Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Cell Physiol ; 236(11): 7682-7697, 2021 11.
Article in English | MEDLINE | ID: mdl-34041750

ABSTRACT

Hepatopulmonary syndrome (HPS) markedly increases the mortality of patients. However, its pathogenesis remains incompletely understood. Rat HPS develops in common bile duct ligation (CBDL)-induced, but not thioacetamide (TAA)-induced cirrhosis. We investigated the mechanisms of HPS by comparing these two models. Pulmonary histology, blood gas exchange, and the related signals regulating macrophage accumulation were assessed in CBDL and TAA rats. Anti-polymorphonuclear leukocyte (antiPMN) and anti-granulocyte-macrophage colony stimulating factor (antiGM-CSF) antibodies, clodronate liposomes (CL), and monocyte chemoattractant protein 1 (MCP1) inhibitor (bindarit) were administrated in CBDL rats, GM-CSF, and MCP1 were administrated in bone marrow-derived macrophages (BMDMs). Pulmonary inflammatory cell recruitment, vascular dilatation, and hypoxemia were progressively developed by 1 week after CBDL, but only occurred at 4 week after TAA. Neutrophils were the primary inflammatory cells within 3 weeks after CBDL and at 4 week after TAA. M2 macrophages were the primary inflammatory cells, meantime, pulmonary fibrosis, GM-CSFR, and CCR2 were specifically increased from 4 week after CBDL. AntiPMN antibody treatment decreased neutrophil and macrophage accumulation, CL or the combination of antiGM-CSF antibody and bindarit treatment decreased macrophage recruitment, resulting in pulmonary fibrosis, vascular dilatation, and hypoxemia in CBDL rats alleviated. The combination treatment of GM-CSF and MCP1 promoted cell migration, M2 macrophage differentiation, and transforming growth factor-ß1 (TGF-ß1) production in BMDMs. Conclusively, our results highlight neutrophil recruitment mediates pulmonary vascular dilatation and hypoxemia in the early stage of rat HPS. Further, M2 macrophage accumulation induced by GM-CSF/GM-CSFR and MCP1/CCR2 leads to pulmonary fibrosis and promotes vascular dilatation and hypoxemia, as a result, HPS develops.


Subject(s)
Hepatopulmonary Syndrome/etiology , Hypoxia/etiology , Lung/metabolism , Macrophages/metabolism , Microvessels/metabolism , Neutrophils/metabolism , Pulmonary Fibrosis/etiology , Animals , Biphenyl Compounds/blood , Cell Movement , Cell Proliferation , Chemokine CCL2/metabolism , Dilatation, Pathologic , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hepatopulmonary Syndrome/immunology , Hepatopulmonary Syndrome/metabolism , Hepatopulmonary Syndrome/pathology , Hypoxia/immunology , Hypoxia/metabolism , Hypoxia/pathology , Inflammation Mediators/metabolism , Leucine/analogs & derivatives , Leucine/blood , Liver Cirrhosis, Experimental/complications , Lung/immunology , Lung/pathology , Macrophages/immunology , Male , Microvessels/immunology , Microvessels/pathology , Neutrophil Infiltration , Neutrophils/immunology , Phenotype , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Rats, Sprague-Dawley , Receptors, CCR2/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Time Factors , Transforming Growth Factor beta1/metabolism
2.
J Cell Mol Med ; 23(8): 5542-5552, 2019 08.
Article in English | MEDLINE | ID: mdl-31144461

ABSTRACT

One central factor in hepatopulmonary syndrome (HPS) pathogenesis is pulmonary vascular remodelling (PVR) which involves dysregulation of proliferation and migration in pulmonary microvascular endothelial cells (PMVECs). Growing evidence suggests that Apical/basolateral polarity plays an important role in cell proliferation, migration, adhesion and differentiation. In this study, we explored whether cell polarity is involved and critical in experimental HPS rats that are induced by common bile duct ligation (CBDL). Cell polarity related proteins were analysed in CBDL rats lung and PMVECs under the HPS serum stimulation by immunofluorescence assay. Cdc42/PTEN activity, cell proliferation and migration and Annexin A2 (AX2) in PMVECs were determined, respectively. Cell polarity related proteins, lost their specialized luminal localization in PMVECs of the CBDL rat. The loss of cell polarity was induced by abnormal activity of Cdc42, which was strongly enhanced by the interaction between p-PTEN and Annexin A2 in PMVECs, after treatment with serum from CBDL rats. It led to over-proliferation and high migration ability of PMVECs. Down-regulation of PTEN-Cdc42 activity in PMVECs restored cell polarity and thus reduced their ability of migration and proliferation. Our study suggested that the loss of cell polarity plays a critical role in the pathogenesis of HPS-associated PVR and may become a potentially effective therapeutic target.


Subject(s)
Cell Polarity , Hepatopulmonary Syndrome/metabolism , Hepatopulmonary Syndrome/pathology , PTEN Phosphohydrolase/metabolism , cdc42 GTP-Binding Protein/metabolism , Actin Cytoskeleton/metabolism , Animals , Annexin A2/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Common Bile Duct/surgery , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hepatopulmonary Syndrome/blood , Ligation , Lung/blood supply , Male , Microvessels/pathology , Models, Biological , Rats, Sprague-Dawley
3.
Hepatology ; 68(2): 634-651, 2018 08.
Article in English | MEDLINE | ID: mdl-29023811

ABSTRACT

Hepatopulmonary syndrome (HPS) is a severe complication of cirrhosis with increased risk of mortality. Pulmonary microvascular alterations are key features of HPS; but underlying mechanisms are incompletely understood, and studies on HPS are limited to rats. Placental growth factor (PlGF), a proangiogenic molecule that is selectively involved in pathological angiogenesis, may play an important role in HPS development; however, its role has never been investigated. In this study, we validated an HPS model by common bile duct ligation (CBDL) in mice, investigated the kinetic changes in pulmonary angiogenesis and inflammation during HPS development, and provide evidence for a novel therapeutic strategy by targeting pathological angiogenesis. Mice with CBDL developed hypoxemia and intrapulmonary shunting on a background of liver fibrosis. Pulmonary alterations included increased levels of proangiogenic and inflammatory markers, which was confirmed in serum of human HPS patients. Increased PlGF production in HPS mice originated from alveolar type II cells and lung macrophages, as demonstrated by immunofluorescent staining. Dysfunctional vessel formation in CBDL mice was visualized by microscopy on vascular corrosion casts. Both prophylactic and therapeutic anti-PlGF (αPlGF) antibody treatment impeded HPS development, as demonstrated by significantly less intrapulmonary shunting and improved gas exchange. αPlGF treatment decreased endothelial cell dysfunction in vivo and in vitro and was accompanied by reduced pulmonary inflammation. Importantly, αPlGF therapy did not affect liver alterations, supporting αPlGF's ability to directly target the pulmonary compartment. CONCLUSION: CBDL in mice induces HPS, which is mediated by PlGF production; αPlGF treatment improves experimental HPS by counteracting pulmonary angiogenesis and might be an attractive therapeutic strategy for human HPS. (Hepatology 2017).


Subject(s)
Hepatopulmonary Syndrome/metabolism , Lung/pathology , Neovascularization, Pathologic/metabolism , Placenta Growth Factor/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers/metabolism , Common Bile Duct/surgery , Disease Models, Animal , Endoglin/blood , Hepatopulmonary Syndrome/physiopathology , Humans , Ligation/methods , Liver/pathology , Liver Cirrhosis/pathology , Male , Mice , Placenta Growth Factor/antagonists & inhibitors
4.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 676-684, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29233725

ABSTRACT

Recent studies have shown that pulmonary angiogenesis is an important pathological process in the development of hepatopulmonary syndrome (HPS), and growing evidence has indicated that Stromal cell-derived factor 1/C-X-C chemokine receptor type 4 (SDF-1/CXCR4) axis is involved in pulmonary vascular disease by mediating the accumulation of c-kit+ cells. This study aimed to test the effect of AMD3100, an antagonist of CXCR4, in HPS pulmonary angiogenesis. Common bile duct ligation (CBDL) rats were used as experimental HPS model and were treated with AMD3100 (1.25mg/kg/day, i.p.) or 0.9% saline for 3weeks. The sham rats underwent common bile duct exposure without ligation. The c-kit+ cells accounts and its angiogenic-related functions, prosurvival signals, pulmonary angiogenesis and arterial oxygenation were analysed in these groups. Our results showed that pulmonary SDF-1/CXCR4, Akt, Erk and VEGF/VEGFR2 were significantly activated in CBDL rats, and the numbers of circulating and pulmonary c-kit+ cells were increased in CBDL rats compared with control rats. Additionally, the angiogenic-related functions of c-kit+ cells and pulmonary microvessel counts were also elevated in CBDL rats. CXCR4 inhibition reduced pulmonary c-kit+ cells and microvessel counts and improved arterial oxygenation within 3weeks in CBDL rats. The pulmonary prosurvival signals and pro-angiogenic activity of c-kit+ cells were also down-regulated in AMD3100-treated rats. In conclusion, AMD3100 treatment attenuated pulmonary angiogenesis in CBDL rats and prevented the development of HPS via reductions in pulmonary c-kit+ cells and inhibition of the prosurvival signals. Our study provides new insights in HPS treatment.


Subject(s)
Hepatopulmonary Syndrome/pathology , Heterocyclic Compounds/pharmacology , Lung/drug effects , Neovascularization, Pathologic/prevention & control , Proto-Oncogene Proteins c-kit/metabolism , Animals , Benzylamines , Cells, Cultured , Common Bile Duct/pathology , Common Bile Duct/surgery , Cyclams , Down-Regulation/drug effects , Hepatopulmonary Syndrome/drug therapy , Hepatopulmonary Syndrome/metabolism , Heterocyclic Compounds/therapeutic use , Ligation , Lung/blood supply , Lung/pathology , Male , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Exp Cell Res ; 359(1): 266-274, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28729092

ABSTRACT

We have established that annexin A2 (ANXA2) is an important factor in the experimental hepatopulmonary syndrome (HPS) serum-induced proliferation of pulmonary arterial smooth muscle cells (PASMCs). However, the detailed mechanism remains unclear. ANXA2 translocated to the caveolin-enriched microdomains (caveolae) in PASMCs upon HPS serum stimulation. The disruption of caveolae by Methyl-ß-cyclodextrin (MßCD) alleviated the caveolae recruitment of ANXA2 and the ANXA2-mediated activation of ERK1/2 and NF-κB, so that ANXA2-modulated PASMC proliferation was suppressed. The over-expression of Cav-1 resulted in the relocation of ANXA2 from caveolae and negatively regulated ERK1/2 and NF-κB activation, which inhibited the ANXA2-modulated PASMC proliferative behavior. These data indicate that caveolae function as a signaling platform for ANXA2-induced proliferative behavior and Cav-1 participates upstream of ANXA2 in the activation of ERK1/2 and NF-κB.


Subject(s)
Annexin A2/metabolism , Caveolae/metabolism , Caveolin 1/metabolism , Hepatopulmonary Syndrome/metabolism , Hepatopulmonary Syndrome/pathology , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/pathology , Animals , Caveolin 1/chemistry , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatopulmonary Syndrome/blood , Male , NF-kappa B/metabolism , Protein Domains , Rats, Sprague-Dawley , beta-Cyclodextrins
6.
Clin Sci (Lond) ; 131(2): 159-168, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27879294

ABSTRACT

Hepatopulmonary syndrome (HPS) is a lung complication in various liver diseases, with high incidence, poor prognosis and no effective non-surgical treatments in patients with hepatocirrhosis. Therefore, assessing HPS pathogenesis to explore proper therapy strategies is clinically relevant. In the present study, male Sprague-Dawley rats underwent sham operation or common bile duct ligation (CBDL). Two weeks post-surgery, the following groups were set up for 2 weeks of treatment: sham + normal saline, CBDL + CXCR2 antagonist SB225002, CBDL + tumour necrosis factor α (TNF-α) antagonist PTX and CBDL + normal saline groups. Liver and lung tissues were collected after mean arterial pressure (MAP) and portal venous pressure (PVP) measurements. Haematoxylin and eosin (H&E) staining (lung) and Masson staining (liver) were performed for pathological analyses. Finally, pulmonary tissue RNA and total protein were assessed for target effectors. The mRNA and protein levels of CXCR2 were significantly increased in the pulmonary tissue of CBDL rats. What's more, CXCR2 inhibition by SB225002 reduced the expression of CD68 and von Willebrand factor (vWf) in CBDL rats. Importantly, CXCR2 inhibition suppressed the activation of Akt and extracellular signal-regulated kinase (ERK) in CBDL rats. Antagonization of TNF-α with PTX down-regulated the expression of CXCR2. During HPS pathogenesis in rats, CXCR2 might be involved in the accumulation of pulmonary intravascular macrophages and angiogenesis, possibly by activating Akt and ERK, with additional regulation by TNF-α that enhanced pulmonary angiogenesis by directly acting on the pulmonary tissue. Finally, the present study may provide novel targets for the treatment of HPS.


Subject(s)
Hepatopulmonary Syndrome/metabolism , Macrophages/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Common Bile Duct/drug effects , Common Bile Duct/metabolism , Common Bile Duct/surgery , Hepatopulmonary Syndrome/drug therapy , Hepatopulmonary Syndrome/genetics , Hepatopulmonary Syndrome/physiopathology , Humans , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Macrophages/drug effects , Male , Neovascularization, Pathologic , Phenylurea Compounds/administration & dosage , Portal Pressure/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-8B/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
7.
Biochem Biophys Res Commun ; 469(1): 70-75, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26616057

ABSTRACT

Hepatopulmonary syndrome (HPS) has been classically associated with intrapulmonary vasodilatation (IPVD) and pulmonary vascular remodelling (PVR), which are the key pathophysiological components of HPS and concerned frequently in the studies of HPS. Little is known about the relevance of pulmonary artery smooth muscle cells (PASMCs) migration or the molecular mechanisms of PVR in HPS. Annexin A2 (ANXA2) plays crucial role in HPS-associated PVR and might activate the activity of paxillin which as a regulatory protein participates in the regulation of PASMCs function in PVR. In addition, it has been identified that ANXA2 could influence the cells migration by some important signaling pathways in many diseases, including lung cancer, pulmonary hypertensionand and liver cancer. In this study, we performed scratch wound motility assay, modified boyden chamber, reverse transcription PCR, western blot and co-immunoprecipitation to determine the role of ANXA2 on HPS-associated PVR. We found that HPS rat serum from a common bile duct ligation (CBDL) rat model enhanced the migration of PASMCs and increased the expression of ANXA2 in PASMCs. We reported that ANXA2 and paxillin could form a co-immunoprecipitation. After silencing ANXA2 with siRNA, we found that the up-regulation of paxillin expression, induced by the HPS rat serum, was reversed. Additionally, we found that down-regulation of ANXA2 could significantly inhibit the migration of PASMCs. These findings indicated that down-regulation of ANXA2 by siRNA results in the inhibition of the aberrant dysregulation of paxillin and migration of PASMCs, which suggesting a potential therapeutic effect on HPS-associated PVR.


Subject(s)
Annexin A2/metabolism , Hepatopulmonary Syndrome/metabolism , Myocytes, Smooth Muscle/metabolism , Paxillin/metabolism , Pulmonary Artery/metabolism , Serum/metabolism , Animals , Cell Movement , Cells, Cultured , Down-Regulation , Hepatopulmonary Syndrome/pathology , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley
9.
J Cell Mol Med ; 19(10): 2453-61, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26147104

ABSTRACT

Hepatopulmonary syndrome (HPS) is characterized by a triad of severe liver disease, intrapulmonary vascular dilation and hypoxaemia. Pulmonary vascular remodelling (PVR) is a key feature of HPS pathology. Our previous studies have established the role of the pulmonary artery smooth muscle cell (PASMC) phenotypic modulation and proliferation in HPS-associated PVR. Myocardin, a robust transcriptional coactivator of serum response factor, plays a critical role in the vascular smooth muscle cell phenotypic switch. However, the mechanism regulating myocardin upstream signalling remains unclear. In this study, treatment of rat PASMCs with serum drawn from common bile duct ligation rats, which model symptoms of HPS, resulted in a significant increase in miR-9 expression correlated with a decrease in expression of myocardin and the phenotypic markers SM-α-actin and smooth muscle-specific myosin heavy chain (SM-MHC). Furthermore, miRNA functional analysis and luciferase reporter assay demonstrated that miR-9 effectively regulated myocardin expression by directly binding to its 3'-untranslated region. Both the knockdown of miR-9 and overexpression of myocardin effectively attenuated the HPS rat serum-induced phenotype switch and proliferation of PASMCs. Taken together, the findings of our present study demonstrate that miR-9 is required in HPS rat serum-induced phenotypic modulation and proliferation of PASMCs for targeting of myocardin and that miR-9 may serve as a potential therapeutic target in HPS.


Subject(s)
Gene Expression Regulation , Hepatopulmonary Syndrome/metabolism , MicroRNAs/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/genetics , Pulmonary Artery/pathology , Serum/metabolism , Trans-Activators/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Blotting, Western , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Down-Regulation/genetics , Gene Knockdown Techniques , Hepatopulmonary Syndrome/pathology , Male , MicroRNAs/genetics , Molecular Sequence Data , Nuclear Proteins/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Trans-Activators/metabolism , Up-Regulation/genetics
10.
Cell Physiol Biochem ; 37(4): 1289-300, 2015.
Article in English | MEDLINE | ID: mdl-26430741

ABSTRACT

BACKGROUND/AIMS: Pulmonary microvascular endothelial cell (PMVEC) proliferation and angiogenesis contribute to the development of hepatopulmonary syndrome (HPS). MicroRNA-199a-5p (miR-199a-5p) has emerged as a potent regulator of angiogenesis, and its expression levels significantly decrease in the serum of patients with hepatopathy. However, it has not been reported about whether miR-199a-5p might control PMVEC proliferation. Here, we described the miR-199a-5p governing PMVEC proliferation in HPS. METHODS: PMVECs were treated with rat serum from common bile duct ligation (CBDL) or sham. MiR-199a-5p mimic or inhibitor was used to change the miR-199a-5p expression. Knockdown of caveolin-1 (Cav-1) was performed using siRNA. NSC-23766 was used to inhibit Rac1 activity. Gene and protein expressions were quantified by qRT-PCR and western blot. Cell proliferation was analyzed by 3H-TdR incorporation and CCK-8 assays. Stress fibers were detected by immunofluorescence. RESULTS: CBDL rat serum induced the down-regulation of miR-199a-5p. Delivery of miR-199a-5p suppressed the CBDL rat serum-induced PMVEC proliferation whereas knockdown of miR-199a-5p promoted PMVEC proliferation. This was accompanied by a decrease and an increase in Cav-1 expression, respectively. Cav-1 siRNA abolished the enhancement of PMVEC proliferation induced by the miR-199a-5p inhibition. Although stress fibers were disrupted in Cav-1 deficient cells, NSC-23766 increased stress fibers and contributed to cell proliferation. CONCLUSIONS: CBDL rat serum induced down-regulation of miR-199a-5p in PMVECs, which led to an increase of Cav-1 gene expression. Increased Cav-1 expression, by inhibiting Rac1 activity, led to the formation of stress fibers, which contribute to PMVEC proliferation and thus the pathogenesis of HPS.


Subject(s)
MicroRNAs/metabolism , Aminoquinolines/pharmacology , Animals , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Proliferation , Cells, Cultured , Down-Regulation , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hepatopulmonary Syndrome/metabolism , Hepatopulmonary Syndrome/pathology , Lung/cytology , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotides, Antisense/metabolism , Pyrimidines/pharmacology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
11.
Am J Pathol ; 184(6): 1706-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24731444

ABSTRACT

Hepatic production and release of endothelin-1 (ET-1) binding to endothelin B (ETB) receptors, overexpressed in the lung microvasculature, is associated with accumulation of pro-angiogenic monocytes and vascular remodeling in experimental hepatopulmonary syndrome (HPS) after common bile duct ligation (CBDL). We have recently found that lung vascular monocyte adhesion and angiogenesis in HPS involve interaction of endothelial C-X3-C motif ligand 1 (CX3CL1) with monocyte CX3C chemokine receptor 1 (CX3CR1), although whether ET-1/ETB receptor activation influences these events is unknown. Our aim was to define if ET-1/ETB receptor activation modulates CX3CL1/CX3CR1 signaling and lung angiogenesis in experimental HPS. A selective ETB receptor antagonist, BQ788, was given for 2 weeks to 1-week CBDL rats. ET-1 (±BQ788) was given to cultured rat pulmonary microvascular endothelial cells overexpressing ETB receptors. BQ788 treatment significantly decreased lung angiogenesis, monocyte accumulation, and CX3CL1 levels after CBDL. ET-1 treatment significantly induced CX3CL1 production in lung microvascular endothelial cells, which was blocked by inhibitors of Ca(2+) and mitogen-activated protein kinase (MEK)/ERK pathways. ET-1-induced ERK activation was Ca(2+) independent. ET-1 administration also increased endothelial tube formation in vitro, which was inhibited by BQ788 or by blocking Ca(2+) and MEK/ERK activation. CX3CR1 neutralizing antibody partially inhibited ET-1 effects on tube formation. These findings identify a novel mechanistic interaction between the ET-1/ETB receptor axis and CX3CL1/CX3CR1 in mediating pulmonary angiogenesis and vascular monocyte accumulation in experimental HPS.


Subject(s)
Chemokine CX3CL1/metabolism , Endothelial Cells/metabolism , Endothelin-1/metabolism , Hepatopulmonary Syndrome/metabolism , Lung/metabolism , Neovascularization, Pathologic/metabolism , Receptor, Endothelin B/metabolism , Animals , Calcium Signaling , Cells, Cultured , Endothelial Cells/pathology , Hepatopulmonary Syndrome/pathology , Lung/blood supply , Lung/pathology , MAP Kinase Signaling System , Male , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Gastrointest Liver Physiol ; 306(1): G72-80, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24200956

ABSTRACT

Pulmonary vascular dilation and angiogenesis underlie experimental hepatopulmonary syndrome (HPS) induced by common bile duct ligation (CBDL) and may respond to receptor tyrosine kinase (RTK) inhibition. Vascular endothelial growth factor-A (VEGF-A) expression occurs in proliferating cholangiocytes and pulmonary intravascular monocytes after CBDL, the latter contributing to angiogenesis. CBDL cholangiocytes also produce endothelin-1 (ET-1), which triggers lung vascular endothelin B receptor-mediated endothelial nitric oxide synthase (eNOS) activation and pulmonary intravascular monocyte accumulation. However, whether RTK pathway activation directly regulates cholangiocyte and pulmonary microvascular alterations in experimental HPS is not defined. We assessed RTK pathway activation in cholangiocytes and lung after CBDL and the effects of the type II RTK inhibitor sorafenib in experimental HPS. Cholangiocyte VEGF-A expression and ERK activation accompanied proliferation and increased hepatic and circulating ET-1 levels after CBDL. Sorafenib decreased each of these events and led to a reduction in lung eNOS activation and intravascular monocyte accumulation. Lung monocyte VEGF-A expression and microvascular Akt and ERK activation were also found in vivo after CBDL, and VEGF-A activated Akt and ERK and angiogenesis in rat pulmonary microvascular endothelial cells in vitro. Sorafenib inhibited VEGF-A-mediated signaling and angiogenesis in vivo and in vitro and improved arterial gas exchange and intrapulmonary shunting. RTK activation in experimental HPS upregulates cholangiocyte proliferation and ET-1 production, leading to pulmonary microvascular eNOS activation, intravascular monocyte accumulation, and VEGF-A-mediated angiogenic signaling pathways. These findings identify a novel mechanism in cholangiocytes through which RTK inhibition ameliorates experimental HPS.


Subject(s)
Common Bile Duct , Endothelium, Vascular , Hepatopulmonary Syndrome , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Signal Transduction/drug effects , Animals , Common Bile Duct/metabolism , Common Bile Duct/pathology , Common Bile Duct/surgery , Disease Models, Animal , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Hepatopulmonary Syndrome/etiology , Hepatopulmonary Syndrome/metabolism , Hepatopulmonary Syndrome/pathology , Hepatopulmonary Syndrome/physiopathology , Ligation , Lung/blood supply , Male , Neovascularization, Pathologic/metabolism , Niacinamide/pharmacology , Nitric Oxide Synthase Type III/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Sorafenib , Vascular Endothelial Growth Factor A/metabolism
13.
Cell Physiol Biochem ; 34(5): 1768-79, 2014.
Article in English | MEDLINE | ID: mdl-25427750

ABSTRACT

BACKGROUND: Hepatopulmonary syndrome (HPS) is a serious complication of advanced liver disease that is characterised by intrapulmonary vascular dilatation (IPVD) and arterial hypoxemia. Pulmonary vascular remodelling (PVR) is an important pathological feature of HPS, but the potential mechanisms underlying PVR remain undefined. Recent findings have established the essential role of changes in Annexin A2 (ANXA2) in controlling the phenotypic modulation of pulmonary artery smooth muscle cells (PASMCs) in PVR associated with HPS. However, the mechanism by which upstream signalling regulates ANXA2 is unclear. METHODS: In the present study, computational analysis was used to predict which miRNA might target the 3´-untranslated region (3´-UTR) of the ANXA2 mRNA. Real-time PCR and western blotting were performed to study the level of correlation between ANXA2 and the differentiation marker with the predicted miRNAs in PASMCs stimulated with serum from normal rats or those with HPS. Functional analysis of the miRNA and a luciferase reporter assay were performed to demonstrate that the predicted miRNA suppressed ANXA2 expression by directly targeting the predicted 3´-UTR site of the ANXA2 mRNA. RESULTS: Computational analysis predicted that miR-206 would target the 3´-UTR of ANXA2 mRNA. In HPS rat serum-stimulated PASMCs, the expression of miR-206 displayed an inverse correlation with ANXA2, while a positive correlation was observed with the phenotypic marker smooth muscle α-actin (SM α-actin). The miRNA functional analysis and luciferase reporter assay demonstrated that miR-206 effectively downregulated the expression of ANXA2 by binding to the 3´-UTR of the ANXA2 mRNA. Consistently, miR-206 effectively inhibited the HPS rat serum-induced phenotypic modulation and proliferation, while these effects were reversed in ANXA2-overexpressing PASMCs. CONCLUSION: This study demonstrates that miR-206 inhibits the HPS rat serum-induced phenotypic modulation and proliferation in PASMCs by down-regulating ANXA2 gene expression.


Subject(s)
Annexin A2/genetics , Annexin A2/metabolism , Hepatopulmonary Syndrome/metabolism , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Serum/metabolism , 3' Untranslated Regions/genetics , Actins/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Down-Regulation/genetics , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
14.
Transl Res ; 271: 93-104, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38797433

ABSTRACT

Hepatopulmonary syndrome (HPS) is a serious pulmonary complication in the advanced stage of liver disease. The occurrence of pulmonary edema in HPS patients is life-threatening. Increased pulmonary vascular permeability is an important mechanism leading to pulmonary edema, and endothelial glycocalyx (EG) is a barrier that maintains stable vascular permeability. However, in HPS, whether the pulmonary vascular EG changes and its regulatory mechanism are still unclear. Spleen derived monocytes are involved in the pathogenesis of HPS. However, whether they regulate the pulmonary vascular permeability in HPS patients or rats and what is the mechanism is still unclear. Healthy volunteers and HPS patients with splenectomy or not were enrolled in this study. We found that the respiration of HPS patients was significantly improved in response to splenectomy, while the EG degradation and pulmonary edema were aggravated. In addition, HPS patients expressed higher levels of oncostatin M (OSM) and fibroblast growth factor (FGF). Subsequently, the co-culture system of monocytes and human umbilical vein endothelial cells (HUVECs) was constructed. It was found that monocytes secreted OSM and activated the FGF/FGFR1 signaling pathway in HUVECs. Then, an HPS rat model was constructed by common bile duct ligation (CBDL) for in vivo verification. HPS rats were intravenously injected with OSM recombinant protein and/or TNF-α into the rats via tail vein 30 min before CBDL. The results showed that the respiration of HPS rats was improved after splenectomy, while the degradation of EG in pulmonary vessels and vascular permeability were increased, and pulmonary edema was aggravated. Moreover, the expression of OSM and FGF was upregulated in HPS rats, while both were downregulated after splenectomy. Intravenous injection of exogenous OSM eliminated the effect of splenectomy on FGF and improved EG degradation. It can be seen that during HPS, spleen-derived monocytes secrete OSM to promote pulmonary vascular EG remodeling by activating the FGF/FGFR1 pathway, thereby maintaining stable vascular permeability, and diminishing pulmonary edema. This study provides a promising therapeutic target for the treatment of HPS.


Subject(s)
Capillary Permeability , Hepatopulmonary Syndrome , Monocytes , Oncostatin M , Receptor, Fibroblast Growth Factor, Type 1 , Signal Transduction , Spleen , Animals , Humans , Hepatopulmonary Syndrome/metabolism , Male , Monocytes/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Spleen/metabolism , Oncostatin M/metabolism , Fibroblast Growth Factors/metabolism , Rats , Human Umbilical Vein Endothelial Cells/metabolism , Splenectomy , Rats, Sprague-Dawley , Lung/metabolism , Lung/blood supply , Female , Middle Aged , Adult , Glycocalyx/metabolism
15.
J Gastroenterol Hepatol ; 28(2): 213-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23190201

ABSTRACT

Hepatopulmonary syndrome (HPS) is an important cause of dyspnea and hypoxia in the setting of liver disease, occurring in 10-30% of patients with cirrhosis. It is due to vasodilation and angiogenesis in the pulmonary vascular bed, which leads to ventilation-perfusion mismatching, diffusion limitation to oxygen exchange, and arteriovenous shunting. There is evidence, primarily from animal studies, that vasodilation is mediated by a number of endogenous vasoactive molecules, including endothelin-1 and nitric oxide (NO). In experimental HPS, liver injury stimulates release of endothelin-1 and results in increased expression of ET(B) receptors on pulmonary endothelial cells, leading to upregulation of endothelial NO synthase (eNOS) and subsequent increased production of NO, which causes vasodilation. In addition, increased phagocytosis of bacterial endotoxin in the lung not only promotes stimulation of inducible NO synthase, which increases NO production, but also contributes to intrapulmonary accumulation of monocytes, which may stimulate angiogenesis via vascular endothelial growth factor pathway. Despite these insights into the pathogenesis of experimental HPS, there is no established medical therapy, and liver transplantation remains the main treatment for symptomatic HPS, although selected patients may benefit from other surgical or radiological interventions. In this review, we focus on recent advances in our understanding of the pathophysiology of HPS, and discuss current approaches to the investigation and treatment of this condition.


Subject(s)
Hepatopulmonary Syndrome , Liver Cirrhosis/complications , Lung/blood supply , Pulmonary Artery/physiopathology , Animals , Early Diagnosis , Endothelin-1/metabolism , Hepatopulmonary Syndrome/diagnosis , Hepatopulmonary Syndrome/etiology , Hepatopulmonary Syndrome/metabolism , Hepatopulmonary Syndrome/physiopathology , Hepatopulmonary Syndrome/therapy , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/physiopathology , Lung/physiopathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Predictive Value of Tests , Prognosis , Pulmonary Artery/metabolism , Pulmonary Circulation , Vasodilation
16.
Cardiol Young ; 23(5): 629-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23458130

ABSTRACT

Severe cyanosis due to pulmonary arteriovenous fistulas occurs often after a bidirectional superior cavopulmonary anastomosis (Glenn operation) and also in some congenital anomalies in which hepatic venous blood bypasses the lungs in the first passage. Relocation of hepatic flow into the lungs usually causes these fistulas to disappear. Similar pulmonary arteriovenous fistulas are observed in hereditary haemorrhagic telangiectasia, and in liver disease (hepatopulmonary syndrome). There is no convincing identification yet of a responsible hepatic factor that produces these lesions. Candidates for such a factor are reviewed, and the possibility of angiotensin or bradykinin contributing to the fistulas is discussed.


Subject(s)
Angiotensins/metabolism , Arteriovenous Fistula/metabolism , Bradykinin/metabolism , Cyanosis/etiology , Fontan Procedure , Hepatopulmonary Syndrome/metabolism , Postoperative Complications/metabolism , Pulmonary Artery/abnormalities , Pulmonary Veins/abnormalities , Telangiectasia, Hereditary Hemorrhagic/metabolism , Arteriovenous Fistula/complications , Hepatic Veins/abnormalities , Hepatopulmonary Syndrome/complications , Humans , Liver/blood supply , Pulmonary Artery/metabolism , Pulmonary Veins/metabolism , Telangiectasia, Hereditary Hemorrhagic/complications
17.
Zhonghua Gan Zang Bing Za Zhi ; 21(9): 701-4, 2013 Sep.
Article in Zh | MEDLINE | ID: mdl-24160348

ABSTRACT

OBJECTIVE: To determine the lung expression of tissue factor (TF) mRNA in hepatopulmonary syndrome (HPS) using a rat model system and to investigate the potential significance of its differential expression. METHODS: Forty male Sprague-Dawley rats were used to establish models of cirrhosis (n = 20) and HPS (n = 20). Blood gas analysis was used to investigate the effects of each model on pulmonary function. Effects on the expression of TF mRNA in lung were determined by qRT-PCR and on lung pathology by histological analysis. RESULTS: The HPS rats showed significantly lower PaO2 than the cirrhosis rats (58.20 +/- 3.19 mmHg vs. 85.00 +/- 2.53 mmHg, P less than 0.05) but significantly higher TF mRNA expression in lung (0.77 +/- 0.22 vs. 0.33 +/- 0.14, P less than 0.05). TF mRNA expression was negatively correlated with the value of PaO2 (r = -0.565, P less than 0.05). The lungs of the cirrhosis rats showed widened alveolar intervals, diversified sizes of alveolar spaces, reduced lung capacity, inflammatory cell infiltration, and hyperemia in the pulmonary vessels. The lungs of the HPS rats showed all of the same changes but also with accumulated macrophages and micro-thrombosis in the pulmonary vessels. Among the HPS rats, those with micro-thrombosis in pulmonary vessels showed a greater increase in TF mRNA expression than those without (0.68 +/- 0.17 vs. 0.40 +/- 0.12, P less than 0.05). CONCLUSION: The expression of TF mRNA in lung of hepatopulmonary syndrome model rats was elevated and might increase the incidence of thromboembolism in the lung.


Subject(s)
Hepatopulmonary Syndrome/metabolism , Lung/metabolism , Thromboplastin/metabolism , Animals , Disease Models, Animal , Hepatopulmonary Syndrome/genetics , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Thromboplastin/genetics
18.
J Hepatol ; 57(4): 752-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22659346

ABSTRACT

BACKGROUND & AIMS: Hepatopulmonary syndrome (HPS), classically attributed to intrapulmonary vascular dilatation, occurs in 15-30% of cirrhotics and causes hypoxemia and increases mortality. In experimental HPS after common bile duct ligation (CBDL), monocytes adhere in the lung vasculature and produce vascular endothelial growth factor (VEGF)-A and angiogenesis ensues and contribute to abnormal gas exchange. However, the mechanisms for these events are unknown. The chemokine fractalkine (CX(3)CL1) can directly mediate monocyte adhesion and activate VEGF-A and angiogenesis via its receptor CX(3)CR1 on monocytes and endothelium during inflammatory angiogenesis. We explored whether pulmonary CX(3)CL1/CX(3)CR1 alterations occur after CBDL and influence pulmonary angiogenesis and HPS. METHODS: Pulmonary CX(3)CL1/CX(3)CR1 expression and localization, CX(3)CL1 signaling pathway activation, monocyte accumulation, and development of angiogenesis and HPS were assessed in 2- and 4-week CBDL animals. The effects of a neutralizing antibody to CX(3)CR1 (anti-CX(3)CR1 Ab) on HPS after CBDL were evaluated. RESULTS: Circulating CX(3)CL1 levels and lung expression of CX(3)CL1 and CX(3)CR1 in intravascular monocytes and microvascular endothelium increased in 2- and 4-week CBDL animals as HPS developed. These events were accompanied by pulmonary angiogenesis, monocyte accumulation, activation of CX(3)CL1 mediated signaling pathways (Akt, ERK) and increased VEGF-A expression and signaling. Anti-CX(3)CR1 Ab treatment reduced monocyte accumulation, decreased lung angiogenesis and improved HPS. These events were accompanied by inhibition of CX(3)CL1 signaling pathways and a reduction in VEGF-A expression and signaling. CONCLUSIONS: Circulating CX(3)CL1 levels and pulmonary CX(3)CL1/CX(3)CR1 expression and signaling increase after CBDL and contribute to pulmonary intravascular monocyte accumulation, angiogenesis and development of experimental HPS.


Subject(s)
Chemokine CX3CL1/metabolism , Hepatopulmonary Syndrome/metabolism , Lung/blood supply , Neovascularization, Pathologic/metabolism , Receptors, Chemokine/metabolism , Animals , Antibodies/pharmacology , CX3C Chemokine Receptor 1 , Cell Adhesion/drug effects , Chemokine CX3CL1/blood , Common Bile Duct/surgery , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatopulmonary Syndrome/blood , Hepatopulmonary Syndrome/pathology , Ligation , Liver/pathology , Lung/metabolism , Lung/pathology , Male , Monocytes/physiology , Portal Pressure/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Chemokine/immunology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Biomed Pharmacother ; 137: 111263, 2021 May.
Article in English | MEDLINE | ID: mdl-33516071

ABSTRACT

BACKGROUND & AIMS: Hepatopulmonary syndrome (HPS) is characterized by pulmonary vasodilation and arterial blood oxygen desaturation in patients with chronic liver disease. Generally, common bile duct ligation (CBDL) rats are a suitable experimental model for studying hepatopulmonary syndrome. Our previous study demonstrated that endotoxin surges markedly, followed by bacterial translocation and the loss of liver immune function in all the stages of CBDL, thereby contributing to the pathogenesis of HPS. However, the mechanisms behind the increase of the endotoxin and how to alleviate it have not yet been elucidated. Pulmonary injury induced by increased bilirubin, endotoxin, and inflammatory mediators occurs in the early and later stages of CBDL. This study assessed the effects of Tea polyphenols (TP) and Levofloxacin on endotoxin reduction and suppression of lung injury in HPS rats in the long and short term, respectively. METHODS: Morphological change of pulmonary injury, HPS relative index, endotoxin concentration, and the activation extent of Malondialdehyde (MDA) and Myeloperoxidase (MPO) were evaluated in CBDL rats with or without TP and Levofloxacin for three weeks or six weeks. The inflammation factors of serum, lung tissue, and BALF were then compared at the same condition for the two time periods. This was followed by adoption of the network pharmacology approach, which was mainly composed of active component gathering, target prediction, HPS gene collection, network analysis, and gene enrichment analysis. Finally, the mRNA and protein levels of the inflammatory factors were studied and relative signaling expression was assessed using RT-PCR and Western blot analysis. RESULTS: The obtained results indicated that the pulmonary injury manifestation was perceived and endotoxin, MDA, and MPO activation were markedly increased in the early and later stages of CBDL. TP and Levofloxacin treatment alleviated endotoxin infection and inflammation factor expression three weeks and six weeks after CBDL. In addition, Levofloxacin displayed a short time anti-bacterial effect, while TP exerted a long period function. TP and Levofloxacin also reduced TNF-α, TGF-ß, IL-1ß, PDGF-BB, NO, ICAM-1, and ET-1 expression on the mRNA or protein expression. With regard to the pharmacological mechanism, the network analysis indicated that 12 targets might be the therapeutic targets of TP and Levofloxacin on HPS, namely ET-1, NOs3, VEGFa, CCl2, TNF, Ptgs2, Hmox1, Alb, Ace, Cav1, and Mmp9. The gene enrichment analysis implied that TP and Levofloxacin probably benefited patients with HPS by modulating pathways associated with the AGE-RAGE signaling pathway, the TNF signaling pathway, the HIF-1 signaling pathway, the VEGF signaling pathway, and the IL-17 signaling pathway, Rheumatoid arthritis, Fluid shear stress, and atherosclerosis. Finally, the TNF-α level was mainly diminished on the protein level following CBDL. CONCLUSIONS: TP and Levofloxacin could alleviate pulmonary injury for short and long period, respectively, while at the same time preventing endotoxin and the development of HPS in CBDL rats. These effects are possibly associated with the regulation of the Endotoxin -TNF-α pathways.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Endotoxins/metabolism , Hepatopulmonary Syndrome/prevention & control , Levofloxacin/pharmacology , Lung Injury/prevention & control , Lung/drug effects , Polyphenols/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Bacterial Translocation , Camellia sinensis , Common Bile Duct/surgery , Disease Models, Animal , Hepatopulmonary Syndrome/metabolism , Hepatopulmonary Syndrome/microbiology , Hepatopulmonary Syndrome/pathology , Ligation , Lung/metabolism , Lung/pathology , Lung Injury/metabolism , Lung Injury/microbiology , Lung Injury/pathology , Male , Protein Interaction Maps , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
20.
Liver Transpl ; 16(7): 870-3, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20583080

ABSTRACT

Spontaneous resolution of hepatopulmonary syndrome (HPS) without liver transplantation or improvement in the underlying liver disease has rarely been reported in the literature. Increased endogenous production of nitric oxide has been implicated in the pathogenesis of HPS. We report the case of a 50-year-old man with hepatitis C cirrhosis who demonstrated dramatic improvement in HPS after withdrawal from chronic methadone therapy. We speculate on the potential role of opiate receptors in the pulmonary vasculature and their effect on nitric oxide signaling as a potential mechanism accounting for the patient's clinical improvement.


Subject(s)
Hepatopulmonary Syndrome/rehabilitation , Methadone/therapeutic use , Narcotics/therapeutic use , Withholding Treatment , Hepatitis C/complications , Hepatopulmonary Syndrome/etiology , Hepatopulmonary Syndrome/metabolism , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/virology , Male , Middle Aged , Nitric Oxide/metabolism , Receptors, Opioid/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL