Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 2235: 181-190, 2021.
Article in English | MEDLINE | ID: mdl-33576978

ABSTRACT

Tumor progression and metastasis are multistep processes that are critically dependent on the interaction of metastasizing tumor cells with other cells of the local microenvironment. Mimicking the single steps of the metastatic cascade in vitro is therefore challenging when investigating not only tumor cell behavior alone but also cellular crosstalk between different cell populations. In particular, the crosstalk of tumor cells with pericytes and endothelial cells when accessing the bloodstream is of great importance for successful intravasation and metastatic dissemination. To examine metastatic intravasation and analyze the interaction of tumor cells with pericytes, which reside within the basement membrane and endothelial cells, aligning the vessel wall, we have designed a 3D in vitro transwell assay mimicking tumor cell intravasation into a vessel. Modifying the Boyden chamber transwell assay by seeding first an endothelial cell layer onto the transwell membrane and covering it with pericytes before adding the tumor cells allows us to investigate the role of pericytes and endothelial cells on tumor cell intravasation and at the same time to study their crosstalk at the molecular level and how this interaction influences tumor cell behavior. It further allows the manipulation of the system for proof-of-principle experimentation.


Subject(s)
Cell Culture Techniques/methods , Neoplasm Invasiveness/pathology , Pericytes/metabolism , Animals , Basement Membrane , Biomimetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Pericytes/pathology , Tumor Microenvironment
2.
Cells ; 9(3)2020 03 04.
Article in English | MEDLINE | ID: mdl-32143367

ABSTRACT

It was previously shown that Bone Morphogenetic Protein (BMP)-9 is constitutively produced and secreted by hepatic stellate cells (HSC). Upon acute liver damage, BMP-9 expression is transiently down-regulated and blocking BMP-9 under conditions of chronic damage ameliorated liver fibrogenesis in C57BL/6 mice. Thereby, BMP-9 acted as a pro-fibrogenic cytokine in the liver but without directly activating isolated HSC in vitro. Lipopolysaccharide (LPS), an endotoxin derived from the membrane of Gram-negative bacteria in the gut, is known to be essential in the pathogenesis of diverse kinds of liver diseases. The aim of the present project was therefore to investigate how high levels of BMP-9 in the context of LPS signalling might result in enhanced liver damage. For this purpose, we stimulated human liver sinusoidal endothelial cells (LSEC) with LPS and incubated primary human liver myofibroblasts (MF) with the conditioned medium of these cells. We found that LPS led to the secretion of factors from LSEC that upregulate BMP-9 expression in MF. At least one of these BMP-9 enhancing factors was defined to be IL-6. High BMP-9 in turn, especially in combination with LPS stimulation, induced the expression of certain capillarization markers in LSEC and enhanced the LPS-mediated induction of pro-inflammatory cytokines in primary human macrophages. In LSEC, pre-treatment with BMP-9 reduced the LPS-mediated activation of the NfkB pathway, whereas in macrophages, LPS partially inhibited the BMP-9/Smad-1 signaling cascade. In vivo, in mice, BMP-9 led to the enhanced presence of F4/80-positive cells in the liver and it modulated the LPS-mediated regulation of inflammatory mediators. In summary, our data point to BMP-9 being a complex and highly dynamic modulator of hepatic responses to LPS: Initial effects of LPS on LSEC led to the upregulation of BMP-9 in MF but sustained high levels of BMP-9 in turn promote pro-inflammatory reactions of macrophages. Thereby, the spatial and timely fine-tuned presence (or absence) of BMP-9 is needed for efficient wound-healing responses in the liver.


Subject(s)
Endothelial Cells/drug effects , Growth Differentiation Factor 2/pharmacology , Lipopolysaccharides/pharmacology , Liver/drug effects , Animals , Cells, Cultured , Cytokines/metabolism , Down-Regulation/drug effects , Endothelial Cells/metabolism , Growth Differentiation Factor 2/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Inflammation/drug therapy , Inflammation/pathology , Inflammation Mediators/metabolism , Mice, Inbred C57BL
3.
EMBO Mol Med ; 9(6): 741-749, 2017 06.
Article in English | MEDLINE | ID: mdl-28373218

ABSTRACT

Hepatocellular carcinoma (HCC) is among the most common and deadliest cancers worldwide. A major contributor to HCC progression is the cross talk between tumor cells and the surrounding stroma including activated hepatic stellate cells (HSC). Activation of HSC during liver damage leads to upregulation of the orphan receptor endosialin (CD248), which contributes to regulating the balance of liver regeneration and fibrosis. Based on the established role of endosialin in regulating HSC/hepatocyte cross talk, we hypothesized that HSC-expressed endosialin might similarly affect cell proliferation during hepatocarcinogenesis. Indeed, the histological analysis of human HCC samples revealed an inverse correlation between tumor cell proliferation and stromal endosialin expression. Correspondingly, global genetic inactivation of endosialin resulted in accelerated tumor growth in an inducible mouse HCC model. A candidate-based screen of tumor lysates and differential protein arrays of cultured HSC identified several established hepatotropic cytokines, including IGF2, RBP4, DKK1, and CCL5 as being negatively regulated by endosialin. Taken together, the experiments identify endosialin-expressing HSC as a negative regulator of HCC progression.


Subject(s)
Antigens, CD/analysis , Antigens, Neoplasm/analysis , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Hepatic Stellate Cells/physiology , Hepatocytes/physiology , Liver Neoplasms/pathology , Animals , Disease Models, Animal , Humans , Mice , Neoplasm Proteins/deficiency
4.
Gut ; 66(5): 939-954, 2017 05.
Article in English | MEDLINE | ID: mdl-28336518

ABSTRACT

OBJECTIVE: Bone morphogenetic protein (BMP)-9, a member of the transforming growth factor-ß family of cytokines, is constitutively produced in the liver. Systemic levels act on many organs and tissues including bone and endothelium, but little is known about its hepatic functions in health and disease. DESIGN: Levels of BMP-9 and its receptors were analysed in primary liver cells. Direct effects of BMP-9 on hepatic stellate cells (HSCs) and hepatocytes were studied in vitro, and the role of BMP-9 was examined in acute and chronic liver injury models in mice. RESULTS: Quiescent and activated HSCs were identified as major BMP-9 producing liver cell type. BMP-9 stimulation of cultured hepatocytes inhibited proliferation, epithelial to mesenchymal transition and preserved expression of important metabolic enzymes such as cytochrome P450. Acute liver injury caused by partial hepatectomy or single injections of carbon tetrachloride (CCl4) or lipopolysaccharide (LPS) into mice resulted in transient downregulation of hepatic BMP-9 mRNA expression. Correspondingly, LPS stimulation led to downregulation of BMP-9 expression in cultured HSCs. Application of BMP-9 after partial hepatectomy significantly enhanced liver damage and disturbed the proliferative response. Chronic liver damage in BMP-9-deficient mice or in mice adenovirally overexpressing the selective BMP-9 antagonist activin-like kinase 1-Fc resulted in reduced deposition of collagen and subsequent fibrosis. CONCLUSIONS: Constitutive expression of low levels of BMP-9 stabilises hepatocyte function in the healthy liver. Upon HSC activation, endogenous BMP-9 levels increase in vitro and in vivo and high levels of BMP-9 cause enhanced damage upon acute or chronic injury.


Subject(s)
Acute Lung Injury/physiopathology , Growth Differentiation Factor 2/metabolism , Growth Differentiation Factor 2/pharmacology , Hepatic Stellate Cells/metabolism , Hepatocytes/physiology , Liver Cirrhosis/metabolism , Liver Regeneration/drug effects , Acute Lung Injury/genetics , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Growth Differentiation Factor 2/antagonists & inhibitors , Growth Differentiation Factor 2/genetics , Hepatectomy , Hepatocytes/drug effects , Hepatocytes/enzymology , Lipopolysaccharides/pharmacology , Liver Cirrhosis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Arterioscler Thromb Vasc Biol ; 37(3): 495-505, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28126825

ABSTRACT

OBJECTIVE: Vascular smooth muscle cells (VSMC) play a key role in the pathogenesis of atherosclerosis, the globally leading cause of death. The transmembrane orphan receptor endosialin (CD248) has been characterized as an activation marker of cells of the mesenchymal lineage including tumor-associated pericytes, stromal myofibroblasts, and activated VSMC. We, therefore, hypothesized that VSMC-expressed endosialin may display functional involvement in the pathogenesis of atherosclerosis. APPROACH AND RESULTS: Expression of endosialin was upregulated during atherosclerosis in apolipoprotein E (ApoE)-null mice and human atherosclerotic samples analyzed by quantitative real-time polymerase chain reaction and immunohistochemistry. Atherosclerosis, assessed by Oil Red O staining of the descending aorta, was significantly reduced in ApoE/endosialin-deficient mice on Western-type diet. Marker analysis of VSMC in lesions induced by shear stress-modifying cast implantation around the right carotid artery identified a more pronounced contractile VSMC phenotype in the absence of endosialin. Moreover, in addition to contributing to neointima formation, endosialin also potentially regulated the proinflammatory phenotype of VSMC as evidenced in surrogate cornea pocket assay experiments in vivo and corresponding flow cytometry and ELISA analyses in vitro. CONCLUSIONS: The experiments identify endosialin as a potential regulator of phenotypic remodeling of VSMC contributing to atherosclerosis. The association of endosialin with atherosclerosis and its absent expression in nonatherosclerotic samples warrant further consideration of endosialin as a therapeutic target and biomarker.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neoplasm Proteins/metabolism , Vascular Remodeling , Aged , Aged, 80 and over , Animals , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Case-Control Studies , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , Neointima , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phenotype , Signal Transduction , Vasoconstriction
6.
Cancer Res ; 76(18): 5313-25, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27635044

ABSTRACT

Metastasis is a multistep process that is critically dependent on the interaction of metastasizing tumor cells with cells in the local microenvironment. Within this tumor stroma, vessel-associated pericytes and myofibroblasts share a number of traits, including the upregulated expression of the transmembrane receptor endosialin (CD248). Comparative experiments in wild-type and endosialin-deficient mice revealed that stromal endosialin does not affect primary tumor growth but strongly promotes spontaneous metastasis. Mechanistically, endosialin-expressing pericytes in the primary tumor facilitate distant site metastasis by promoting tumor cell intravasation in a cell contact-dependent manner, resulting in elevated numbers of circulating tumor cells. Corresponding to these preclinical experiments, in independent cohorts of primary human breast cancers, upregulated endosialin expression significantly correlates with increased metastasis and poorer patient survival. Together, the data demonstrate a critical role for endosialin-expressing primary tumor pericytes in mediating metastatic dissemination and identify endosialin as a promising therapeutic target in breast cancer. Cancer Res; 76(18); 5313-25. ©2016 AACR.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Breast Neoplasms/pathology , Pericytes/metabolism , Animals , Breast Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Neoplasm Invasiveness/pathology , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Polymerase Chain Reaction
7.
EMBO Mol Med ; 7(3): 332-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25680861

ABSTRACT

Liver fibrosis is a reversible wound-healing response to injury reflecting the critical balance between liver repair and scar formation. Chronic damage leads to progressive substitution of liver parenchyma by scar tissue and ultimately results in liver cirrhosis. Stromal cells (hepatic stellate cells [HSC] and endothelial cells) have been proposed to control the balance between liver fibrosis and regeneration. Here, we show that endosialin, a C-type lectin, expressed in the liver exclusively by HSC and portal fibroblasts, is upregulated in liver fibrosis in mouse and man. Chronic chemically induced liver damage resulted in reduced fibrosis and enhanced hepatocyte proliferation in endosialin-deficient (EN(KO)) mice. Correspondingly, acute-liver-damage-induced hepatocyte proliferation (partial hepatectomy) was increased in EN(KO) mice. A candidate-based screen of known regulators of hepatocyte proliferation identified insulin-like growth factor 2 (IGF2) as selectively endosialin-dependent hepatocyte mitogen. Collectively, the study establishes a critical role of HSC in the reciprocal regulation of fibrogenesis vs. hepatocyte proliferation and identifies endosialin as a therapeutic target in non-neoplastic settings.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Cell Proliferation , Hepatic Stellate Cells/metabolism , Hepatocytes/cytology , Liver Cirrhosis/pathology , Animals , Humans , Liver Cirrhosis/chemically induced , Liver Regeneration , Mice , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism
8.
J Biol Chem ; 288(4): 2210-22, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23195957

ABSTRACT

Glioblastomas are very difficult tumors to treat because they are highly invasive and disseminate within the normal brain, resulting in newly growing tumors. We have identified netrin-1 as a molecule that promotes glioblastoma invasiveness. As evidence, netrin-1 stimulates glioblastoma cell invasion directly through Matrigel-coated transwells, promotes tumor cell sprouting and enhances metastasis to lymph nodes in vivo. Furthermore, netrin-1 regulates angiogenesis as shown in specific angiogenesis assays such as enhanced capillary endothelial cells (EC) sprouting and by increased EC infiltration into Matrigel plugs in vivo, as does VEGF-A. This netrin-1 signaling pathway in glioblastoma cells includes activation of RhoA and cyclic AMP response element-binding protein (CREB). A novel finding is that netrin-1-induced glioblastoma invasiveness and angiogenesis are mediated by activated cathepsin B (CatB), a cysteine protease that translocates to the cell surface as an active enzyme and co-localizes with cell surface annexin A2 (ANXA2). The specific CatB inhibitor CA-074Me inhibits netrin-1-induced cell invasion, sprouting, and Matrigel plug angiogenesis. Silencing of CREB suppresses netrin-1-induced glioblastoma cell invasion, sprouting, and CatB expression. It is concluded that netrin-1 plays an important dual role in glioblastoma progression by promoting both glioblastoma cell invasiveness and angiogenesis in a RhoA-, CREB-, and CatB-dependent manner. Targeting netrin-1 pathways may be a promising strategy for brain cancer therapy.


Subject(s)
Cathepsin B/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Neovascularization, Pathologic , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Annexin A2/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Collagen/chemistry , Drug Combinations , Female , Gene Silencing , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Laminin/chemistry , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Biological , Neoplasm Invasiveness , Netrin-1 , Protein Binding , Proteoglycans/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...