Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Hepatology ; 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38271673

ABSTRACT

BACKGROUND AND AIMS: Transforming growth factor-beta 1 (TGFß1) induces HSC activation into metastasis-promoting cancer-associated fibroblasts (CAFs), but how the process is fueled remains incompletely understood. We studied metabolic reprogramming induced by TGFß1 in HSCs. APPROACHES AND RESULTS: Activation of cultured primary human HSCs was assessed by the expression of myofibroblast markers. Glucose transporter 1 (Glut1) of murine HSC was disrupted by Cre recombinase/LoxP sequence derived from bacteriophage P1 recombination (Cre/LoxP). Plasma membrane (PM) Glut1 and glycolysis were studied by biotinylation assay and the Angilent Seahorse XFe96 Analyzer. S.c. HSC/tumor co-implantation and portal vein injection of MC38 colorectal cancer cells into HSC-specific Glut1 knockout mice were performed to determine in vivo relevance. Transcriptome was obtained by RNA sequencing of HSCs and spatialomics with MC38 liver metastases. TGFß1-induced CAF activation of HSCs was accompanied by elevation of PM Glut1, glucose uptake, and glycolysis. Targeting Glut1 or Src by short hairpin RNA, pharmacologic inhibition, or a Src SH3 domain deletion mutant abrogated TGFß1-stimulated PM accumulation of Glut1, glycolysis, and CAF activation. Mechanistically, binding of the Src SH3 domain to SH3 domain-binding protein 5 led to a Src/SH3 domain-binding protein 5/Rab11/Glut1 complex that activated Rab11-dependent Glut1 PM transport under TGFß1 stimulation. Deleting the Src SH3 domain or targeting Glut1 of HSCs by short hairpin RNA or Cre recombinase/LoxP sequence derived from bacteriophage P1 recombination suppressed CAF activation in mice and MC38 colorectal liver metastasis. Multi-omics revealed that Glut1 deficiency in HSCs/CAFs suppressed HSC expression of tumor-promoting factors and altered MC38 transcriptome, contributing to reduced MC38 liver metastases. CONCLUSION: The Src SH3 domain-facilitated metabolic reprogramming induced by TGFß1 represents a target to inhibit CAF activation and the pro-metastatic liver microenvironment.

2.
Hepatology ; 78(5): 1602-1624, 2023 11 01.
Article in English | MEDLINE | ID: mdl-36626639

ABSTRACT

Cancer cells often encounter hypoxic and hypo-nutrient conditions, which force them to make adaptive changes to meet their high demands for energy and various biomaterials for biomass synthesis. As a result, enhanced catabolism (breakdown of macromolecules for energy production) and anabolism (macromolecule synthesis from bio-precursors) are induced in cancer. This phenomenon is called "metabolic reprogramming," a cancer hallmark contributing to cancer development, metastasis, and drug resistance. HCC and cholangiocarcinoma (CCA) are 2 different liver cancers with high intertumoral heterogeneity in terms of etiologies, mutational landscapes, transcriptomes, and histological representations. In agreement, metabolism in HCC or CCA is remarkably heterogeneous, although changes in the glycolytic pathways and an increase in the generation of lactate (the Warburg effect) have been frequently detected in those tumors. For example, HCC tumors with activated ß-catenin are addicted to fatty acid catabolism, whereas HCC tumors derived from fatty liver avoid using fatty acids. In this review, we describe common metabolic alterations in HCC and CCA as well as metabolic features unique for their subsets. We discuss metabolism of NAFLD as well, because NAFLD will likely become a leading etiology of liver cancer in the coming years due to the obesity epidemic in the Western world. Furthermore, we outline the clinical implication of liver cancer metabolism and highlight the computation and systems biology approaches, such as genome-wide metabolic models, as a valuable tool allowing us to identify therapeutic targets and develop personalized treatments for liver cancer patients.


Subject(s)
Bile Duct Neoplasms , Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/pathology , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology
3.
Cell Rep ; 38(6): 110349, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139382

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) contains abundant myofibroblasts derived from hepatic stellate cells (HSCs) through an activation process mediated by TGF-ß. To determine the role of programmed death-ligand 1 (PD-L1) in myofibroblastic activation of HSCs, we disrupted PD-L1 of HSCs by shRNA or anti-PD-L1 antibody. We find that PD-L1, produced by HSCs, is required for HSC activation by stabilizing TGF-ß receptors I (TßRI) and II (TßRII). While the extracellular domain of PD-L1 (amino acids 19-238) targets TßRII protein to the plasma membrane and protects it from lysosomal degradation, a C-terminal 260-RLRKGR-265 motif on PD-L1 protects TßRI mRNA from degradation by the RNA exosome complex. PD-L1 is required for HSC expression of tumor-promoting factors, and targeting HSC PD-L1 by shRNA or Cre/loxP recombination suppresses HSC activation and ICC growth in mice. Thus, myofibroblast PD-L1 can modulate the tumor microenvironment and tumor growth by a mechanism independent of immune suppression.


Subject(s)
B7-H1 Antigen/metabolism , Hepatic Stellate Cells/metabolism , Liver Neoplasms/pathology , Myofibroblasts/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Animals , Cell Movement , Cell Proliferation/physiology , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Mice , Myofibroblasts/pathology , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Tumor Microenvironment/immunology
4.
J Hepatol ; 76(4): 921-933, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34953958

ABSTRACT

BACKGROUND & AIMS: Biliary disease is associated with a proliferative/fibrogenic ductular reaction (DR). p300 is an epigenetic regulator that acetylates lysine 27 on histone 3 (H3K27ac) and is activated during fibrosis. Long non-coding RNAs (lncRNAs) are aberrantly expressed in cholangiopathies, but little is known about how they recruit epigenetic complexes and regulate DR. We investigated epigenetic complexes, including transcription factors (TFs) and lncRNAs, contributing to p300-mediated transcription during fibrosis. METHODS: We evaluated p300 in vivo using tamoxifen-inducible, cholangiocyte-selective, p300 knockout (KO) coupled with bile duct ligation (BDL) and Mdr KO mice treated with SGC-CBP30. Primary cholangiocytes and liver tissue were analyzed for expression of Acta2-as1 lncRNA by qPCR and RNA in situ hybridization. In vitro, we performed RNA-sequencing in human cholangiocytes with a p300 inhibitor. Cholangiocytes were exposed to lipopolysaccharide (LPS) as an injury model. We confirmed formation of a p300/ELK1 complex by immunoprecipitation (IP). RNA IP was used to examine interactions between ACTA2-AS1 and p300. Chromatin IP assays were used to evaluate p300/ELK1 occupancy and p300-mediated H3K27ac. Organoids were generated from ACTA2-AS1-depleted cholangiocytes. RESULTS: BDL-induced DR and fibrosis were reduced in Krt19-CreERT/p300fl/fl mice. Similarly, Mdr KO mice were protected from DR and fibrosis after SGC-CBP30 treatment. In vitro, depletion of ACTA2-AS1 reduced expression of proliferative/fibrogenic markers, reduced LPS-induced cholangiocyte proliferation, and impaired organoid formation. ACTA2-AS1 regulated transcription by facilitating p300/ELK1 binding to the PDGFB promoter after LPS exposure. Correspondingly, LPS-induced H3K27ac was mediated by p300/ELK1 and was reduced in ACTA2-AS1-depleted cholangiocytes. CONCLUSION: Cholangiocyte-selective p300 KO or p300 inhibition attenuate DR/fibrosis in mice. ACTA2-AS1 influences recruitment of p300/ELK1 to specific promoters to drive H3K27ac and epigenetic activation of proliferative/fibrogenic genes. This suggests that cooperation between epigenetic co-activators and lncRNAs facilitates DR/fibrosis in biliary diseases. LAY SUMMARY: We identified a three-part complex containing an RNA molecule, a transcription factor, and an epigenetic enzyme. The complex is active in injured bile duct cells and contributes to activation of genes involved in proliferation and fibrosis.


Subject(s)
RNA, Long Noncoding , Animals , Bile Ducts/pathology , Cell Proliferation , Fibrosis , Lipopolysaccharides , Liver/pathology , Mice , Mice, Knockout , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
5.
Explor Res Hypothesis Med ; 6(3): 110-117, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34589656

ABSTRACT

Yes-associated protein-1 (YAP1) is a potent transcriptional co-activator and functions as an important downstream effector of the Hippo signaling pathway, which is key to regulating cell proliferation, apoptosis, and organ growth. YAP1 has been implicated as an oncogene for various human cancers including gastrointestinal cancers and hepatocellular carcinoma (HCC). YAP1 promotes tumorigenesis and cancer progression by multiple mechanisms, such as by promoting malignant phenotypes, expanding cancer stem cells, and inducing epithelial-mesenchymal transition. YAP1 overexpression or its activated forms are associated with advanced pathological grades and poor prognosis of cancer, and therefore targeting YAP1 may open a fertile avenue for cancer therapy. In this review, we summarize the recent evidence regarding the role of YAP1 in the carcinogenesis of gastrointestinal cancers and HCC.

6.
Hepatology ; 73(6): 2468-2483, 2021 06.
Article in English | MEDLINE | ID: mdl-33159815

ABSTRACT

BACKGROUND AND AIMS: During liver fibrosis, liver sinusoidal endothelial cells (LSECs) release angiocrine signals to recruit inflammatory cells into the liver. p300, a master regulator of gene transcription, is associated with pathological inflammatory response. Therefore, we examined how endothelial p300 regulates angiocrine signaling and inflammation related to portal hypertension and fibrogenesis. APPROACH AND RESULTS: CCl4 or partial inferior vena cava ligation (pIVCL) was used to induce liver injury. Mice with LSEC-specific p300 deletion (p300LSECΔ/Δ ) or C-C motif chemokine ligand 2 (Ccl2) deficiency, nuclear factor kappa B (NFκB)-p50 knockout mice, and bromodomain containing 4 (BRD4) inhibitors in wild-type mice were used to investigate mechanisms of inflammation regulation. Leukocytes were analyzed by mass cytometry by time-of-flight. Epigenetic histone marks were modified by CRISPR endonuclease-deficient CRISPR-associated 9-fused with the Krüppel associated box domain (CRISPR-dCas9-KRAB)-mediated epigenome editing. Portal pressure and liver fibrosis were reduced in p300LSECΔ/Δ mice compared to p300fl/fl mice following liver injury. Accumulation of macrophages was also reduced in p300LSECΔ/Δ mouse livers. Ccl2 was the most up-regulated chemokine in injured LSECs, but its increase was abrogated in p300LSECΔ/Δ mice. While the macrophage accumulation was increased in NFκB-p50 knockout mice with enhanced NFκB activity, it was reduced in mice with LSEC-specific Ccl2 deficiency and mice treated with specific BRD4 inhibitors. In vitro, epigenome editing of CCL2 enhancer and promoter regions by CRISPR-dCas9-KRAB technology repressed TNFα-induced CCL2 transcription through H3K9 trimethylation. In contrast, TNFα activated CCL2 transcription by promoting p300 interaction with NFκB and BRD4, leading to histone H3 lysine 27 acetylation at CCL2 enhancer and promoter regions. CONCLUSIONS: In summary, endothelial p300 interaction with NFκB and BRD4 increases CCL2 expression, leading to macrophage accumulation, portal hypertension, and liver fibrosis. Inhibition of p300 and its binding partners might serve as therapy in the treatment of liver diseases.


Subject(s)
Chemokine CCL2/metabolism , E1A-Associated p300 Protein/metabolism , Endothelial Cells/metabolism , Hypertension, Portal/metabolism , Liver Cirrhosis/metabolism , NF-kappa B p50 Subunit/metabolism , Nuclear Proteins , Transcription Factors , Animals , Cell Movement/drug effects , Chemotactic Factors , Drug Discovery , E1A-Associated p300 Protein/antagonists & inhibitors , Liver Cirrhosis/drug therapy , Mice , Mice, Knockout , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
7.
FASEB J ; 34(6): 7345-7359, 2020 06.
Article in English | MEDLINE | ID: mdl-32304339

ABSTRACT

TGFß induces the differentiation of hepatic stellate cells (HSCs) into tumor-promoting myofibroblasts but underlying mechanisms remain incompletely understood. Because endocytosis of TGFß receptor II (TßRII), in response to TGFß stimulation, is a prerequisite for TGF signaling, we investigated the role of protein diaphanous homolog 1 (known as Diaph1 or mDia1) for the myofibroblastic activation of HSCs. Using shRNA to knockdown Diaph1 or SMIFH2 to target Diaph1 activity of HSCs, we found that the inactivation of Diaph1 blocked internalization and intracellular trafficking of TßRII and reduced SMAD3 phosphorylation induced by TGFß1. Mechanistic studies revealed that the N-terminal portion of Diaph1 interacted with both TßRII and Rab5a directly and that Rab5a activity of HSCs was increased by Diaph1 overexpression and decreased by Diaph1 knockdown. Additionally, expression of Rab5aQ79L (active Rab5a mutant) increased whereas the expression of Rab5aS34N (inactive mutant) reduced the endosomal localization of TßRII in HSCs compared to the expression of wild-type Rab5a. Functionally, TGFß stimulation promoted HSCs to express tumor-promoting factors, and α-smooth muscle actin, fibronection, and CTGF, markers of myofibroblastic activation of HSCs. Targeting Diaph1 or Rab5a suppressed HSC activation and limited tumor growth in a tumor implantation mouse model. Thus, Dipah1 and Rab5a represent targets for inhibiting HSC activation and the hepatic tumor microenvironment.


Subject(s)
Endocytosis/physiology , Formins/metabolism , Hepatic Stellate Cells/metabolism , Myofibroblasts/metabolism , Receptors, Transforming Growth Factor beta/metabolism , rab5 GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Biomarkers/metabolism , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cell Transdifferentiation/physiology , HT29 Cells , Hepatic Stellate Cells/physiology , Humans , Male , Mice , Mice, Nude , Myofibroblasts/physiology , Phosphorylation/physiology , Signal Transduction/physiology , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
8.
Hepatol Commun ; 4(2): 268-283, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32025610

ABSTRACT

Transforming growth factor ß (TGFß) induces hepatic stellate cell (HSC) differentiation into tumor-promoting myofibroblast, although underlying mechanism remains incompletely understood. Focal adhesion kinase (FAK) is activated in response to TGFß stimulation, so it transmits TGFß stimulus to extracellular signal-regulated kinase and P38 mitogen-activated protein kinase signaling. However, it is unknown whether FAK can, in return, modulate TGFß receptors. In this study, we tested whether FAK phosphorylated TGFß receptor 2 (TGFßR2) and regulated TGFßR2 intracellular trafficking in HSCs. The FAKY397F mutant and PF-573,228 were used to inhibit the kinase activity of FAK, the TGFßR2 protein level was quantitated by immunoblotting, and HSC differentiation into myofibroblast was assessed by expression of HSC activation markers, alpha-smooth muscle actin, fibronectin, or connective tissue growth factor. We found that targeting FAK kinase activity suppressed the TGFßR2 protein level, TGFß1-induced mothers against decapentaplegic homolog phosphorylation, and myofibroblastic activation of HSCs. At the molecular and cellular level, active FAK (phosphorylated FAK at tyrosine 397) bound to TGFßR2 and kept TGFßR2 at the peripheral plasma membrane of HSCs, and it induced TGFßR2 phosphorylation at tyrosine 336. In contrast, targeting FAK or mutating Y336 to F on TGFßR2 led to lysosomal sorting and degradation of TGFßR2. Using RNA sequencing, we identified that the transcripts of 764 TGFß target genes were influenced by FAK inhibition, and that through FAK, TGFß1 stimulated HSCs to produce a panel of tumor-promoting factors, including extracellular matrix remodeling proteins, growth factors and cytokines, and immune checkpoint molecule PD-L1. Functionally, targeting FAK inhibited tumor-promoting effects of HSCs in vitro and in a tumor implantation mouse model. Conclusion: FAK targets TGFßR2 to the plasma membrane and protects TGFßR2 from lysosome-mediated degradation, thereby promoting TGFß-mediated HSC activation. FAK is a target for suppressing HSC activation and the hepatic tumor microenvironment.

9.
Liver Res ; 4(4): 212-217, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33520338

ABSTRACT

BACKGROUND AND AIM: The transcriptional co-activator Yes-associated protein-1 (YAP1) has been implicated as an oncogene and is overexpressed in different kinds of human cancers, especially hepatocellular carcinoma (HCC). However, the role of YAP1 has not been reported in residual/recurrent HCC after transarterial chemoembolization (TACE). Our aim is to determine whether YAP1 is overexpressed in the residual/recurrent HCC after TACE. METHODS: A total of 105 tumor tissues from 71 patients including 30 cases of primary HCC without prior treatment, 35 cases of residual/recurrent HCC post TACE, and 6 cases of hepatoblastoma were included in the immunohistochemical study. YAP1 immunoreactivity was blindly scored as 0, 1+, 2+ or 3+ in density and percentages of positive cells. RESULTS: About 33.3% (10/30) of primary HCC without prior treatment showed 2+ of YAP1 immunoreactivity. While 82.8% (29/35) of residual/recurrent HCCs after TACE treatment displayed 2-3+ of YAP1 immunoreactivity, which was significantly higher compared to primary HCC without prior treatment (P = 0.0002). YAP1 immunoreactivity was moderately to strongly positive (2-3+) in 100% of the hepatoblastoma, particularly in the embryonal components (3+ in 100% cases). CONCLUSIONS: YAP1 is significantly upregulated in the residual/recurrent HCCs post TACE treatment, suggesting that YAP1 may serve as a sensitive diagnostic marker and a treatment target for residual/recurrent HCC post TACE.

10.
Semin Liver Dis ; 40(1): 84-90, 2020 02.
Article in English | MEDLINE | ID: mdl-31683318

ABSTRACT

Chronic liver diseases, such as fibrosis and cancer, lead to a rigid or stiff liver because of perpetual activation of hepatic stellate cells or portal fibroblasts into matrix-producing myofibroblasts. Mechanical forces, as determined by the mechanical properties of extracellular matrix or pressure of circulating blood flow/shear stress, are sensed by mechanoreceptors at the plasma membrane and transmitted into a cell to impact cell function. This process is termed as mechanotransduction. This review includes basic knowledge regarding how external forces are sensed, amplified, and transmitted into the interior of a cell as far as the nucleus to regulate gene transcription and generate biological responses. It also reviews literatures to highlight the mechanisms by which mechanical forces in a normal or diseased liver influence the phenotype of hepatocytes, hepatic stellate cells, portal fibroblasts, and sinusoidal endothelial cells, and these cells in turn participate in the initiation and progression of liver diseases.


Subject(s)
Liver Diseases/pathology , Mechanotransduction, Cellular , Animals , Extracellular Matrix , Hepatocytes/metabolism , Humans , Myofibroblasts/metabolism
11.
Hepatology ; 70(4): 1409-1423, 2019 10.
Article in English | MEDLINE | ID: mdl-31004519

ABSTRACT

Nuclear translocation of mothers against decapentaplegic homolog 2/3 (SMAD2/3), core transcription factors of transforming growth factor ß (TGF-ß) signaling, is critical for hepatic stellate cell (HSC) differentiation into metastasis-promoting myofibroblasts. SMAD2/3 have multiple coactivators, including WW domain-containing transcription regulator protein 1 (WWTR1 or TAZ) and p300 acetyltransferase. In the nucleus, TAZ binds to SMAD2/3 to prevent SMAD2/3 nuclear export. However, how TAZ and SMAD2/3 enter the nucleus remains poorly understood because neither contains a nuclear localization signal (NLS), an amino acid sequence tagging proteins for nuclear transport. p300 is an NLS-containing large scaffold protein, so we hypothesized that SMAD2/3 and TAZ may undergo nuclear import through complexing with p300. Coimmunoprecipitation, immunofluorescence, and nuclear fractionation assays revealed that TGF-ß1 promoted binding of SMAD2/3 and TAZ to p300 and that p300 inactivation disrupted TGF-ß1-mediated SMAD2/3 and TAZ nuclear accumulation. Deleting the p300 NLS blocked TGF-ß1-induced SMAD2/3 and TAZ nuclear transport. Consistently, p300 inactivation suppressed TGF-ß1-mediated HSC activation and transcription of genes encoding tumor-promoting factors, such as connective tissue growth factor, Tenascin C, Periostin, platelet-derived growth factor C, and fibroblast growth factor 2, as revealed by microarray analysis. Chromatin immunoprecipitation-real-time quantitative PCR showed that canonical p300-mediated acetylation of histones also facilitated transcription in response to TGF-ß1 stimulation. Interestingly, although both TGF-ß1-mediated and stiffness-mediated HSC activation require p300, comparison of gene expression data sets revealed that transcriptional targets of TGF-ß1 were distinct from those of stiffness-p300 mechanosignaling. Lastly, in tumor/HSC coinjection and intrasplenic tumor injection models, targeting p300 of activated-HSC/myofibroblasts by C646, short hairpin RNA, or cre-mediated gene disruption reduced tumor and liver metastatic growth in mice. Conclusion: p300 facilitates TGF-ß1-stimulated HSC activation by both noncanonical (cytoplasm-to-nucleus shuttle for SMAD2/3 and TAZ) and canonical (histone acetylation) mechanisms. p300 is an attractive target for inhibiting HSC activation and the prometastatic liver microenvironment.


Subject(s)
Active Transport, Cell Nucleus/genetics , Hepatic Stellate Cells/metabolism , Liver Neoplasms/genetics , Smad2 Protein/genetics , p300-CBP Transcription Factors/genetics , Acyltransferases , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Blotting, Western , Cell Differentiation/genetics , Humans , Liver Neoplasms/pathology , Mice , Myofibroblasts/cytology , Myofibroblasts/metabolism , RNA, Small Interfering/genetics , Signal Transduction/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/pharmacology
12.
J Biol Chem ; 294(9): 3137-3151, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30610118

ABSTRACT

Transforming growth factor ß (TGFß) potently activates hepatic stellate cells (HSCs), which promotes production and secretion of extracellular matrix (ECM) proteins and hepatic fibrogenesis. Increased ECM synthesis and secretion in response to TGFß is associated with endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). TGFß and UPR signaling pathways are tightly intertwined during HSC activation, but the regulatory mechanism that connects these two pathways is poorly understood. Here, we found that TGFß treatment of immortalized HSCs (i.e. LX-2 cells) induces phosphorylation of the UPR sensor inositol-requiring enzyme 1α (IRE1α) in a SMAD2/3-procollagen I-dependent manner. We further show that IRE1α mediates HSC activation downstream of TGFß and that its role depends on activation of a signaling cascade involving apoptosis signaling kinase 1 (ASK1) and c-Jun N-terminal kinase (JNK). ASK1-JNK signaling promoted phosphorylation of the UPR-associated transcription factor CCAAT/enhancer binding protein ß (C/EBPß), which is crucial for TGFß- or IRE1α-mediated LX-2 activation. Pharmacological inhibition of C/EBPß expression with the antiviral drug adefovir dipivoxil attenuated TGFß-mediated activation of LX-2 or primary rat HSCs in vitro and hepatic fibrogenesis in vivo Finally, we identified a critical relationship between C/EBPß and the transcriptional regulator p300 during HSC activation. p300 knockdown disrupted TGFß- or UPR-induced HSC activation, and pharmacological inhibition of the C/EBPß-p300 complex decreased TGFß-induced HSC activation. These results indicate that TGFß-induced IRE1α signaling is critical for HSC activation through a C/EBPß-p300-dependent mechanism and suggest C/EBPß as a druggable target for managing fibrosis.


Subject(s)
Hepatic Stellate Cells/cytology , Transforming Growth Factor beta/metabolism , Unfolded Protein Response , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , E1A-Associated p300 Protein/metabolism , Endoribonucleases/metabolism , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Rats , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Unfolded Protein Response/drug effects
13.
NPJ Precis Oncol ; 2(1): 2, 2018.
Article in English | MEDLINE | ID: mdl-29872721

ABSTRACT

Extracellular matrix (ECM)-induced ß1-integrin-FAK signaling promotes cell attachment, survival, and migration of cancer cells in a distant organ so as to enable cancer metastasis. However, mechanisms governing activation of the ß1-integrin-FAK signaling remain incompletely understood. Here, we report that vasodilator-stimulated phosphoprotein (VASP), an actin binding protein, is required for ECM-mediated ß1-integrin-FAK-YAP1/TAZ signaling in gastrointestinal (GI) cancer cells and their liver metastasis. In patient-derived samples, VASP is upregulated in 53 of 63 colorectal cancers and 43 of 53 pancreatic ductal adenocarcinomas and high VASP levels correlate with liver metastasis and reduced patient survival. In a Matrigel-based 3-dimensional (3D) culture model, short hairpin RNA (shRNA)-mediated VASP knockdown in colorectal cancer cells (KM12L4, HCT116, and HT29) and pancreatic cancer cells (L3.6 and MIA PaCa-1) suppresses the growth of 3D cancer spheroids. Mechanistic studies reveal that VASP knockdown suppresses FAK phosphorylation and YAP1/TAZ protein levels, but not Akt or Erk-related pathways and that YAP1/TAZ proteins are enhanced by the ß1-integrin-FAK signaling. Additionally, VASP regulates the ß1-integrin-FAK-YAP1/TAZ signaling by at least two mechanisms: (1) promoting ECM-mediated ß1-integrin activation and (2) regulating YAP1/TAZ dephosphorylation at downstream of RhoA to enhance the stability of YAP1/TAZ proteins. In agreement with these, preclinical studies with two experimental liver metastasis mouse models demonstrate that VASP knockdown suppresses GI cancer liver metastasis, ß1-integrin activation, and YAP1/TAZ levels of metastatic cancer cells. Together, our data support VASP as a treatment target for liver metastasis of colorectal and pancreatic cancers.

14.
Gastroenterology ; 154(8): 2209-2221.e14, 2018 06.
Article in English | MEDLINE | ID: mdl-29454793

ABSTRACT

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) contribute to desmoplasia and stiffness of liver metastases by differentiating into matrix-producing myofibroblasts. We investigated whether stiffness due to the presence of tumors increases activation of HSCs into myofibroblasts and their tumor-promoting effects, as well as the role of E1A binding protein p300, a histone acetyltransferase that regulates transcription, in these processes. METHODS: HSCs were isolated from liver tissues of patients, mice in which the p300 gene was flanked by 2 loxP sites (p300F/F mice), and p300+/+ mice (controls). The HSCs were placed on polyacrylamide gels with precisely defined stiffness, and their activation (differentiation into myofibroblasts) was assessed by immunofluorescence and immunoblot analyses for alpha-smooth muscle actin. In HSCs from mice, the p300 gene was disrupted by cre recombinase. In human HSCs, levels of p300 were knocked down with small hairpin RNAs or a mutant form of p300 that is not phosphorylated by AKT (p300S1834A) was overexpressed. Human HSCs were also cultured with inhibitors of p300 (C646), PI3K signaling to AKT (LY294002), or RHOA (C3 transferase) and effects on stiffness-induced activation were measured. RNA sequencing and chromatin immunoprecipitation-quantitative polymerase chain reaction were used to identify HSC genes that changed expression levels in response to stiffness. We measured effects of HSC-conditioned media on proliferation of HT29 colon cancer cells and growth of tumors following subcutaneous injection of these cells into mice. MC38 colon cancer cells were injected into portal veins of p300F/Fcre and control mice, and liver metastases were measured. p300F/Fcre and control mice were given intraperitoneal injections of CCl4 to induce liver fibrosis. Liver tissues were collected and analyzed by immunofluorescence, immunoblot, and histology. RESULTS: Substrate stiffness was sufficient to activate HSCs, leading to nuclear accumulation of p300. Disrupting p300 level or activity blocked stiffness-induced activation of HSCs. In HSCs, substrate stiffness activated AKT signaling via RHOA to induce phosphorylation of p300 at serine 1834; this caused p300 to translocate to the nucleus, where it up-regulated transcription of genes that increase activation of HSCs and metastasis, including CXCL12. MC38 cells, injected into portal veins, formed fewer metastases in livers of p300F/Fcre mice than control mice. Expression of p300 was increased in livers of mice following injection of CCl4; HSC activation and collagen deposition were reduced in livers of p300F/Fcre mice compared with control mice. CONCLUSIONS: In studies of mice, we found liver stiffness to activate HSC differentiation into myofibroblasts, which required nuclear accumulation of p300. p300 increases HSC expression of genes that promote metastasis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/metabolism , E1A-Associated p300 Protein/metabolism , Hepatic Stellate Cells/pathology , Liver Neoplasms/pathology , Myofibroblasts/pathology , Animals , Benzoates/pharmacology , Carbon Tetrachloride/toxicity , Cell Nucleus/metabolism , Cell Transdifferentiation , E1A-Associated p300 Protein/genetics , Gene Expression Profiling , Gene Knockdown Techniques , HT29 Cells , Hepatic Stellate Cells/metabolism , Humans , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Mice, SCID , Myofibroblasts/metabolism , Nitrobenzenes , Phosphorylation , Primary Cell Culture , Pyrazoles/pharmacology , Pyrazolones , RNA, Small Interfering/antagonists & inhibitors , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/metabolism
15.
Mol Cancer Res ; 13(4): 604-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25548101

ABSTRACT

Cancer-associated fibroblasts (CAFs), the most abundant cells in the tumor microenvironment (TME), are a key source of the extracellular matrix (ECM) that constitutes the desmoplastic stroma. Through remodeling of the reactive tumor stroma and paracrine actions, CAFs regulate cancer initiation, progression, and metastasis, as well as tumor resistance to therapies. The CAFs found in stroma-rich primary hepatocellular carcinomas (HCC) and liver metastases of primary cancers of other organs predominantly originate from hepatic stellate cells (HSTC), which are pericytes associated with hepatic sinusoids. During tumor invasion, HSTCs transdifferentiate into myofibroblasts in response to paracrine signals emanating from either tumor cells or a heterogeneous cell population within the hepatic tumor microenvironment. Mechanistically, HSTC-to-myofibroblast transdifferentiation, also known as, HSTC activation, requires cell surface receptor activation, intracellular signal transduction, gene transcription, and epigenetic signals, which combined ultimately modulate distinct gene expression profiles that give rise to and maintain a new phenotype. The current review defines a paradigm that explains how HSTCs are activated into CAFs to promote liver metastasis. Furthermore, a focus on the most relevant intracellular signaling networks and epigenetic mechanisms that control HSTC activation is provided. Finally, we discuss the feasibility of targeting CAF/activated HSTCs, in isolation or in conjunction with targeting cancer cells, which constitutes a promising and viable therapeutic approach for the treatment of primary stroma-rich liver cancers and liver metastasis.


Subject(s)
Epigenesis, Genetic , Hepatic Stellate Cells/metabolism , Liver Neoplasms/pathology , Myofibroblasts/metabolism , Signal Transduction , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Transdifferentiation , Hepatic Stellate Cells/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Molecular Targeted Therapy , Myofibroblasts/pathology , Paracrine Communication
16.
Hepatology ; 61(1): 361-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24917558

ABSTRACT

UNLABELLED: Liver microenvironment is a critical determinant for development and progression of liver metastasis. Under transforming growth factor beta (TGF-ß) stimulation, hepatic stellate cells (HSCs), which are liver-specific pericytes, transdifferentiate into tumor-associated myofibroblasts that promote tumor implantation (TI) and growth in the liver. However, the regulation of this HSC activation process remains poorly understood. In this study, we tested whether vasodilator-stimulated phosphoprotein (VASP) of HSCs regulated the TGF-ß-mediated HSC activation process and tumor growth. In both an experimental liver metastasis mouse model and cancer patients, colorectal cancer cells reaching liver sinusoids induced up-regulation of VASP and alpha-smooth muscle actin (α-SMA) in adjacent HSCs. VASP knockdown in HSCs inhibited TGF-ß-mediated myofibroblastic activation of HSCs, TI, and growth in mice. Mechanistically, VASP formed protein complexes with TGF-ß receptor II (TßRII) and Rab11, a Ras-like small GTPase and key regulator of recycling endosomes. VASP knockdown impaired Rab11 activity and Rab11-dependent targeting of TßRII to the plasma membrane, thereby desensitizing HSCs to TGF-ß1 stimulation. CONCLUSIONS: Our study demonstrates a requirement of VASP for TGF-ß-mediated HSC activation in the tumor microenvironment by regulating Rab11-dependent recycling of TßRII to the plasma membrane. VASP and its effector, Rab11, in the tumor microenvironment thus present therapeutic targets for reducing TI and metastatic growth in the liver.


Subject(s)
Cell Adhesion Molecules/metabolism , Colorectal Neoplasms/pathology , Hepatic Stellate Cells/metabolism , Liver Neoplasms, Experimental/secondary , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HT29 Cells , Hepatic Stellate Cells/pathology , Humans , Liver Neoplasms, Experimental/metabolism , Mice , Microfilament Proteins/genetics , Myofibroblasts/pathology , Paracrine Communication , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
17.
Am J Physiol Gastrointest Liver Physiol ; 307(7): G749-59, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25169976

ABSTRACT

Platelet-derived growth factor (PDGF) and transforming growth factor-ß (TGF-ß) signaling are required for hepatic stellate cell (HSC) activation under pathological conditions such as liver metastatic tumor growth. These two signaling pathways are functionally divergent; PDGF signaling promotes proliferation and migration of HSCs, and TGF-ß induces transdifferentiation of quiescent HSCs into myofibroblasts. Although PDGF signaling is implicated in TGF-ß-mediated epithelial mesenchymal transition of tumor cells, the role of PDGF receptors in TGF-ß activation of HSCs has not been investigated. Here we report that PDGF receptor-α (PDGFR-α) is required for TGF-ß signaling of cultured human HSCs although HSCs express both PDGF-α and -ß receptors. PDGFR-α knockdown inhibits TGF-ß-induced phosphorylation and nuclear accumulation of SMAD2 with no influence on AKT or ERK phosphorylation associated with noncanonical TGF-ß signaling. PDGFR-α knockdown suppresses TGF-ß receptor I (TßRI) but increases TßRII gene transcription. At the protein level, PDGFR-α is recruited to TßRI/TßRII complexes by TGF-ß stimulation. PDGFR-α knockdown blocks TGF-ß-mediated internalization of TßRII and induces accumulation of TßRII at the plasma membrane, thereby inhibiting TGF-ß phosphorylation of SMAD2. Functionally, knockdown of PDGFR-α reduces paracrine effects of HSCs on colorectal cancer cell proliferation and migration in vitro. In mice and patients, colorectal cancer cell invasion of the liver induces upregulation of PDGFR-α of HSCs. In summary, our finding that PDGFR-α knockdown inhibits SMAD-dependent TGF-ß signaling by repressing TßRI transcriptionally and blocking endocytosis of TGF-ß receptors highlights a convergence of PDGF and TGF-ß signaling for HSC activation and PDGFR-α as a therapeutic target for liver metastasis and other settings of HSC activation.


Subject(s)
Hepatic Stellate Cells/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Active Transport, Cell Nucleus , Animals , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Gene Expression Regulation , HEK293 Cells , HT29 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice, SCID , Paracrine Communication , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad2 Protein/metabolism , Time Factors , Transfection
18.
Am J Pathol ; 184(10): 2791-802, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25111230

ABSTRACT

Sphingosine-1-phosphate (S1P) is produced by sphingosine kinase 1 and is implicated in tumor growth, although the mechanisms remain incompletely understood. Pancreatic stellate cells (PSCs) reside within the tumor microenvironment and may regulate tumor progression. We hypothesized that S1P activates PSCs to release paracrine factors, which, in turn, increase cancer cell invasion and growth. We used a combination of human tissue, in vitro, and in vivo studies to mechanistically evaluate this concept. Sphingosine kinase 1 was overexpressed in human pancreatic tissue, especially within tumor cells. S1P activated PSCs in vitro and conditioned medium from S1P-stimulated PSCs, increased pancreatic cancer cell migration, and invasion, which was dependent on S1P2, ABL1 (alias c-Abl) kinase, and matrix metalloproteinase-9. In vivo studies showed that pancreatic cancer cells co-implanted with S1P2 receptor knockdown PSCs led to less cancer growth and metastasis in s.c. and orthotopic pancreatic cancer models compared with control PSCs. Pancreatic cancer cell-derived S1P activates PSCs to release paracrine factors, including matrix metalloproteinase-9, which reciprocally promotes tumor cell migration and invasion in vitro and cancer growth in vivo.


Subject(s)
Lysophospholipids/metabolism , Matrix Metalloproteinase 9/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/metabolism , Tumor Microenvironment , Up-Regulation
19.
J Clin Invest ; 123(3): 1138-56, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23454766

ABSTRACT

In the tumor microenvironment, TGF-ß induces transdifferentiation of quiescent pericytes and related stromal cells into myofibroblasts that promote tumor growth and metastasis. The mechanisms governing myofibroblastic activation remain poorly understood, and its role in the tumor microenvironment has not been explored. Here, we demonstrate that IQ motif containing GTPase activating protein 1 (IQGAP1) binds to TGF-ß receptor II (TßRII) and suppresses TßRII-mediated signaling in pericytes to prevent myofibroblastic differentiation in the tumor microenvironment. We found that TGF-ß1 recruited IQGAP1 to TßRII in hepatic stellate cells (HSCs), the resident liver pericytes. Iqgap1 knockdown inhibited the targeting of the E3 ubiquitin ligase SMAD ubiquitination regulatory factor 1 (SMURF1) to the plasma membrane and TßRII ubiquitination and degradation. Thus, Iqgap1 knockdown stabilized TßRII and potentiated TGF-ß1 transdifferentiation of pericytes into myofibroblasts in vitro. Iqgap1 deficiency in HSCs promoted myofibroblast activation, tumor implantation, and metastatic growth in mice via upregulation of paracrine signaling molecules. Additionally, we found that IQGAP1 expression was downregulated in myofibroblasts associated with human colorectal liver metastases. Taken together, our studies demonstrate that IQGAP1 in the tumor microenvironment suppresses TßRII and TGF-ß dependent myofibroblastic differentiation to constrain tumor growth.


Subject(s)
Colorectal Neoplasms/metabolism , Liver Neoplasms/metabolism , Myofibroblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , ras GTPase-Activating Proteins/physiology , Adult , Aged , Aged, 80 and over , Animals , Cell Movement , Cell Proliferation , Cell Survival , Cell Transdifferentiation , Colorectal Neoplasms/pathology , Endosomes/metabolism , Female , Hepatic Stellate Cells/metabolism , Humans , Liver/pathology , Liver Neoplasms/secondary , Lysosomes/metabolism , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Pericytes/metabolism , Pericytes/physiology , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Transport , Proteolysis , Receptor, Transforming Growth Factor-beta Type II , Transforming Growth Factor beta1/physiology , Tumor Burden , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
20.
J Biol Chem ; 287(10): 7190-202, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22247553

ABSTRACT

The extracellular matrix microenvironment regulates cell phenotype and function. One mechanism by which this is achieved is the transactivation of receptor tyrosine kinases by specific matrix molecules. Here, we demonstrate that the provisional matrix protein, fibronectin (FN), activates fibroblast growth factor (FGF) receptor-1 (FGFR1) independent of FGF ligand in liver endothelial cells. FN activation of FGFR1 requires ß1 integrin, as evidenced by neutralizing antibody and siRNA-based studies. Complementary genetic and pharmacologic approaches identify that the non-receptor tyrosine kinase Src is required for FN transactivation of FGFR1. Whereas FGF ligand-induced phosphorylation of FGFR1 preferentially activates ERK, FN-induced phosphorylation of FGFR1 preferentially activates AKT, indicating differential downstream signaling of FGFR1 in response to alternate stimuli. Mutation analysis of known tyrosine residues of FGFR1 reveals that tyrosine 653/654 and 766 residues are required for FN-FGFR1 activation of AKT and chemotaxis. Thus, our study mechanistically dissects a new signaling pathway by which FN achieves endothelial cell chemotaxis, demonstrates how differential phosphorylation profiles of FGFR1 can achieve alternate downstream signals, and, more broadly, highlights the diversity of mechanisms by which the extracellular matrix microenvironment regulates cell behavior through transactivation of receptor tyrosine kinases.


Subject(s)
Chemotaxis/physiology , Endothelial Cells/metabolism , Fibronectins/metabolism , Integrin beta1/metabolism , Liver/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , src-Family Kinases/metabolism , Amino Acid Substitution , Animals , Endothelial Cells/cytology , Fibronectins/genetics , Humans , Integrin beta1/genetics , Liver/cytology , Mice , Mice, Knockout , Mutation, Missense , Phosphorylation/physiology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/physiology , src-Family Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...