study the contribution of choroid ECs to this process by comparing their gene expression profile before (P5) and after (P30) the critical postnatal period when mice acquire mature visual function. Transcriptome analyses show that expression of extracellular matrix-related genes changes dramatically over this period. Co-culture experiments support the existence of a novel regulatory pathway: ECs secrete factors that remodel RPE basement membrane, and integrin receptors sense these changes triggering Rho GTPase signals that modulate RPE tight junctions and enhance RPE barrier function. We anticipate our results will spawn a search for additional roles of choroid ECs in RPE physiology and disease.


Assuntos
Membrana Basal/metabolismo , Lâmina Basilar da Corioide/metabolismo , Matriz Extracelular/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Junções Íntimas/metabolismo , Animais , Biotinilação , Barreira Hematorretiniana/metabolismo , Adesão Celular , Sobrevivência Celular , Células Cultivadas , Corioide/metabolismo , Técnicas de Cocultura , Eletrorretinografia , Feminino , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Permeabilidade , Proteína-Lisina 6-Oxidase/metabolismo , RNA Mensageiro/metabolismo , Análise de Sequência de RNA
7.
Artigo em Inglês | MEDLINE | ID: mdl-28003183

RESUMO

Directional fluid flow is an essential process for embryo development as well as for organ and organism homeostasis. Here, we review the diverse structure of various organ-blood barriers, the driving forces, transporters, and polarity mechanisms that regulate fluid transport across them, focusing on kidney-, eye-, and brain-blood barriers. We end by discussing how cross talk between barrier epithelial and endothelial cells, perivascular cells, and basement membrane signaling contribute to generate and maintain organ-blood barriers.


Assuntos
Transporte Biológico/fisiologia , Polaridade Celular , Células Endoteliais/citologia , Matriz Extracelular/metabolismo , Homeostase , Humanos , Transdução de Sinais
8.
Hepatology ; 64(2): 535-48, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26999313

RESUMO

UNLABELLED: The adenoviral gene transfer of human aquaporin-1 (hAQP1) water channels to the liver of 17α-ethinylestradiol-induced cholestatic rats improves bile flow, in part by enhancing canalicular hAQP1-mediated osmotic water secretion. To gain insight into the mechanisms of 17α-ethinylestradiol cholestasis improvement, we studied the biliary output of bile salts (BS) and the functional expression of the canalicular BS export pump (BSEP; ABCB11). Adenovector encoding hAQP1 (AdhAQP1) or control vector was administered by retrograde intrabiliary infusion. AdhAQP1-transduced cholestatic rats increased the biliary output of major endogenous BS (50%-80%, P < 0.05) as well as that of taurocholate administered in choleretic or trace radiolabel amounts (around 60%, P < 0.05). Moreover, liver transduction with AdhAQP1 normalized serum BS levels, otherwise markedly elevated in cholestatic animals. AdhAQP1 treatment was unable to improve BSEP protein expression in cholestasis; however, its transport activity, assessed by adenosine triphosphate-dependent taurocholate transport in canalicular membrane vesicles, was induced by 90% (P < 0.05). AdhAQP1 administration in noncholestatic rats induced no significant changes in either biliary BS output or BSEP activity. Canalicular BSEP, mostly present in raft (high cholesterol) microdomains in control rats, was largely found in nonraft (low cholesterol) microdomains in cholestasis. Considering that BSEP activity directly depends on canalicular membrane cholesterol content, decreased BSEP presence in rafts may contribute to BSEP activity decline in 17α-ethinylestradiol cholestasis. In AdhAQP1-transduced cholestatic rats, BSEP showed a canalicular microdomain distribution similar to that of control rats, which provides an explanation for the improved BSEP activity. CONCLUSION: Hepatocyte canalicular expression of hAQP1 through adenoviral gene transfer promotes biliary BS output by modulating BSEP activity in estrogen-induced cholestasis, a novel finding that might help us to better understand and treat cholestatic disorders. (Hepatology 2016;64:535-548).


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Aquaporina 1/genética , Ácidos e Sais Biliares/metabolismo , Colestase/terapia , Terapia Genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Animais , Aquaporina 1/metabolismo , Ácidos e Sais Biliares/sangue , Canalículos Biliares/metabolismo , Colestase/induzido quimicamente , Etinilestradiol , Hepatócitos/metabolismo , Humanos , Masculino , Ratos Wistar
9.
Mol Biol Cell ; 26(9): 1728-42, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25739457

RESUMO

In spite of the many key cellular functions of chloride channels, the mechanisms that mediate their subcellular localization are largely unknown. ClC-2 is a ubiquitous chloride channel usually localized to the basolateral domain of epithelia that regulates cell volume, ion transport, and acid-base balance; mice knocked out for ClC-2 are blind and sterile. Previous work suggested that CLC-2 is sorted basolaterally by TIFS(812)LL, a dileucine motif in CLC-2's C-terminal domain. However, our in silico modeling of ClC-2 suggested that this motif was buried within the channel's dimerization interface and identified two cytoplasmically exposed dileucine motifs, ESMI(623)LL and QVVA(635)LL, as candidate sorting signals. Alanine mutagenesis and trafficking assays support a scenario in which ESMI(623)LL acts as the authentic basolateral signal of ClC-2. Silencing experiments and yeast three-hybrid assays demonstrated that both ubiquitous (AP-1A) and epithelium-specific (AP-1B) forms of the tetrameric clathrin adaptor AP-1 are capable of carrying out basolateral sorting of ClC-2 through interactions of ESMI(623)LL with a highly conserved pocket in their γ1-σ1A hemicomplex.


Assuntos
Complexo 1 de Proteínas Adaptadoras/metabolismo , Canais de Cloreto/metabolismo , Complexo 1 de Proteínas Adaptadoras/química , Motivos de Aminoácidos , Animais , Canais de Cloro CLC-2 , Canais de Cloreto/química , Cães , Células Madin Darby de Rim Canino , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico
10.
Exp Eye Res ; 126: 5-15, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25152359

RESUMO

The retinal pigment epithelium (RPE) comprises a monolayer of polarized pigmented epithelial cells that is strategically interposed between the neural retina and the fenestrated choroid capillaries. The RPE performs a variety of vectorial transport functions (water, ions, metabolites, nutrients and waste products) that regulate the composition of the subretinal space and support the functions of photoreceptors (PRs) and other cells in the neural retina. To this end, RPE cells display a polarized distribution of channels, transporters and receptors in their plasma membrane (PM) that is remarkably different from that found in conventional extra-ocular epithelia, e.g. intestine, kidney, and gall bladder. This characteristic PM protein polarity of RPE cells depends on the interplay of sorting signals in the RPE PM proteins and sorting mechanisms and biosynthetic/recycling trafficking routes in the RPE cell. Although considerable progress has been made in our understanding of the RPE trafficking machinery, most available data have been obtained from immortalized RPE cell lines that only partially maintain the RPE phenotype and by extrapolation of data obtained in the prototype Madin-Darby Canine Kidney (MDCK) cell line. The increasing availability of RPE cell cultures that more closely resemble the RPE in vivo together with the advent of advanced live imaging microscopy techniques provides a platform and an opportunity to rapidly expand our understanding of how polarized protein trafficking contributes to RPE PM polarity.


Assuntos
Membrana Celular/fisiologia , Polaridade Celular , Proteínas de Membrana/fisiologia , Epitélio Pigmentado da Retina/fisiologia , Animais , Transporte Biológico/fisiologia , Polaridade Celular/fisiologia , Cães , Humanos , Sinais Direcionadores de Proteínas/fisiologia , Epitélio Pigmentado da Retina/citologia
11.
Proc Natl Acad Sci U S A ; 111(14): E1402-8, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24706818

RESUMO

Accumulation of lipofuscin bisretinoids (LBs) in the retinal pigment epithelium (RPE) is the alleged cause of retinal degeneration in genetic blinding diseases (e.g., Stargardt) and a possible etiological agent for age-related macular degeneration. Currently, there are no approved treatments for these diseases; hence, agents that efficiently remove LBs from RPE would be valuable therapeutic candidates. Here, we show that beta cyclodextrins (ß-CDs) bind LBs and protect them against oxidation. Computer modeling and biochemical data are consistent with the encapsulation of the retinoid arms of LBs within the hydrophobic cavity of ß-CD. Importantly, ß-CD treatment reduced by 73% and 48% the LB content of RPE cell cultures and of eyecups obtained from Abca4-Rdh8 double knock-out (DKO) mice, respectively. Furthermore, intravitreal administration of ß-CDs reduced significantly the content of bisretinoids in the RPE of DKO animals. Thus, our results demonstrate the effectiveness of ß-CDs to complex and remove LB deposits from RPE cells and provide crucial data to develop novel prophylactic approaches for retinal disorders elicited by LBs.


Assuntos
Lipofuscina/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Retinoides/metabolismo , beta-Ciclodextrinas/metabolismo , Animais , Sítios de Ligação , Cromatografia Líquida de Alta Pressão , Simulação por Computador , Fluorescência , Técnicas In Vitro , Lipofuscina/isolamento & purificação , Camundongos , Camundongos Knockout , Oxirredução , Retinoides/isolamento & purificação
12.
FEBS Lett ; 588(9): 1686-91, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24642373

RESUMO

We recently reported that hepatocyte mitochondrial aquaporin-8 (mtAQP8) channels facilitate the uptake of ammonia and its metabolism into urea. Here we studied the effect of bacterial lipopolysaccharides (LPS) on ammonia-derived ureagenesis. In LPS-treated rats, hepatic mtAQP8 protein expression and diffusional ammonia permeability (measured utilizing ammonia analogues) of liver inner mitochondrial membranes were downregulated. NMR studies using 15N-labeled ammonia indicated that basal and glucagon-induced ureagenesis from ammonia were significantly reduced in hepatocytes from LPS-treated rats. Our data suggest that hepatocyte mtAQP8-mediated ammonia removal via ureagenesis is impaired by LPS, a mechanism potentially relevant to the molecular pathogenesis of defective hepatic ammonia detoxification in sepsis.


Assuntos
Amônia/metabolismo , Aquaporinas/metabolismo , Hepatócitos/metabolismo , Lipopolissacarídeos/farmacologia , Mitocôndrias Hepáticas/metabolismo , Ureia/metabolismo , Animais , Transporte Biológico , Células Cultivadas , Glucagon/fisiologia , Hepatócitos/imunologia , Masculino , Metilaminas/metabolismo , Mitocôndrias Hepáticas/imunologia , Ratos , Ratos Wistar
13.
Gastroenterology ; 139(1): 304-14.e2, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20399209

RESUMO

BACKGROUND & AIMS: In polycystic liver diseases, cyst formation involves cholangiocyte hyperproliferation. In polycystic kidney (PCK) rats, an animal model of autosomal-recessive polycystic kidney disease (ARPKD), decreased intracellular calcium [Ca(2+)](i) in cholangiocytes is associated with hyperproliferation. We recently showed transient receptor potential vanilloid 4 (Trpv4), a calcium-entry channel, is expressed in normal cholangiocytes and its activation leads to [Ca(2+)](i) increase. Thus, we hypothesized that pharmacologic activation of Trpv4 might reverse the hyperproliferative phenotype of PCK cholangiocytes. METHODS: Trpv4 expression was examined in liver of normal and PCK rats, normal human beings, and patients with autosomal-dominant polycystic kidney disease or ARPKD. Trpv4 activation effect on cell proliferation and cyst formation was assessed in cholangiocytes derived from normal and PCK rats. The in vivo effects of Trpv4 activation on kidney and liver cysts was analyzed in PCK rats. RESULTS: Trpv4 was overexpressed both at messenger RNA (8-fold) and protein (3-fold) levels in PCK cholangiocytes. Confocal and immunogold electron microscopy supported Trpv4 overexpression in the livers of PCK rats and ARPKD or autosomal-dominant polycystic kidney disease patients. Trpv4 activation in PCK cholangiocytes increased [Ca(2+)](i) by 30%, inhibiting cell proliferation by approximately 25%-50% and cyst growth in 3-dimensional culture (3-fold). Trpv4-small interfering RNA silencing blocked effects of Trpv4 activators by 70%. Trpv4 activation was associated with Akt phosphorylation and beta-Raf and Erk1/2 inhibition. In vivo, Trpv4 activation induced a significant decrease in renal cystic area and a nonsignificant decrease in liver cysts. CONCLUSIONS: Taken together, our in vitro and in vivo data suggest that increasing intracellular calcium by Trpv4 activation may represent a potential therapeutic approach in PKD.


Assuntos
Ductos Biliares/citologia , Rim Policístico Autossômico Recessivo/terapia , Canais de Cátion TRPV/fisiologia , Animais , Cálcio/metabolismo , Proliferação de Células , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Humanos , Leucina/análogos & derivados , Leucina/farmacologia , Fenótipo , Ésteres de Forbol/farmacologia , Rim Policístico Autossômico Recessivo/patologia , Proteínas Proto-Oncogênicas B-raf/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
14.
Am J Physiol Gastrointest Liver Physiol ; 296(1): G93-100, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18948439

RESUMO

Aquaporin-8 (AQP8) water channels, which are expressed in rat hepatocyte bile canalicular membranes, are involved in water transport during bile formation. Nevertheless, there is no conclusive evidence that AQP8 mediates water secretion into the bile canaliculus. In this study, we directly evaluated whether AQP8 gene silencing by RNA interference inhibits canalicular water secretion in the human hepatocyte-derived cell line, HepG2. By RT-PCR and immunoblotting we found that HepG2 cells express AQP8 and by confocal immunofluorescence microscopy that it is localized intracellularly and on the canalicular membrane, as described in rat hepatocytes. We also verified the expression of AQP8 in normal human liver. Forty-eight hours after transfection of HepG2 cells with RNA duplexes targeting two different regions of human AQP8 molecule, the levels of AQP8 protein specifically decreased by 60-70%. We found that AQP8 knockdown cells showed a significant decline in the canalicular volume of approximately 70% (P < 0.01), suggesting an impairment in the basal (nonstimulated) canalicular water movement. We also found that the decreased AQP8 expression inhibited the canalicular water transport in response either to an inward osmotic gradient (-65%, P < 0.05) or to the bile secretory agonist dibutyryl cAMP (-80%, P < 0.05). Our data suggest that AQP8 plays a major role in water transport across canalicular membrane of HepG2 cells and support the notion that defective expression of AQP8 causes bile secretory dysfunction in human hepatocytes.


Assuntos
Aquaporinas/metabolismo , Canalículos Biliares/metabolismo , Técnicas de Silenciamento de Genes , Hepatócitos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Água/metabolismo , Aquaporinas/genética , Canalículos Biliares/efeitos dos fármacos , Linhagem Celular Tumoral , CMP Cíclico/análogos & derivados , CMP Cíclico/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Microscopia Confocal , Osmose , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção
16.
World J Gastroenterol ; 14(46): 7059-67, 2008 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-19084912

RESUMO

This review focuses on current knowledge on hepatocyte aquaporins (AQPs) and their significance in bile formation and cholestasis. Canalicular bile secretion results from a combined interaction of several solute transporters and AQP water channels that facilitate water flow in response to the osmotic gradients created. During choleresis, hepatocytes rapidly increase their canalicular membrane water permeability by modulating the abundance of AQP8. The question was raised as to whether the opposite process, i.e. a decreased canalicular AQP8 expression would contribute to the development of cholestasis. Studies in several experimental models of cholestasis, such as extrahepatic obstructive cholestasis, estrogen-induced cholestasis, and sepsis-induced cholestasis demonstrated that the protein expression of hepatocyte AQP8 was impaired. In addition, biophysical studies in canalicular plasma membranes revealed decreased water permeability associated with AQP8 protein downregulation. The combined alteration in hepatocyte solute transporters and AQP8 would hamper the efficient coupling of osmotic gradients and canalicular water flow. Thus cholestasis may result from a mutual occurrence of impaired solute transport and decreased water permeability.


Assuntos
Aquaporinas/fisiologia , Colestase Intra-Hepática/fisiopatologia , Aquaporinas/metabolismo , Bile/metabolismo , Colestase Intra-Hepática/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Osmose/fisiologia
17.
Am J Physiol Gastrointest Liver Physiol ; 294(2): G567-75, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18174273

RESUMO

Although bacterial lipopolysaccharides (LPS) are known to cause cholestasis in sepsis, the molecular mechanisms accounting for this effect are only partially known. Because aquaporin-8 (AQP8) seems to facilitate the canalicular osmotic water movement during hepatocyte bile formation, we studied its gene and functional expression in LPS-induced cholestasis. By subcellular fractionation and immunoblotting analysis, we found that 34-kDa AQP8 was significantly decreased by 70% in plasma (canalicular) and intracellular (vesicular) liver membranes. However, expression and subcellular localization of hepatocyte sinusoidal AQP9 were unaffected. Immunohistochemistry for liver AQPs confirmed these observations. Osmotic water permeability (P(f)) of canalicular membranes, measured by stopped-flow spectrophotometry, was significantly reduced (65 +/- 1 vs. 49 +/- 1 microm/s) by LPS, consistent with defective canalicular AQP8 functional expression. By Northern blot analysis, we found that 1.5-kb AQP8 mRNA expression was increased by 80%, suggesting a posttranscriptional mechanism of protein reduction. The tumor necrosis factor-alpha (TNF-alpha) receptor fusion protein TNFp75:Fc prevented the LPS-induced impairment of AQP8 expression and bile flow, suggesting the cytokine TNF-alpha as a major mediator of LPS effect. Accordingly, studies in hepatocyte primary cultures indicated that recombinant TNF-alpha downregulated AQP8. The effect of TNF-alpha was prevented by the lysosomal protease inhibitors leupeptin or chloroquine or by the proteasome inhibitors MG132 or lactacystin, suggesting a cytokine-induced AQP8 proteolysis. In conclusion, our data suggest that LPS induces the TNF-alpha-mediated posttranscriptional downregulation of AQP8 functional expression in hepatocytes, a mechanism potentially relevant to the molecular pathogenesis of sepsis-associated cholestasis.


Assuntos
Aquaporinas/fisiologia , Colestase/etiologia , Colestase/fisiopatologia , Lipopolissacarídeos/farmacologia , Fígado/metabolismo , Sepse/complicações , Sepse/fisiopatologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Northern Blotting , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citocinas/sangue , Regulação para Baixo/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Imuno-Histoquímica , Luz , Fígado/efeitos dos fármacos , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Espalhamento de Radiação , Frações Subcelulares/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/sangue
18.
Am J Physiol Gastrointest Liver Physiol ; 292(3): G905-12, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17110522

RESUMO

Our previous work supports a role for aquaporin-8 (AQP8) water channels in rat hepatocyte bile formation mainly by facilitating the osmotically driven canalicular secretion of water. In this study, we tested whether a condition with compromised canalicular bile secretion, i.e., the estrogen-induced intrahepatic cholestasis, displays defective hepatocyte AQP8 functional expression. After 17alpha-ethinylestradiol administration (5 mg x kg body wt(-1).day(-1) for 5 days) to rats, the bile flow was reduced by 58% (P < 0.05). By subcellular fractionation and immunoblotting analysis, we found that 34 kDa AQP8 was significantly decreased by approximately 70% in plasma (canalicular) and intracellular (vesicular) liver membranes. However, 17alpha-ethinylestradiol-induced cholestasis did not significantly affect the protein level or the subcellular localization of sinusoidal AQP9. Immunohistochemistry for liver AQPs confirmed these observations. Osmotic water permeability (P(f)) of canalicular membranes, measured by stopped-flow spectrophotometry, was significantly reduced (73 +/- 1 vs. 57 +/- 2 microm/s) in cholestasis, consistent with defective canalicular AQP8 functional expression. By Northern blotting, we found that AQP8 mRNA expression was increased by 115% in cholestasis, suggesting a posttranscriptional mechanism of protein level reduction. Accordingly, studies in primary cultured rat hepatocytes indicated that the lysosomal protease inhibitor leupeptin prevented the estrogen-induced AQP8 downregulation. In conclusion, hepatocyte AQP8 protein expression is downregulated in estrogen-induced intrahepatic cholestasis, presumably by lysosomal-mediated degradation. Reduced canalicular membrane AQP8 expression is associated with impaired osmotic membrane water permeability. Our data support the novel notion that a defective expression of canalicular AQP8 contributes as a mechanism for bile secretory dysfunction of cholestatic hepatocytes.


Assuntos
Aquaporinas/metabolismo , Permeabilidade da Membrana Celular/fisiologia , Colestase/metabolismo , Hepatócitos/metabolismo , Água/metabolismo , Animais , Aquaporinas/análise , Aquaporinas/genética , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colestase/induzido quimicamente , Colestase/fisiopatologia , Inibidores de Cisteína Proteinase/farmacologia , Regulação para Baixo/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios , Etinilestradiol/farmacologia , Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Leupeptinas/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Lisossomos/metabolismo , Masculino , Ratos , Ratos Wistar
19.
Biol Cell ; 97(11): 831-6, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15859947

RESUMO

BACKGROUND INFORMATION: PI3K (phosphoinositide 3-kinase) mediates several signal transduction pathways in hepatocytes, including some involved in the regulation of vesicle trafficking. Hepatocytes express the water channel AQP8 (aquaporin-8) predominantly in an intracellular location, and it redistributes to the canalicular membrane, upon stimulation with the hormone glucagon, by a cAMP/protein kinase A-dependent mechanism. Since glucagon is capable of stimulating PI3K activity in hepatocytes and a cross talk between cAMP and PI3K has been suggested, in the present study, we examine whether PI3K activation is involved in the glucagon-induced translocation of AQP8. RESULTS: By quantitative immunoblotting of purified hepatocyte plasma membranes, we found that the preincubation of cells with two structurally different PI3K inhibitors, wortmannin or LY294002, prevented the glucagon-induced translocation of AQP8 to hepatocyte plasma membrane. Confocal immunofluorescence microscopy in cultured hepatocytes confirmed the dependence of the hormone-induced redistribution of AQP8 on PI3K activity. Functional studies showed that the PI3K inhibitors were also capable of preventing the glucagon-induced increase in hepatocyte osmotic membrane water permeability. CONCLUSIONS: Our results suggest that PI3K activation is involved in the glucagon-dependent signal transduction pathways leading to hepatocyte AQP8 translocation.


Assuntos
Aquaporinas/metabolismo , Glucagon/farmacologia , Hepatócitos/metabolismo , Canais Iônicos/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Transporte Proteico/efeitos dos fármacos , Androstadienos/farmacologia , Animais , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cromonas/farmacologia , Hepatócitos/efeitos dos fármacos , Masculino , Morfolinas/farmacologia , Ratos , Ratos Wistar , Wortmanina
20.
Curr Neurovasc Res ; 1(4): 293-303, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16181079

RESUMO

Aquaporins (AQPs) are a family of water-selective channels that provide a major pathway for osmotically driven water transport through cell membranes. Some members of the aquaporin family have been identified in the central nervous system (CNS). The water channel aquaporin 1 (AQP1) is restricted to the apical domain of the choroid plexus epithelial cells. The AQP4 is abundantly expressed in astrocyte foot processes and ependymocytes facing capillaries and brain-cerebrospinal fluid (CSF) interfaces, whereas AQP9 is localized in tanycytes and astrocytes processes. The mRNA for other aquaporin homologs (i.e., AQP3, 5, and 8) have been recently found in cultured astrocytes. Based on their subcellular localization and data obtained from functional studies, it is assumed that aquaporins are implicated in water movements in nervous tissue and may play a role in central osmoreception, K+ siphoning, and cerebrospinal fluid formation. There have been recent reports describing different aquaporin-responses under pathologic states leading to brain edema. The data available provide a better understanding of the mechanisms responsible for brain edema and indicate that aquaporins are potential targets for drug development.


Assuntos
Aquaporinas/classificação , Aquaporinas/metabolismo , Sistema Nervoso Central/metabolismo , Animais , Aquaporinas/química , Aquaporinas/fisiologia , Encefalopatias/metabolismo , Sistema Nervoso Central/anatomia & histologia , Regulação da Expressão Gênica/fisiologia , Humanos , Equilíbrio Hidroeletrolítico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...