Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 51(4): 638-654.e9, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31561945

RESUMEN

Macrophages are strongly adapted to their tissue of residence. Yet, little is known about the cell-cell interactions that imprint the tissue-specific identities of macrophages in their respective niches. Using conditional depletion of liver Kupffer cells, we traced the developmental stages of monocytes differentiating into Kupffer cells and mapped the cellular interactions imprinting the Kupffer cell identity. Kupffer cell loss induced tumor necrosis factor (TNF)- and interleukin-1 (IL-1) receptor-dependent activation of stellate cells and endothelial cells, resulting in the transient production of chemokines and adhesion molecules orchestrating monocyte engraftment. Engrafted circulating monocytes transmigrated into the perisinusoidal space and acquired the liver-associated transcription factors inhibitor of DNA 3 (ID3) and liver X receptor-α (LXR-α). Coordinated interactions with hepatocytes induced ID3 expression, whereas endothelial cells and stellate cells induced LXR-α via a synergistic NOTCH-BMP pathway. This study shows that the Kupffer cell niche is composed of stellate cells, hepatocytes, and endothelial cells that together imprint the liver-specific macrophage identity.


Asunto(s)
Células Endoteliales/fisiología , Células Estrelladas Hepáticas/fisiología , Hepatocitos/fisiología , Macrófagos del Hígado/fisiología , Hígado/citología , Macrófagos/fisiología , Monocitos/fisiología , Animales , Comunicación Celular , Diferenciación Celular , Células Cultivadas , Microambiente Celular , Femenino , Regulación de la Expresión Génica , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Notch/metabolismo
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1024-1036, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29329986

RESUMEN

Liver fibrosis is the result of persistent liver injury, and is characterized by sustained scar formation and disruption of the normal liver architecture. The extent of fibrosis is considered as an important prognostic factor for the patient outcome, as an absence of (early) treatment can lead to the development of liver cirrhosis and hepatocellular carcinoma. Till date, the most sensitive and specific way for the diagnosis and staging of liver fibrosis remains liver biopsy, an invasive diagnostic tool, which is associated with high costs and discomfort for the patient. Over time, non-invasive scoring systems have been developed, of which the measurements of serum markers and liver stiffness are validated for use in the clinic. These tools lack however the sensitivity and specificity to detect small changes in the progression or regression of both early and late stages of fibrosis. Novel non-invasive diagnostic markers with the potential to overcome these limitations have been developed, but often lack validation in large patient cohorts. In this review, we will summarize novel trends in non-invasive markers of liver fibrosis development and will discuss their (dis-)advantages for use in the clinic.


Asunto(s)
Metabolismo de los Lípidos , Lípidos , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/metabolismo , Animales , Biomarcadores/metabolismo , Biopsia , Humanos , Cirrosis Hepática/patología
3.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 819-830, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29246445

RESUMEN

Pannexins are transmembrane proteins that form communication channels connecting the cytosol of an individual cell with its extracellular environment. A number of studies have documented the presence of pannexin1 in liver as well as its involvement in inflammatory responses. In this study, it was investigated whether pannexin1 plays a role in acute liver failure and non-alcoholic steatohepatitis, being prototypical acute and chronic liver pathologies, respectively, both featured by liver damage, oxidative stress and inflammation. To this end, wild-type and pannexin1-/- mice were overdosed with acetaminophen for 1, 6, 24 or 48h or were fed a choline-deficient high-fat diet for 8weeks. Evaluation of the effects of genetic pannexin1 deletion was based on a number of clinically relevant read-outs, including markers of liver damage, histopathological analysis, lipid accumulation, protein adduct formation, oxidative stress and inflammation. In parallel, in order to elucidate molecular pathways affected by pannexin1 deletion as well as to mechanistically anchor the clinical observations, whole transcriptome analysis of liver tissue was performed. The results of this study show that pannexin1-/- diseased mice present less liver damage and oxidative stress, while inflammation was only decreased in pannexin1-/- mice in which non-alcoholic steatohepatitis was induced. A multitude of genes related to inflammation, oxidative stress and xenobiotic metabolism were differentially modulated in both liver disease models in wild-type and in pannexin1-/- mice. Overall, the results of this study suggest that pannexin1 may play a role in the pathogenesis of liver disease.


Asunto(s)
Conexinas/genética , Citoprotección/genética , Eliminación de Gen , Hepatopatías/genética , Hígado/metabolismo , Proteínas del Tejido Nervioso/genética , Enfermedad Aguda , Animales , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Hígado/patología , Hepatopatías/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Hepatol ; 63(3): 679-88, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25908270

RESUMEN

BACKGROUND & AIMS: Hepatic stellate cell activation is a wound-healing response to liver injury. However, continued activation of stellate cells during chronic liver damage causes excessive matrix deposition and the formation of pathological scar tissue leading to fibrosis and ultimately cirrhosis. The importance of sustained stellate cell activation for this pathological process is well recognized, and several signalling pathways that can promote stellate cell activation have been identified, such as the TGFß-, PDGF-, and LPS-dependent pathways. However, the mechanisms that trigger and drive the early steps in activation are not well understood. METHODS AND RESULTS: We identified the Hippo pathway and its effector YAP as a key pathway that controls stellate cell activation. YAP is a transcriptional co-activator and we found that it drives the earliest changes in gene expression during stellate cell activation. Activation of stellate cells in vivo by CCl4 administration to mice or activation in vitro caused rapid activation of YAP as revealed by its nuclear translocation and by the induction of YAP target genes. YAP was also activated in stellate cells of human fibrotic livers as evidenced by its nuclear localization. Importantly, knockdown of YAP expression or pharmacological inhibition of YAP prevented hepatic stellate cell activation in vitro and pharmacological inhibition of YAP impeded fibrogenesis in mice. CONCLUSIONS: YAP activation is a critical driver of hepatic stellate cell activation and inhibition of YAP presents a novel approach for the treatment of liver fibrosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Células Estrelladas Hepáticas/fisiología , Fosfoproteínas/fisiología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular , Vía de Señalización Hippo , Humanos , Cirrosis Hepática/prevención & control , Ratones , Ratones Endogámicos BALB C , Fosfoproteínas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Factores de Transcripción , Proteínas Señalizadoras YAP
5.
Liver Int ; 35(4): 1253-64, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25243526

RESUMEN

BACKGROUND & AIMS: Liver fibrosis is induced by the accumulation of extracellular matrix, deposited mainly by activated hepatic stellate cells (HSCs). One key characteristic of stellate cell activation is the directional migration to the site of injury during the wound-healing process. P311 is a protein that has been shown to play a role in migration and we aimed to study a possible role for this protein during stellate cell migration. METHODS: Mouse stellate cells were isolated and cultured in vitro to investigate P311 protein and gene expression during HSC activation by immunocytochemistry and RT-qPCR respectively. Expression of P311 during in vivo activation was evaluated in CCl4 and bile duct ligation-induced liver fibrosis. Production of reactive oxygen species was determined using the fluorescent probe DCFH-DA. By siRNA-mediated knockdown of P311, we investigated a possible effect on proliferation by incorporation of EdU and on migration by Boyden chamber assays. RESULTS: P311 gene expression was increased during both in vitro and in vivo activation of HSCs. siRNA-mediated knockdown led to a decrease in reactive oxygen production and cell proliferation. Migration induced by different chemokines, such as PDGF-bb and MCP-1 was inhibited by knockdown of P311. CONCLUSIONS: P311 is central to reactive oxygen species-mediated HSC migration induced by different chemokines.


Asunto(s)
Movimiento Celular , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática Experimental/metabolismo , Hígado/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Becaplermina , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Quimiocina CCL2/farmacología , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática Experimental/genética , Cirrosis Hepática Experimental/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/genética , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-sis/farmacología , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta/farmacología
6.
JHEP Rep ; 6(5): 101036, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38694958

RESUMEN

Background & Aims: Chronic liver disease (CLD) remains a global health issue associated with a significant disease burden. Liver fibrosis, a hallmark of CLD, is characterised by the activation of hepatic stellate cells (HSCs) that gain profibrotic characteristics including increased production of extracellular matrix protein. Currently, no antifibrotic therapies are available clinically, in part because of the lack of HSC-specific drug targets. Here, we aimed to identify HSC-specific membrane proteins that can serve as targets for antifibrotic drug development. Methods: Small interfering RNA-mediated knockdown of GPR176 was used to assess the in vitro function of GPR176 in HSCs and in precision cut liver slices (PCLS). The in vivo role of GPR176 was assessed using the carbon tetrachloride (CCl4) and common bile duct ligation (BDL) models in wild-type and GPR176 knockout mice. GPR176 in human CLD was assessed by immunohistochemistry of diseased human livers and RNA expression analysis in human primary HSCs and transcriptomic data sets. Results: We identified Gpr176, an orphan G-protein coupled receptor, as an HSC-enriched activation associated gene. In vitro, Gpr176 is strongly induced upon culture-induced and hepatocyte-damage-induced activation of primary HSCs. Knockdown of GPR176 in primary mouse HSCs or PCLS cultures resulted in reduced fibrogenic characteristics. Absence of GPR176 did not influence liver homeostasis, but Gpr176-/- mice developed less severe fibrosis in CCl4 and BDL fibrosis models. In humans, GPR176 expression was correlated with in vitro HSC activation and with fibrosis stage in patients with CLD. Conclusions: GPR176 is a functional protein during liver fibrosis and reducing its activity attenuates fibrogenesis. These results highlight the potential of GPR176 as an HSC-specific antifibrotic candidate to treat CLD. Impact and implications: The lack of effective antifibrotic drugs is partly attributed to the insufficient knowledge about the mechanisms involved in the development of liver fibrosis. We demonstrate that the G-protein coupled receptor GPR176 contributes to fibrosis development. Since GPR176 is specifically expressed on the membrane of activated hepatic stellate cells and is linked with fibrosis progression in humans, it opens new avenues for the development of targeted interventions.

7.
Toxicol Appl Pharmacol ; 271(2): 276-84, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707763

RESUMEN

Histone deacetylase (HDAC) inhibitors are promising new compounds for the therapy of fibrotic diseases. In this study we compared the effect of two HDAC inhibitors, trichostatin A and valproic acid, in an experimental model of kidney fibrosis. In mice, doxorubicin (adriamycin) can cause nephropathy characterized by chronic proteinuria, glomerular damage and interstitial inflammation and fibrosis, as seen in human focal segmental glomerulosclerosis. Two treatment regimens were applied, treatment was either started prior to the doxorubicin insult or delayed until a significant degree of proteinuria and fibrosis was present. Pre-treatment of trichostatin A significantly hampered glomerulosclerosis and tubulointerstitial fibrosis, as did the pre-treatment with valproic acid. In contrast, the development of proteinuria was only completely inhibited in the pre-treated valproic acid group, and not in the pre-treated trichostatin A animals. In the postponed treatment with valproic acid, a complete resolution of established doxorubicin-induced proteinuria was achieved within three days, whereas trichostatin A could not correct proteinuria in such a treatment regimen. However, both postponed regimens have comparable efficacy in maintaining the kidney fibrosis to the level reached at the start of the treatments. Moreover, not only the process of fibrosis, but also renal inflammation was attenuated by both HDAC inhibitors. Our data confirm a role for HDACs in renal fibrogenesis and point towards a therapeutic potential for HDAC inhibitors. The effect on renal disease progression and manifestation can however be different for individual HDAC inhibitors.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Ácido Valproico/uso terapéutico , Acetilación , Animales , Doxorrubicina , Femenino , Fibrosis , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Glomeruloesclerosis Focal y Segmentaria/patología , Inmunohistoquímica , Inflamación/inducido químicamente , Inflamación/patología , Riñón/patología , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Infiltración Neutrófila , ARN Mensajero/biosíntesis , ARN Mensajero/genética
8.
Methods Mol Biol ; 2669: 193-206, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37247061

RESUMEN

In vitro models of liver fibrosis have evolved from mono-cultures of primary rodent hepatic stellate cells and stellate cell lines, to more complex co-cultures of primary or stem cell-derived liver cells. Great progress has been made in the development of stem cell-derived liver cultures; however, the liver cells obtained from stem cells do not yet fully recapitulate the phenotype of their in vivo counterparts. Freshly isolated rodent cells remain the most representative cell type to use for in vitro culture. To study liver injury-induced fibrosis, co-cultures of hepatocytes and stellate cells are an informative minimal model. Here, we describe a robust protocol to isolate hepatocytes and hepatic stellate cells from one mouse and a method for the subsequent seeding and culture as free-floating spheroids.


Asunto(s)
Células Estrelladas Hepáticas , Hepatocitos , Ratones , Animales , Hepatocitos/metabolismo , Hígado , Cirrosis Hepática/metabolismo , Técnicas de Cocultivo
9.
J Am Soc Nephrol ; 22(10): 1863-75, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21868496

RESUMEN

Inhibitors of histone deacetylase (HDAC) have anti-inflammatory and antifibrotic effects in several organs and tissues, but their effect on the progression of renal disease is unknown. Here, we studied the effect of valproic acid in adriamycin-induced nephropathy in mice. Administration of valproic acid before kidney injury prevented the development of proteinuria and the onset of glomerulosclerosis. Even after postponing treatment until the peak of adriamycin-induced proteinuria, valproic acid rapidly decreased the quantity of proteinuria and attenuated the progression of renal disease. Valproic acid abrogated the decrease in glomerular acetylation observed during adriamycin-induced nephropathy. Furthermore, valproic acid attenuated the significant upregulation of profibrotic and proinflammatory genes, the deposition of collagen, and the infiltration of macrophages into the kidney. Valproic acid decreased glomerular apoptosis and proliferation induced by adriamycin. Ultrastructural studies further supported the protective effect of valproic acid on podocytes in this model. Taken together, these data suggest that HDACs contribute to the pathogenesis of renal disease and that HDAC inhibitors may have therapeutic potential in CKD.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácido Valproico/uso terapéutico , Lesión Renal Aguda/prevención & control , Animales , Antibióticos Antineoplásicos , Modelos Animales de Enfermedad , Doxorrubicina , Evaluación Preclínica de Medicamentos , Femenino , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Ratones , Ratones Endogámicos BALB C , Proteinuria/prevención & control
10.
Biomaterials ; 290: 121817, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36208587

RESUMEN

Chronic liver disease can lead to liver fibrosis and ultimately cirrhosis, which is a significant health burden and a major cause of death worldwide. Reliable in vitro models are lacking and thus mono-cultures of cell lines are still used to study liver disease and evaluate candidate anti-fibrotic drugs. We established functional multicellular liver spheroid (MCLS) cultures using primary mouse hepatocytes, hepatic stellate cells, liver sinusoidal endothelial cells and Kupffer cells. Cell-aggregation and spheroid formation was enhanced with 96-well U-bottom plates generating over ±700 spheroids from one mouse. Extensive characterization showed that MCLS cultures contain functional hepatocytes, quiescent stellate cells, fenestrated sinusoidal endothelium and responsive Kupffer cells that can be maintained for 17 days. MCLS cultures display a fibrotic response upon chronic exposure to acetaminophen, and present steatosis and fibrosis when challenged with free fatty acid and lipopolysaccharides, reminiscent of non-alcoholic fatty liver disease (NAFLD) stages. Treatment of MCLS cultures with potential anti-NAFLD drugs such as Elafibranor, Lanifibranor, Pioglitazone and Obeticholic acid shows that all can inhibit steatosis, but only Elafibranor and especially Lanifibranor inhibit fibrosis. Therefore, primary mouse MCLS cultures can be used to model acute and chronic liver disease and are suitable for the assessment of anti-NAFLD drugs.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Esferoides Celulares , Ratones , Animales , Esferoides Celulares/metabolismo , Células Endoteliales , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Cirrosis Hepática/metabolismo , Hígado/patología , Hepatocitos
11.
Cell Mol Gastroenterol Hepatol ; 13(6): 1701-1716, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35219894

RESUMEN

BACKGROUND & AIMS: Liver fibrosis arises from long-term chronic liver injury, accompanied by an accelerated wound healing response with interstitial accumulation of extracellular matrix (ECM). Activated hepatic stellate cells (HSC) are the main source for ECM production. MicroRNA29a (miR-29a) is a crucial antifibrotic miRNA that is repressed during fibrosis, resulting in up-regulation of collagen synthesis. METHODS: Intracellular and extracellular miRNA levels of primary and immortalized myofibroblastic HSC in response to profibrogenic stimulation by transforming growth factor ß (TGFß) or platelet-derived growth factor-BB (PDGF-BB) or upon inhibition of vesicular transport and autophagy processes were determined by quantitative polymerase chain reaction. Autophagy flux was studied by electron microscopy, flow cytometry, immunoblotting, and immunocytochemistry. Hepatic and serum miR-29a levels were quantified by using both liver tissue and serum samples from a cohort of chronic hepatitis C virus patients and a murine CCl4 induced liver fibrosis model. RESULTS: In our study, we show that TGFß and PDGF-BB resulted in decrease of intracellular miR-29a and a pronounced increase of vesicular miR-29a release into the supernatant. Strikingly, miR-29a vesicular release was accompanied by enhanced autophagic activity and up-regulation of the autophagy marker protein LC3. Moreover, autophagy inhibition strongly prevented miR-29a secretion and repressed its targets' expression such as Col1A1. Consistently, hepatic miR-29a loss and increased LC3 expression in myofibroblastic HSC were associated with increased serum miR-29a levels in CCl4-treated murine liver fibrosis and specimens of hepatitis C virus patients with chronic liver disease. CONCLUSIONS: We provide evidence that activation-associated autophagy in HSC induces release of miR-29a, whereas inhibition of autophagy represses fibrogenic gene expression in part through attenuated miR-29a secretion.


Asunto(s)
Hepatitis C Crónica , MicroARNs/genética , Animales , Autofagia , Becaplermina/metabolismo , Células Estrelladas Hepáticas/patología , Hepatitis C Crónica/metabolismo , Hepatitis C Crónica/patología , Humanos , Cirrosis Hepática/patología , Ratones , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
12.
Cardiovasc Res ; 118(5): 1262-1275, 2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33909875

RESUMEN

AIMS: Hepatic capillaries are lined with specialized liver sinusoidal endothelial cells (LSECs) which support macromolecule passage to hepatocytes and prevent fibrosis by keeping hepatic stellate cells (HSCs) quiescent. LSEC specialization is co-determined by transcription factors. The zinc-finger E-box-binding homeobox (Zeb)2 transcription factor is enriched in LSECs. Here, we aimed to elucidate the endothelium-specific role of Zeb2 during maintenance of the liver and in liver fibrosis. METHODS AND RESULTS: To study the role of Zeb2 in liver endothelium we generated EC-specific Zeb2 knock-out (ECKO) mice. Sequencing of liver EC RNA revealed that deficiency of Zeb2 results in prominent expression changes in angiogenesis-related genes. Accordingly, the vascular area was expanded and the presence of pillars inside ECKO liver vessels indicated that this was likely due to increased intussusceptive angiogenesis. LSEC marker expression was not profoundly affected and fenestrations were preserved upon Zeb2 deficiency. However, an increase in continuous EC markers suggested that Zeb2-deficient LSECs are more prone to dedifferentiation, a process called 'capillarization'. Changes in the endothelial expression of ligands that may be involved in HSC quiescence together with significant changes in the expression profile of HSCs showed that Zeb2 regulates LSEC-HSC communication and HSC activation. Accordingly, upon exposure to the hepatotoxin carbon tetrachloride (CCl4), livers of ECKO mice showed increased capillarization, HSC activation, and fibrosis compared to livers from wild-type littermates. The vascular maintenance and anti-fibrotic role of endothelial Zeb2 was confirmed in mice with EC-specific overexpression of Zeb2, as the latter resulted in reduced vascularity and attenuated CCl4-induced liver fibrosis. CONCLUSION: Endothelial Zeb2 preserves liver angioarchitecture and protects against liver fibrosis. Zeb2 and Zeb2-dependent genes in liver ECs may be exploited to design novel therapeutic strategies to attenuate hepatic fibrosis.


Asunto(s)
Células Endoteliales , Cirrosis Hepática , Animales , Biomarcadores/metabolismo , Células Endoteliales/metabolismo , Endotelio , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/prevención & control , Ratones
13.
J Hepatol ; 55(6): 1353-60, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21803012

RESUMEN

BACKGROUND & AIMS: Autophagy is a metabolic process that degrades and recycles intracellular organelles and proteins with many connections to human disease and physiology. We studied the role of autophagy during hepatic stellate cell (HSC) activation, a key event in liver fibrogenesis. METHODS: Analysis of the autophagic flux during in vitro activation of primary mouse HSCs was performed using a DsRed-GFP-LC3B encoding plasmid. The effect of autophagy inhibition by bafilomycin A1 on the in vitro activation process of human and mouse HSCs was examined by measuring proliferation, presence of activation markers by RT-qPCR, immunofluorescence, and Western blotting. Analysis of lipid droplet and microtubule-associated protein light chain 3 beta (LC3B) colocalization in the presence of PDGF-BB was investigated by immunocytochemistry. RESULTS: A significant increased autophagic flux was observed during culture induced mouse HSC activation. Treatment of mouse HSCs and human HSCs with autophagy inhibitor bafilomycin A1 results in a significant decreased proliferation and expression of activation markers. In addition, lipid droplets and LC3B colocalization was increased after PDGF-BB treatment in quiescent HSCs. CONCLUSIONS: During HSC activation, autophagic flux is increased. The demonstration of partly inhibition of in vitro HSC activation after treatment with an autophagy inhibitor unveils a potential new therapeutic strategy for liver fibrosis.


Asunto(s)
Autofagia/fisiología , Células Estrelladas Hepáticas/fisiología , Actinas/genética , Animales , Autofagia/efectos de los fármacos , Becaplermina , Tetracloruro de Carbono/toxicidad , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Técnicas In Vitro , Metabolismo de los Lípidos/efectos de los fármacos , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Macrólidos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína-Lisina 6-Oxidasa , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Hepatology ; 51(2): 603-14, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19957378

RESUMEN

UNLABELLED: Hepatic stellate cell (HSC) activation is a pivotal step in the pathogenesis of liver fibrosis. The clarification of this transdifferentiation process is therefore important for the development of effective therapies for fibrosis. We analyzed the effect of a histone deacetylase inhibitor, valproic acid (VPA), on mouse HSC transdifferentiation in vitro and in vivo. The exposure of freshly isolated mouse HSCs to 2.5 mM VPA led to increased histone H4 acetylation and inhibited cell proliferation. Expression of stellate cell activation markers analyzed by quantitative polymerase chain reaction and western blotting revealed that treatment with VPA inhibited the induction of activation markers such as Acta2, Lox, Spp1, and Myh11. Treatment of mice with VPA decreased collagen deposition and in vivo activation of stellate cells in the livers of CCl(4)-treated mice. Class I histone deacetylase silencing through RNA interference in mouse HSCs only partially mimicked treatment with VPA. CONCLUSION: Chronic administration of VPA results in a marked decrease in stellate cell activation both in vitro and in vivo. We hypothesize that the VPA effect results partially from class I histone deacetylase inhibition, but that also non-histone deacetylase class I VPA targets are involved in the stellate cell activation process.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/fisiología , Histona Desacetilasa 1/antagonistas & inhibidores , Ácido Valproico/administración & dosificación , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ácido Valproico/farmacología
15.
Cancers (Basel) ; 13(23)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34885024

RESUMEN

Primary and secondary liver cancer are the third cause of death in the world, and as the incidence is increasing, liver cancer represents a global health burden. Current treatment strategies are insufficient to permanently cure patients from this devastating disease, and therefore other approaches are under investigation. The importance of cancer-associated fibroblasts (CAFs) in the tumour microenvironment is evident, and many pre-clinical studies have shown increased tumour aggressiveness in the presence of CAFs. However, it remains unclear how hepatic stellate cells are triggered by the tumour to become CAFs and how the recently described CAF subtypes originate and orchestrate pro-tumoural effects. Specialized in vitro systems will be needed to address these questions. In this review, we present the currently used in vitro models to study CAFs in primary and secondary liver cancer and highlight the trend from using oversimplified 2D culture systems to more complex 3D models. Relatively few studies report on the impact of cancer (sub)types on CAFs and the tumour microenvironment, and most studies investigated the impact of secreted factors due to the nature of the models.

16.
Front Med (Lausanne) ; 8: 750044, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34746184

RESUMEN

Liver sinusoidal endothelial cells have a gatekeeper function in liver homeostasis by permitting substrates from the bloodstream into the space of Disse and regulating hepatic stellate cell activation status. Maintenance of LSEC's highly specialized phenotype is crucial for liver homeostasis. During liver fibrosis and cirrhosis, LSEC phenotype and functions are lost by processes known as capillarization and LSEC dysfunction. LSEC capillarization can be demonstrated by the loss of fenestrae (cytoplasmic pores) and the manifestation of a basement membrane. Currently, no protein or genetic markers can clearly distinguish healthy from damaged LSECs in acute or chronic liver disease. Single cell (sc)RNA sequencing efforts have identified several LSEC populations in mouse models for liver disease and in human cirrhotic livers. Still, there are no clearly defined genesets that can identify LSECs or dysfunctional LSEC populations in transcriptome data. Here, we developed genesets that are enriched in healthy and damaged LSECs which correlated very strongly with healthy and early stage- vs. advanced human liver diseases. A damaged LSEC signature comprised of Fabp4/5 and Vwf/a1 was established which could efficiently identify damaged endothelial cells in single cell RNAseq data sets. In LSECs from an acute CCl4 liver injury mouse model, Fabp4/5 and Vwf/a1 expression is induced within 1-3 days while in cirrhotic human livers these 4 genes are highly enriched in damaged LSECs. In conclusion, our newly developed gene signature of damaged LSECs can be applicable to a wide range of liver disease etiologies, implicating a common transcriptional alteration mechanism in LSEC damage.

17.
Cell Death Dis ; 12(12): 1110, 2021 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-34839349

RESUMEN

Activated hepatic stellate cells (aHSC) are the main source of extra cellular matrix in liver fibrosis. Activation is classically divided in two phases: initiation and perpetuation. Currently, HSC-based therapeutic candidates largely focus on targeting the aHSCs in the perpetuation phase. However, the importance of HSC initiation during chronic liver disease (CLD) remains unclear. Here, we identified transcriptional programs of initiating and activated HSCs by RNA sequencing, using in vitro and in vivo mouse models of fibrosis. Importantly, we show that both programs are active in HSCs during murine and human CLD. In human cirrhotic livers, scar associated mesenchymal cells employ both transcriptional programs at the single cell level. Our results indicate that the transcriptional programs that drive the initiation of HSCs are still active in humans suffering from CLD. We conclude that molecules involved in the initiation of HSC activation, or in the maintenance of aHSCs can be considered equally important in the search for druggable targets of chronic liver disease.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Hepatopatías/fisiopatología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones
18.
Nat Protoc ; 16(5): 2542-2563, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33864055

RESUMEN

Hepatic stellate cells (HSCs) are nonparenchymal liver cells responsible for extracellular matrix homeostasis and are the main cells involved in the development of liver fibrosis following injury. The lack of reliable sources of HSCs has hence limited the development of complex in vitro systems to model liver diseases and toxicity. Here we describe a protocol to differentiate human induced pluripotent stem cells (iPSCs) into hepatic stellate cells (iPSC-HSCs). The protocol is based on the addition of several growth factors important for liver development sequentially over 12 d. iPSC-HSCs present phenotypic and functional characteristics of primary HSCs and can be expanded or frozen and used to perform high-throughput in vitro studies. We also describe how to coculture iPSC-HSCs with hepatocytes, which self-assemble into three-dimensional (3D) hepatic spheroids. This protocol enables the generation of HSC-like cells for in vitro modeling and drug screening studies.


Asunto(s)
Diferenciación Celular , Técnicas Citológicas/métodos , Células Estrelladas Hepáticas/citología , Células Madre Pluripotentes Inducidas/citología , Humanos , Esferoides Celulares/citología
19.
Biomaterials ; 261: 120335, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32891040

RESUMEN

A major obstacle in the development of efficient therapies for progressive liver fibrosis is the lack of representative in vitro models of liver fibrosis to aid in understanding the mechanisms of the disease and to promote the development of pharmaceuticals. Our aim was to develop a relevant in vitro mouse liver fibrosis model, based on the central hypothesis that liver fibrosis in vitro cannot be studied using only hepatic stellate cells (HSCs)-the main producer of scar tissue during fibrosis-, but requires cultures in which at least hepatocytes are integrated. We established robust methods to generate co-culture spheroids from freshly isolated mouse hepatocytes and HSCs. Characteristics and functionality of these spheroids were analyzed by qPCR of cell-type specific markers, CYP induction and immunohistochemistry. Compound toxicity was determined by ATP-assays. Hepatocytes and HSCs maintained their cell-type specific marker expression over a 15-day culture period without major hepatocyte dedifferentiation or HSC activation. Exposure of spheroids to TGFß can directly activate HSCs, while acetaminophen exposure mounts a hepatocyte damage dependent activation of HSCs. Pharmaceuticals with known anti-fibrotic properties, such as Valproic acid and Verteporfin, reduce HSC activation in response to hepatocyte damage in these cultures. A comparison between the fibrotic response of the spheroid co-cultures and in vivo activated HSCs showed that these 3D co-cultures are more representative than the commonly used 2D HSC monocultures. Finally, we showed that the 3D cultures can be integrated in microfluidic chips. We conclude that our hepatocyte-stellate cell-spheroid cultures are a robust in vitro model of liver fibrosis. This model could be used to further unravel the mechanism of HSC activation and facilitate the discovery of, or testing for novel anti-fibrotic compounds, as these spheroids better reproduce HSC in vivo activation compared to the more traditional 2D mono-culture models.


Asunto(s)
Células Estrelladas Hepáticas , Hígado , Animales , Fibrosis , Células Estrelladas Hepáticas/patología , Hepatocitos , Hígado/patología , Cirrosis Hepática/patología , Ratones
20.
EBioMedicine ; 43: 501-512, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31036530

RESUMEN

BACKGROUND: Platelet Derived Growth Factor Receptor beta (PDGFRß) has been associated to hepatic stellate cell activation and has been the target of multiple therapeutic studies. However, little is known concerning its use as a diagnostic agent. METHODS: Circulating PDGFRß levels were analysed in a cohort of patients with liver fibrosis/cirrhosis due to chronic alcohol abuse, viral hepatitis, or non-alcoholic fatty liver disease (NAFLD). The diagnostic performance of PDGFRß as individual blood parameter, or in combination with other metabolic factors was evaluated. FINDINGS: sPDGFRß levels are progressively increased with increasing fibrosis stage and the largest difference was observed in patients with significant fibrosis, compared to no or mild fibrosis. The accuracy of sPDGFRß-levels predicting fibrosis could be increased by combining it with albumin levels and platelet counts into a novel diagnostic algorithm, the PRTA-score, generating a predictive value superior to Fib-4, APRI, and AST/ALT. The sPDGFRß levels and the PRTA-score are independent of liver disease aetiology, thus overcoming one of the major weaknesses of current non-invasive clinical and experimental scores. Finally, we confirmed the diagnostic value of sPDGFRß levels and the PRTA-score in an independent patient cohort with NAFLD which was staged for fibrosis by liver biopsy. INTERPRETATION: The PRTA-score is an accurate tool for detecting significant liver fibrosis in a broad range of liver disease aetiologies. FUND: Vrije Universiteit Brussel, the Institute for the Promotion of Innovation through Science and Technology in Flanders (IWT-Flanders) (HILIM-3D; SBO140045), and the Fund of Scientific Research Flanders (FWO).


Asunto(s)
Hígado Graso Alcohólico/complicaciones , Hepatitis Viral Animal/complicaciones , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto , Algoritmos , Animales , Biomarcadores , Biopsia , Enfermedad Hepática Inducida por Sustancias y Drogas/complicaciones , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Células Estrelladas Hepáticas/metabolismo , Humanos , Cirrosis Hepática/diagnóstico , Masculino , Ratones , Persona de Mediana Edad , Curva ROC , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA