Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Hepatol ; 77(3): 723-734, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35421427

RESUMEN

BACKGROUND & AIMS: Liver sinusoidal endothelial cells (LSECs) are ideally situated to sense stiffness and generate angiocrine programs that potentially regulate liver fibrosis and portal hypertension. We explored how specific focal adhesion (FA) proteins parlay LSEC mechanotransduction into stiffness-induced angiocrine signaling in vitro and in vivo. METHODS: Primary human and murine LSECs were placed on gels with incremental stiffness (0.2 kPa vs. 32 kPa). Cell response was studied by FA isolation, actin polymerization assay, RNA-sequencing and electron microscopy. Glycolysis was assessed using radioactive tracers. Epigenetic regulation of stiffness-induced genes was analyzed by chromatin-immunoprecipitation (ChIP) analysis of histone activation marks, ChIP sequencing and circularized chromosome conformation capture (4C). Mice with LSEC-selective deletion of glycolytic enzymes (Hk2fl/fl/Cdh5cre-ERT2) or treatment with the glycolysis inhibitor 3PO were studied in portal hypertension (partial ligation of the inferior vena cava, pIVCL) and early liver fibrosis (CCl4) models. RESULTS: Glycolytic enzymes, particularly phosphofructokinase 1 isoform P (PFKP), are enriched in isolated FAs from LSECs on gels with incremental stiffness. Stiffness resulted in PFKP recruitment to FAs, which paralleled an increase in glycolysis. Glycolysis was associated with expansion of actin dynamics and was attenuated by inhibition of integrin ß1. Inhibition of glycolysis attenuated a stiffness-induced CXCL1-dominant angiocrine program. Mechanistically, glycolysis promoted CXCL1 expression through nuclear pore changes and increases in NF-kB translocation. Biochemically, this CXCL1 expression was mediated through spatial re-organization of nuclear chromatin resulting in formation of super-enhancers, histone acetylation and NF-kB interaction with the CXCL1 promoter. Hk2fl/fl/Cdh5cre-ERT2 mice showed attenuated neutrophil infiltration and portal hypertension after pIVCL. 3PO treatment attenuated liver fibrosis in a CCl4 model. CONCLUSION: Glycolytic enzymes are involved in stiffness-induced angiocrine signaling in LSECs and represent druggable targets in early liver disease. LAY SUMMARY: Treatment options for liver fibrosis and portal hypertension still represent an unmet need. Herein, we uncovered a novel role for glycolytic enzymes in promoting stiffness-induced angiocrine signaling, which resulted in inflammation, fibrosis and portal hypertension. This work has revealed new targets that could be used in the prevention and treatment of liver fibrosis and portal hypertension.


Asunto(s)
Células Endoteliales , Hipertensión Portal , Actinas/metabolismo , Animales , Quimiocina CXCL1/metabolismo , Cromatina/metabolismo , Células Endoteliales/metabolismo , Epigénesis Genética , Glucólisis , Histonas/metabolismo , Humanos , Hipertensión Portal/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Mecanotransducción Celular , Ratones , FN-kappa B/metabolismo
2.
Liver Int ; 40(7): 1670-1685, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32378324

RESUMEN

BACKGROUND & AIMS: Polycystic liver diseases (PLDs) are genetic disorders characterized by progressive development of multiple biliary cysts. Recently, novel PLD-causative genes, encoding for endoplasmic reticulum (ER)-resident proteins involved in protein biogenesis and transport, were identified. We hypothesized that aberrant proteostasis contributes to PLD pathogenesis, representing a potential therapeutic target. METHODS: ER stress was analysed at transcriptional (qPCR), proteomic (mass spectrometry), morphological (transmission electron microscopy, TEM) and functional (proteasome activity) levels in different PLD models. The effect of ER stress inhibitors [4-phenylbutyric acid (4-PBA)] and/or activators [tunicamycin (TM)] was tested in polycystic (PCK) rats and cystic cholangiocytes in vitro. RESULTS: The expression levels of unfolded protein response (UPR) components were upregulated in liver tissue from PLD patients and PCK rats, as well as in primary cultures of human and rat cystic cholangiocytes, compared to normal controls. Cystic cholangiocytes showed altered proteomic profiles, mainly related to proteostasis (ie synthesis, folding, trafficking and degradation of proteins), marked enlargement of the ER lumen (by TEM) and hyperactivation of the proteasome. Notably, chronic treatment of PCK rats with 4-PBA decreased liver weight, as well as both liver and cystic volumes, of animals under baseline conditions or after TM administration compared to controls. In vitro, 4-PBA downregulated the expression (mRNA) of UPR effectors, normalized proteomic profiles related to protein synthesis, folding, trafficking and degradation and reduced the proteasome hyperactivity in cystic cholangiocytes, reducing their hyperproliferation and apoptosis. CONCLUSIONS: Restoration of proteostasis in cystic cholangiocytes with 4-PBA halts hepatic cystogenesis, emerging as a novel therapeutic strategy.


Asunto(s)
Quistes , Hepatopatías , Animales , Conductos Biliares , Proliferación Celular , Quistes/tratamiento farmacológico , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Humanos , Hepatopatías/tratamiento farmacológico , Hepatopatías/metabolismo , Proteómica , Proteostasis , Ratas
3.
Hepatology ; 67(3): 1088-1108, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29023824

RESUMEN

Polycystic liver disease (PLD) is a group of genetic disorders with limited treatment options and significant morbidity. Hepatic cysts arise from cholangiocytes exhibiting a hyperproliferative phenotype. Considering that hyperproliferation of many cell types is associated with alterations in autophagy, we hypothesized that autophagy is altered in PLD cholangiocytes, contributes to hepatic cystogenesis, and might represent a potential therapeutic target. We employed functional pathway cluster analysis and next-generation sequencing, transmission electron microscopy, immunofluorescence confocal microscopy, and western blotting to assess autophagy in human and rodent PLD cholangiocytes. A three-dimensional culture model was used to study the effects of molecular and pharmacologic inhibition of autophagy on hepatic cystogenesis in vitro, and the polycystic kidney disease-specific rat, an animal model of PLD, to study the effects of hydroxychloroquine, a drug that interferes with the autophagy pathway, on disease progression in vivo. Assessment of the transcriptome of PLD cholangiocytes followed by functional pathway cluster analysis revealed that the autophagy-lysosomal pathway is one of the most altered pathways in PLD. Direct evaluation of autophagy in PLD cholangiocytes both in vitro and in vivo showed increased number and size of autophagosomes, lysosomes, and autolysosomes; overexpression of autophagy-related proteins (Atg5, Beclin1, Atg7, and LC3); and enhanced autophagic flux associated with activation of the cAMP-protein kinase A-cAMP response element-binding protein signaling pathway. Molecular and pharmacologic intervention in autophagy with ATG7 small interfering RNA, bafilomycin A1 , and hydroxychloroquine reduced proliferation of PLD cholangiocytes in vitro and growth of hepatic cysts in three-dimensional cultures. Hydroxychloroquine also efficiently inhibited hepatic cystogenesis in the polycystic kidney disease-specific rat. CONCLUSION: Autophagy is increased in PLD cholangiocytes, contributes to hepatic cystogenesis, and represents a potential therapeutic target for disease treatment. (Hepatology 2018;67:1088-1108).


Asunto(s)
Autofagia/efectos de los fármacos , Conductos Biliares/citología , Quistes/fisiopatología , Hepatopatías/fisiopatología , Hígado/patología , Animales , Autofagia/genética , Autofagia/fisiología , Conductos Biliares/metabolismo , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Análisis por Conglomerados , Quistes/tratamiento farmacológico , Quistes/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Células Epiteliales/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Hidroxicloroquina/farmacología , Hígado/metabolismo , Hepatopatías/metabolismo , Masculino , Microscopía Electrónica de Transmisión , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Somatostatina/análogos & derivados , Somatostatina/farmacología
4.
Hepatology ; 66(4): 1197-1218, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28543567

RESUMEN

Hepatic cystogenesis in polycystic liver disease is associated with increased levels of cyclic adenosine monophosphate (cAMP) in cholangiocytes lining liver cysts. Takeda G protein receptor 5 (TGR5), a G protein-coupled bile acid receptor, is linked to cAMP and expressed in cholangiocytes. Therefore, we hypothesized that TGR5 might contribute to disease progression. We examined expression of TGR5 and Gα proteins in cultured cholangiocytes and in livers of animal models and humans with polycystic liver disease. In vitro, we assessed cholangiocyte proliferation, cAMP levels, and cyst growth in response to (1) TGR5 agonists (taurolithocholic acid, oleanolic acid [OA], and two synthetic compounds), (2) a novel TGR5 antagonist (m-tolyl 5-chloro-2-[ethylsulfonyl] pyrimidine-4-carboxylate [SBI-115]), and (3) a combination of SBI-115 and pasireotide, a somatostatin receptor analogue. In vivo, we examined hepatic cystogenesis in OA-treated polycystic kidney rats and after genetic elimination of TGR5 in double mutant TGR5-/- ;Pkhd1del2/del2 mice. Compared to control, expression of TGR5 and Gαs (but not Gαi and Gαq ) proteins was increased 2-fold to 3-fold in cystic cholangiocytes in vitro and in vivo. In vitro, TGR5 stimulation enhanced cAMP production, cell proliferation, and cyst growth by ∼40%; these effects were abolished after TGR5 reduction by short hairpin RNA. OA increased cystogenesis in polycystic kidney rats by 35%; in contrast, hepatic cystic areas were decreased by 45% in TGR5-deficient TGR5-/- ;Pkhd1del2/del2 mice. TGR5 expression and its colocalization with Gαs were increased ∼2-fold upon OA treatment. Levels of cAMP, cell proliferation, and cyst growth in vitro were decreased by ∼30% in cystic cholangiocytes after treatment with SBI-115 alone and by ∼50% when SBI-115 was combined with pasireotide. CONCLUSION: TGR5 contributes to hepatic cystogenesis by increasing cAMP and enhancing cholangiocyte proliferation; our data suggest that a TGR5 antagonist alone or concurrently with somatostatin receptor agonists represents a potential therapeutic approach in polycystic liver disease. (Hepatology 2017;66:1197-1218).


Asunto(s)
AMP Cíclico/metabolismo , Quistes/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Hepatopatías/metabolismo , Pirimidinas/uso terapéutico , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proliferación Celular/efectos de los fármacos , Quistes/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Humanos , Hepatopatías/tratamiento farmacológico , Ratones , Ácido Oleanólico , Enfermedades Renales Poliquísticas/metabolismo , Cultivo Primario de Células , Pirimidinas/farmacología , Ratas , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Somatostatina/análogos & derivados , Somatostatina/farmacología , Somatostatina/uso terapéutico
5.
Lab Invest ; 95(6): 684-96, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25867762

RESUMEN

Cholangiocytes are the target of a heterogeneous group of liver diseases known as the cholangiopathies. An evolving understanding of the mechanisms driving biliary development provides the theoretical underpinnings for rational development of induced pluripotent stem cell (iPSC)-derived cholangiocytes (iDCs). Therefore, the aims of this study were to develop an approach to generate iDCs and to fully characterize the cells in vitro and in vivo. Human iPSC lines were generated by forced expression of the Yamanaka pluripotency factors. We then pursued a stepwise differentiation strategy toward iDCs, using precise temporal exposure to key biliary morphogens, and we characterized the cells, using a variety of morphologic, molecular, cell biologic, functional, and in vivo approaches. Morphology shows a stepwise phenotypic change toward an epithelial monolayer. Molecular analysis during differentiation shows appropriate enrichment in markers of iPSC, definitive endoderm, hepatic specification, hepatic progenitors, and ultimately cholangiocytes. Immunostaining, western blotting, and flow cytometry demonstrate enrichment of multiple functionally relevant biliary proteins. RNA sequencing reveals that the transcriptome moves progressively toward that of human cholangiocytes. iDCs generate intracellular calcium signaling in response to ATP, form intact primary cilia, and self-assemble into duct-like structures in three-dimensional culture. In vivo, the cells engraft within mouse liver, following retrograde intrabiliary infusion. In summary, we have developed a novel approach to generate mature cholangiocytes from iPSCs. In addition to providing a model of biliary differentiation, iDCs represent a platform for in vitro disease modeling, pharmacologic testing, and individualized, cell-based, regenerative therapies for the cholangiopathies.


Asunto(s)
Conductos Biliares/citología , Células Epiteliales/citología , Células Madre Pluripotentes Inducidas/citología , Animales , Conductos Biliares/química , Conductos Biliares/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Señalización del Calcio , Diferenciación Celular , Ingeniería Celular , Línea Celular , Células Epiteliales/química , Células Epiteliales/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/química , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/química , Hígado/citología , Hígado/metabolismo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
PLoS Pathog ; 9(4): e1003261, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23592986

RESUMEN

Exosomes are membranous nanovesicles released by most cell types from multi-vesicular endosomes. They are speculated to transfer molecules to neighboring or distant cells and modulate many physiological and pathological procedures. Exosomes released from the gastrointestinal epithelium to the basolateral side have been implicated in antigen presentation. Here, we report that luminal release of exosomes from the biliary and intestinal epithelium is increased following infection by the protozoan parasite Cryptosporidium parvum. Release of exosomes involves activation of TLR4/IKK2 signaling through promoting the SNAP23-associated vesicular exocytotic process. Downregulation of let-7 family miRNAs by activation of TLR4 signaling increases SNAP23 expression, coordinating exosome release in response to C. parvum infection. Intriguingly, exosomes carry antimicrobial peptides of epithelial cell origin, including cathelicidin-37 and beta-defensin 2. Activation of TLR4 signaling enhances exosomal shuttle of epithelial antimicrobial peptides. Exposure of C. parvum sporozoites to released exosomes decreases their viability and infectivity both in vitro and ex vivo. Direct binding to the C. parvum sporozoite surface is required for the anti-C. parvum activity of released exosomes. Biliary epithelial cells also increase exosomal release and display exosome-associated anti-C. parvum activity following LPS stimulation. Our data indicate that TLR4 signaling regulates luminal exosome release and shuttling of antimicrobial peptides from the gastrointestinal epithelium, revealing a new arm of mucosal immunity relevant to antimicrobial defense.


Asunto(s)
Criptosporidiosis/inmunología , Cryptosporidium parvum/inmunología , Exosomas/metabolismo , Mucosa Intestinal/inmunología , Receptor Toll-Like 4/metabolismo , Presentación de Antígeno , Catelicidinas/metabolismo , Línea Celular , Activación Enzimática , Células Epiteliales/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , MicroARNs/biosíntesis , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Interferencia de ARN , Transducción de Señal/inmunología , Esporozoítos/inmunología , Esporozoítos/metabolismo , beta-Defensinas/metabolismo
7.
Kidney Int ; 85(5): 1225-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24196483

RESUMEN

Urinary exosome-like vesicles (ELVs) are a heterogenous mixture (diameter 40-200 nm) containing vesicles shed from all segments of the nephron including glomerular podocytes. Contamination with Tamm-Horsfall protein (THP) oligomers has hampered their isolation and proteomic analysis. Here we improved ELV isolation protocols employing density centrifugation to remove THP and albumin, and isolated a glomerular membranous vesicle (GMV)-enriched subfraction from 7 individuals identifying 1830 proteins and in 3 patients with glomerular disease identifying 5657 unique proteins. The GMV fraction was composed of podocin/podocalyxin-positive irregularly shaped membranous vesicles and podocin/podocalyxin-negative classical exosomes. Ingenuity pathway analysis identified integrin, actin cytoskeleton, and Rho GDI signaling in the top three canonical represented signaling pathways and 19 other proteins associated with inherited glomerular diseases. The GMVs are of podocyte origin and the density gradient technique allowed isolation in a reproducible manner. We show many nephrotic syndrome proteins, proteases, and complement proteins involved in glomerular disease are in GMVs and some were only shed in the disease state (nephrin, TRPC6, INF2 and phospholipase A2 receptor). We calculated sample sizes required to identify new glomerular disease biomarkers, expand the ELV proteome, and provide a reference proteome in a database that may prove useful in the search for biomarkers of glomerular disease.


Asunto(s)
Exosomas/química , Membrana Basal Glomerular/química , Enfermedades Renales/orina , Podocitos/química , Proteinuria/orina , Proteómica/métodos , Urinálisis , Orina/química , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Biomarcadores/orina , Estudios de Casos y Controles , Centrifugación por Gradiente de Densidad , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Enfermedades Renales/diagnóstico , Masculino , Datos de Secuencia Molecular , Proteinuria/diagnóstico , Adulto Joven
8.
Am J Physiol Gastrointest Liver Physiol ; 304(11): G1013-24, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23578785

RESUMEN

TGR5, the G protein-coupled bile acid receptor that transmits bile acid signaling into a cell functional response via the intracellular cAMP signaling pathway, is expressed in human and rodent cholangiocytes. However, detailed information on the localization and function of cholangiocyte TGR5 is limited. We demonstrated that in human (H69 cells) and rat cholangiocytes, TGR5 is localized to multiple, diverse subcellular compartments, with its strongest expression on the apical plasma, ciliary, and nuclear membranes. To evaluate the relationship between ciliary TGR5 and the cholangiocyte functional response to bile acid signaling, we used a model of ciliated and nonciliated H69 cells and demonstrated that TGR5 agonists induce opposite changes in cAMP and ERK levels in cells with and without primary cilia. The cAMP level was increased in nonciliated cholangiocytes but decreased in ciliated cells. In contrast, ERK signaling was induced in ciliated cholangiocytes but suppressed in cells without cilia. TGR5 agonists inhibited proliferation of ciliated cholangiocytes but activated proliferation of nonciliated cells. The observed differential effects of TGR5 agonists were associated with the coupling of TGR5 to Gαi protein in ciliated cells and Gαs protein in nonciliated cholangiocytes. The functional responses of nonciliated and ciliated cholangiocytes to TGR5-mediated bile acid signaling may have important pathophysiological significance in cilia-related liver disorders (i.e., cholangiociliopathies), such as polycystic liver disease. In summary, TGR5 is expressed on diverse cholangiocyte compartments, including a primary cilium, and its ciliary localization determines the cholangiocyte functional response to bile acid signaling.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Conductos Biliares Intrahepáticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Conductos Biliares Intrahepáticos/citología , Línea Celular , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Cilios/metabolismo , Cilios/ultraestructura , AMP Cíclico/metabolismo , Células Epiteliales/metabolismo , Exosomas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas
9.
J Am Soc Nephrol ; 22(12): 2266-77, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22021705

RESUMEN

Mutations in the PKHD1 gene, which encodes fibrocystin, cause autosomal recessive polycystic kidney disease (ARPKD). Unfortunately, the lack of specific antibodies to the mouse protein impairs the study of splicing, post-translational processing, shedding, and temporal and spatial expression of endogenous fibrocystin at the cellular and subcellular level. Here, we report using a knock-in strategy to generate a null Pkhd1 strain and a strain that expresses fibrocystin along with two SV5-Pk epitope tags engineered in-frame into the third exon, immediately C-terminal to the signal-peptide cleavage site in a poorly conserved region. By 6 mo of age, the Pkhd1-null mouse develops massive cystic hepatomegaly and proximal tubule dilation, whereas the mouse with epitope-tagged fibrocystin has histologically normal liver and kidneys at 14 mo. Although Pkhd1 was believed to generate many splice forms, our western analysis resolved fibrocystin as a 500 kD product without other forms in the 15-550 kD range. Western analysis also revealed that exosome-like vesicles (ELVs) secrete the bulk of fibrocystin in its mature cleaved form, and scanning electron microscopy identified that fibrocystin on ELVs attached to cilia. Furthermore, the addition of ELVs with epitope-tagged fibrocystin to wild-type cells showed that label transferred to primary cilia within 5 min. In summary, tagging of the endogenous Pkhd1 gene facilitates the study of the glycosylation, proteolytic cleavage, and shedding of fibrocystin.


Asunto(s)
Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/fisiología , Animales , Epítopos , Femenino , Masculino , Ratones
10.
J Extracell Vesicles ; 11(4): e12208, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35383410

RESUMEN

Extracellular vesicles (EVs) are of growing interest due to their potential diagnostic, disease surveillance, and therapeutic applications. While several studies have evaluated EV isolation methods in various biofluids, there are few if any data on these techniques when applied to stool. The latter is an ideal biospecimen for studying EVs and colorectal cancer (CRC) because the release of tumour markers by luminal exfoliation into stool occurs earlier than vascular invasion. Since EV release is a conserved mechanism, bacteria in stool contribute to the overall EV population. In this study, we assessed five EV separation methods (ultracentrifugation [UC], precipitation [EQ-O, EQ-TC], size exclusion chromatography [SEC], and ultrafiltration [UF]) for total recovery, reproducibility, purity, RNA composition, and protein expression in stool supernatant. CD63, TSG101, and ompA proteins were present in EV fractions from all methods except UC. Human (18s) and bacterial (16s) rRNA was detected in stool EV preparations. Enzymatic treatment prior to extraction is necessary to avoid non-vesicular RNA contamination. Ultrafiltration had the highest recovery, RNA, and protein yield. After assessing purity further, SEC was the isolation method of choice. These findings serve as the groundwork for future studies that use high throughput omics technologies to investigate the potential of stool-derived EVs as a source for novel biomarkers for early CRC detection.


Asunto(s)
Vesículas Extracelulares , Cromatografía en Gel , Vesículas Extracelulares/metabolismo , Humanos , Reproducibilidad de los Resultados , Ultracentrifugación , Ultrafiltración
11.
Clin Cancer Res ; 28(14): 3021-3031, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35552651

RESUMEN

PURPOSE: This phase Ib/2 trial investigated pembrolizumab-containing trimodality therapy in patients with gastroesophageal junction (GEJ) adenocarcinoma. PATIENTS AND METHODS: Patients with GEJ adenocarcinoma (cT1-3NanyM0) received neoadjuvant pembrolizumab-containing chemoradiation (CROSS regimen) followed by surgical resection and adjuvant pembrolizumab. The primary endpoints were tolerability in the first 16 patients and pathologic complete response [pCR (ypT0N0)]. Secondary endpoints included progression-free survival (PFS) and overall survival (OS). An independent propensity-score-matched cohort (treated with CROSS without immunotherapy) was used for comparison. Exploratory analyses included immune biomarkers in the tumor microenvironment (TME) and plasma. RESULTS: We enrolled 31 eligible patients, of whom 29 received all expected doses of neoadjuvant pembrolizumab and 28 underwent R0 resection. Safety endpoints were met. The primary efficacy endpoint was not met [7/31 (22.6%) achieved pCR]. Patients with high [i.e., combined positive score (CPS) ≥ 10] baseline expression of programmed death (PD)-L1 in the TME had a significantly higher pCR rate than those with low expression [50.0% (4/8) vs. 13.6% (3/22); P = 0.046]. Patients with high PD-L1 expression also experienced longer PFS and OS than propensity-score-matched patients. Among trial patients with PD-L1 CPS < 10, unprespecified analysis explored whether extracellular vesicles (EV) could identify further responders: an elevated plasma level of PD-L1-expressing EVs was significantly associated with higher pCR. CONCLUSIONS: Adding pembrolizumab to trimodality therapy showed acceptable tolerability but did not meet the pre-specified pCR endpoint. Exploratory analyses suggested that high PD-L1 expression in the TME and/or on EVs may identify patients most likely to achieve tumor response.


Asunto(s)
Adenocarcinoma , Antineoplásicos Inmunológicos , Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados , Antineoplásicos Inmunológicos/efectos adversos , Antígeno B7-H1/metabolismo , Neoplasias Esofágicas , Unión Esofagogástrica/patología , Humanos , Terapia Neoadyuvante , Microambiente Tumoral
12.
Gastroenterology ; 139(1): 304-14.e2, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20399209

RESUMEN

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.


Asunto(s)
Conductos Biliares/citología , Riñón Poliquístico Autosómico Recesivo/terapia , Canales Catiónicos TRPV/fisiología , Animales , Calcio/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Fenotipo , Ésteres del Forbol/farmacología , Riñón Poliquístico Autosómico Recesivo/patología , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
13.
Hepatology ; 52(1): 238-48, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20578142

RESUMEN

UNLABELLED: Increasing evidence suggests that hepatic fibrosis and pathological angiogenesis are interdependent processes that occur in parallel. Endothelial cell invasion is requisite for angiogenesis, and thus studies of the mechanisms governing liver endothelial cell (LEC) invasion during cirrhosis are of great importance. Emerging research implicates amoeboid-type motility and membrane blebbing as features that may facilitate invasion through matrix-rich microenvironments. Aquaporins (AQPs) are integral membrane water channels, recognized for their importance in epithelial secretion and absorption. However, recent studies also suggest links between water transport and cell motility or invasion. Therefore, the purpose of this study was to test the hypothesis that AQP-1 is involved in amoeboid motility and angiogenic invasion during cirrhosis. AQP-1 expression and localization was examined in normal and cirrhotic liver tissues derived from human and mouse. AQP-1 levels were modulated in LEC using retroviral overexpression or small interfering RNA (siRNA) knockdown and functional effects on invasion, membrane blebbing dynamics, and osmotic water permeability were assayed. Results demonstrate that AQP-1 is up-regulated in the small, angiogenic, neovasculature within the fibrotic septa of cirrhotic liver. AQP-1 overexpression promotes fibroblast growth factor (FGF)-induced dynamic membrane blebbing in LEC, which is sufficient to augment invasion through extracellular matrix. Additionally, AQP-1 localizes to plasma membrane blebs, where it increases osmotic water permeability and locally facilitates the rapid, trans-membrane flux of water. CONCLUSION: AQP-1 enhances osmotic water permeability and FGF-induced dynamic membrane blebbing in LEC and thereby drives invasion and pathological angiogenesis during cirrhosis.


Asunto(s)
Acuaporina 1/metabolismo , Movimiento Celular , Endotelio Vascular/patología , Cirrosis Hepática/patología , Neovascularización Patológica/metabolismo , Animales , Acuaporina 1/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/genética , Ósmosis , ARN Interferente Pequeño/genética , Agua/metabolismo
14.
Lab Invest ; 90(12): 1770-81, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20644520

RESUMEN

Hepatic sinusoidal endothelial cells (HSECs) are a unique subpopulation of fenestrated endothelial cells lining the hepatic sinusoids and comprising the majority of endothelial cells within the liver. HSECs not only have important roles in blood clearance, vascular tone, and immunity, but also undergo pathological changes, contributing to fibrosis, angiogenesis, and portal hypertension. There are few cell culture models for in vitro studies of motility and angiogenesis as primary cells are time-consuming to isolate, are limited in number, and often lack features of pathological vasculature. The aim of this study was to generate an immortalized cell line derived from HSECs that mimic pathological vasculature and allows detailed molecular interventions to be pursued. HSECs were isolated from mouse liver using CD31-based immunomagnetic separation, immortalized with SV40 large T-antigen, and subcloned on the basis of their ability to endocytose the acetylated low-density lipoprotein (AcLDL). The resulting cell line, transformed sinusoidal endothelial cells (TSECs), maintains an endothelial phenotype as well as some HSEC-specific features. This is evidenced by typical microscopic features of endothelia, including formation of lamellipodia and filopodia, and a cobblestone morphology of cell monolayers. Electron microscopy showed maintenance of a limited number of fenestrae organized in sieve plates. TSECs express numerous endothelia-specific markers, including CD31 and von Willebrand's factor (vWF), as detected by PCR array, immunoblotting, and immunofluorescence (IF). Functionally, TSECs maintain a number of key endothelial features, including migration in response to angiogenic factors, formation of vascular tubes, endocytosis of AcLDL, and remodeling of extracellular matrix. Their phenotype most closely resembles the pathological neovasculature associated with chronic liver disease, in which cells become proliferative, defenestrated, and angiogenic. Importantly, the cells can be transduced efficiently with viral vectors. TSECs should provide a reproducible cell culture model for high-throughput in vitro studies pertaining to a broad range of liver endothelial cell functions, but likely broader endothelial cell biology as well.


Asunto(s)
Línea Celular Transformada , Movimiento Celular , Células Endoteliales/citología , Células Endoteliales/inmunología , Hígado/citología , Hígado/inmunología , Neovascularización Fisiológica , Animales , Técnicas de Cultivo de Célula/métodos , Línea Celular Transformada/citología , Movimiento Celular/inmunología , Transformación Celular Viral/inmunología , Quimiotaxis/inmunología , Células Endoteliales/ultraestructura , Separación Inmunomagnética , Lentivirus/inmunología , Hepatopatías/inmunología , Ratones , Neovascularización Fisiológica/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Factor de von Willebrand/inmunología
16.
Am J Physiol Gastrointest Liver Physiol ; 299(4): G990-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20634433

RESUMEN

Exosomes are small extracellular vesicles that are thought to participate in intercellular communication. Recent work from our laboratory suggests that, in normal and cystic liver, exosome-like vesicles accumulate in the lumen of intrahepatic bile ducts, presumably interacting with cholangiocyte cilia. However, direct evidence for exosome-ciliary interaction is limited and the physiological relevance of such interaction remains unknown. Thus, in this study, we tested the hypothesis that biliary exosomes are involved in intercellular communication by interacting with cholangiocyte cilia and inducing intracellular signaling and functional responses. Exosomes were isolated from rat bile by differential ultracentrifugation and characterized by scanning, transmission, and immunoelectron microscopy. The exosome-ciliary interaction and its effects on ERK1/2 signaling, expression of the microRNA, miR-15A, and cholangiocyte proliferation were studied on ciliated and deciliated cultured normal rat cholangiocytes. Our results show that bile contains vesicles identified as exosomes by their size, characteristic "saucer-shaped" morphology, and specific markers, CD63 and Tsg101. When NRCs were exposed to isolated biliary exosomes, the exosomes attached to cilia, inducing a decrease of the phosphorylated-to-total ERK1/2 ratio, an increase of miR-15A expression, and a decrease of cholangiocyte proliferation. All these effects of biliary exosomes were abolished by the pharmacological removal of cholangiocyte cilia. Our findings suggest that bile contains exosomes functioning as signaling nanovesicles and influencing intracellular regulatory mechanisms and cholangiocyte proliferation through interaction with primary cilia.


Asunto(s)
Sistema Biliar/citología , Cilios/fisiología , Exosomas/fisiología , Vesícula Biliar/citología , Animales , Proliferación Celular , Expresión Génica , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
17.
Proc Natl Acad Sci U S A ; 104(48): 19138-43, 2007 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-18024594

RESUMEN

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.


Asunto(s)
Bicarbonatos/metabolismo , Conductos Biliares Intrahepáticos/citología , Cilios/metabolismo , Canales Catiónicos TRPV/fisiología , Adenosina Trifosfato/metabolismo , Animales , Bilis/metabolismo , Conductos Biliares Intrahepáticos/metabolismo , Señalización del Calcio , Concentración de Iones de Hidrógeno , Soluciones Hipotónicas/farmacología , Masculino , Concentración Osmolar , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/biosíntesis , Canales Catiónicos TRPV/genética , Agua/metabolismo
18.
J Am Soc Nephrol ; 20(2): 278-88, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19158352

RESUMEN

Proteins associated with autosomal dominant and autosomal recessive polycystic kidney disease (polycystin-1, polycystin-2, and fibrocystin) localize to various subcellular compartments, but their functional site is thought to be on primary cilia. PC1+ vesicles surround cilia in Pkhd1(del2/del2) mice, which led us to analyze these structures in detail. We subfractionated urinary exosome-like vesicles (ELVs) and isolated a subpopulation abundant in polycystin-1, fibrocystin (in their cleaved forms), and polycystin-2. This removed Tamm-Horsfall protein, the major contaminant, and subfractionated ELVs into at least three different populations, demarcated by the presence of aquaporin-2, polycystin-1, and podocin. Proteomic analysis of PKD ELVs identified 552 proteins (232 not yet in urinary proteomic databases), many of which have been implicated in signaling, including the molecule Smoothened. We also detected two other protein products of genes involved in cystic disease: Cystin, the product of the mouse cpk locus, and ADP-ribosylation factor-like 6, the product of the human Bardet-Biedl syndrome gene (BBS3). Our proteomic analysis confirmed that cleavage of polycystin-1 and fibrocystin occurs in vivo, in manners consistent with cleavage at the GPS site in polycystin-1 and the proprotein convertase site in fibrocystin. In vitro, these PKD ELVs preferentially interacted with primary cilia of kidney and biliary epithelial cells in a rapid and highly specific manner. These data suggest that PKD proteins are shed in membrane particles in the urine, and these particles interact with primary cilia.


Asunto(s)
Exosomas/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Animales , Biotinilación , Humanos , Riñón/metabolismo , Ratones , Modelos Biológicos , Proteoma , Proteómica/métodos , Ratas , Receptores de Superficie Celular/metabolismo , Canales Catiónicos TRPP/metabolismo
19.
Am J Pathol ; 173(6): 1637-46, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18988797

RESUMEN

Polycystic kidney (PCK) rats are a spontaneous model of autosomal recessive polycystic kidney disease that exhibit cholangiocyte-derived liver cysts. We have previously reported that in normal cholangiocytes a subset of vesicles contain three proteins (ie, the water channel AQP1, the chloride channel CFTR, and the anion exchanger AE2) that account for ion-driven water transport. Thus, we hypothesized that altered expression and location of these functionally related proteins contribute to hepatic cystogenesis. We show here that under basal conditions and in response to secretin and hypotonicity, cysts from PCK rats expanded to a greater degree than cysts formed by normal bile ducts. Quantitative reverse transcriptase-polymerase chain reaction, immunoblot analysis, and confocal and immunoelectron microscopy all indicated increased expression of these three proteins in PCK cholangiocytes versus normal cholangiocytes. AQP1, CFTR, and AE2 were localized preferentially to the apical membrane in normal rats while overexpressed at the basolateral membrane in PCK rats. Exposure of the cholangiocyte basolateral membrane to CFTR inhibitors [5-nitro-2-(3-phenylpropylamino)-benzoic acid and CFTRinh172], or Cl(-)/HCO(3)(-) exchange inhibitors (4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid disodium salt hydrate and 4-acetamido-4'-isothiocyanato-2,2'-stilbenedisulfonic acid disodium salt hydrate) blocked secretin-stimulated fluid accumulation in PCK but not in normal cysts. Our data suggest that hepatic cystogenesis in autosomal recessive polycystic kidney disease may involve increased fluid accumulation because of overexpression and abnormal location of AQP1, CFTR, and AE2 in cystic cholangiocytes. Therapeutic interventions that block the activation of these proteins might inhibit cyst expansion in polycystic liver disease.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Antiportadores/metabolismo , Acuaporina 1/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Hígado/patología , Riñón Poliquístico Autosómico Recesivo/metabolismo , Animales , Proteínas de Transporte de Anión/genética , Antiportadores/genética , Acuaporina 1/genética , Conductos Biliares/citología , Técnicas de Cultivo de Célula , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Hígado/metabolismo , Riñón Poliquístico Autosómico Recesivo/patología , Ratas , Ratas Endogámicas , Ratas Sprague-Dawley , Proteínas SLC4A , Secretina/metabolismo
20.
J Parasitol ; 91(5): 995-9, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16419739

RESUMEN

Although accumulating evidence supports an active role for host cells during Cryptosporidium parvum invasion of epithelia, our knowledge of the underlying parasite-specific processes triggering such events is limited. In an effort to better understand the invasion strategy of C. parvum, we characterized the presence and distribution of the apical organelles (micronemes, dense granules, and rhoptry) through the stages of attachment to, and internalization by, human biliary epithelia, using serial-section electron microscopy. Novel findings include an apparent organized rearrangement of micronemes upon host cell attachment. The apically segregated micronemes were apposed to a central microtubule-like filamentous structure, and the more distal micronemes localized to the periphery and apical region of the parasite during internalization, coinciding with the formation of the anterior vacuole. The morphological observations presented here extend our understanding of parasite-specific processes that occur during attachment to, and internalization by, host epithelial cells.


Asunto(s)
Conductos Biliares/parasitología , Cryptosporidium parvum/ultraestructura , Orgánulos/ultraestructura , Animales , Conductos Biliares/citología , Línea Celular Transformada , Cryptosporidium parvum/fisiología , Gránulos Citoplasmáticos/parasitología , Gránulos Citoplasmáticos/ultraestructura , Células Epiteliales/parasitología , Interacciones Huésped-Parásitos , Humanos , Microscopía Electrónica de Transmisión/métodos , Microtúbulos/parasitología , Microtúbulos/ultraestructura , Orgánulos/parasitología , Vacuolas/parasitología , Vacuolas/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA