Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Publication year range
1.
Cell Mol Life Sci ; 79(6): 293, 2022 May 13.
Article in English | MEDLINE | ID: mdl-35562519

ABSTRACT

Atypical chemokine receptor 3 (ACKR3, formerly CXC chemokine receptor 7) is a G protein-coupled receptor that recruits ß-arrestins, but is devoid of functional G protein signaling after receptor stimulation. In preclinical models of liver and lung fibrosis, ACKR3 was previously shown to be upregulated after acute injury in liver sinusoidal and pulmonary capillary endothelial cells, respectively. This upregulation was linked with a pro-regenerative and anti-fibrotic role for ACKR3. A recently described ACKR3-targeting small molecule agonist protected mice from isoproterenol-induced cardiac fibrosis. Here, we aimed to evaluate its protective role in preclinical models of liver and lung fibrosis. After confirming its in vitro pharmacological activity (i.e., ACKR3-mediated ß-arrestin recruitment and receptor binding), in vivo administration of this ACKR3 agonist led to increased mouse CXCL12 plasma levels, indicating in vivo interaction of the agonist with ACKR3. Whereas twice daily in vivo administration of the ACKR3 agonist lacked inhibitory effect on bleomycin-induced lung fibrosis, it had a modest, but significant anti-fibrotic effect in the carbon tetrachloride (CCl4)-induced liver fibrosis model. In the latter model, ACKR3 stimulation affected the expression of several fibrosis-related genes and led to reduced collagen content as determined by picro-sirius red staining and hydroxyproline quantification. These data confirm that ACKR3 agonism, at least to some extent, attenuates fibrosis, although this effect is rather modest and heterogeneous across various tissue types. Stimulating ACKR3 alone without intervening in other signaling pathways involved in the multicellular crosstalk leading to fibrosis will, therefore, most likely not be sufficient to deliver a satisfactory clinical outcome.


Subject(s)
Pulmonary Fibrosis , Receptors, CXCR , Animals , Mice , beta-Arrestins/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL12/pharmacology , Endothelial Cells/metabolism , Liver/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Receptors, CXCR/chemistry , Receptors, CXCR/genetics , Receptors, CXCR/metabolism
2.
Molecules ; 27(3)2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35164317

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, has led to a pandemic, that continues to be a huge public health burden. Despite the availability of vaccines, there is still a need for small-molecule antiviral drugs. In an effort to identify novel and drug-like hit matter that can be used for subsequent hit-to-lead optimization campaigns, we conducted a high-throughput screening of a 160 K compound library against SARS-CoV-2, yielding a 1-heteroaryl-2-alkoxyphenyl analog as a promising hit. Antiviral profiling revealed this compound was active against various beta-coronaviruses and preliminary mode-of-action experiments demonstrated that it interfered with viral entry. A systematic structure-activity relationship (SAR) study demonstrated that a 3- or 4-pyridyl moiety on the oxadiazole moiety is optimal, whereas the oxadiazole can be replaced by various other heteroaromatic cycles. In addition, the alkoxy group tolerates some structural diversity.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Heterocyclic Compounds/pharmacology , SARS-CoV-2/drug effects , Virus Replication/drug effects , Animals , Chlorocebus aethiops , High-Throughput Screening Assays , Microbial Sensitivity Tests , Structure-Activity Relationship , Vero Cells
3.
Gastroenterology ; 153(4): 1054-1067, 2017 10.
Article in English | MEDLINE | ID: mdl-28642198

ABSTRACT

BACKGROUND: Intestinal fibrosis resulting in (sub)obstruction is a common complication of Crohn's disease (CD). Rho kinases (ROCKs) play multiple roles in TGFß-induced myofibroblast activation that could be therapeutic targets. Because systemic ROCK inhibition causes cardiovascular side effects, we evaluated the effects of a locally acting ROCK inhibitor (AMA0825) on intestinal fibrosis. METHODS: Fibrosis was assessed in mouse models using dextran sulfate sodium (DSS) and adoptive T-cell transfer. The in vitro and ex vivo effects of AMA0825 were studied in different cell types and in CD biopsy cultures. RESULTS: ROCK is expressed in fibroblastic, epithelial, endothelial, and muscle cells of the human intestinal tract and is activated in inflamed and fibrotic tissue. Prophylactic treatment with AMA0825 inhibited myofibroblast accumulation, expression of pro-fibrotic factors, and accumulation of fibrotic tissue without affecting clinical disease activity and histologic inflammation in 2 models of fibrosis. ROCK inhibition reversed established fibrosis in a chronic DSS model and impeded ex vivo pro-fibrotic protein secretion from stenotic CD biopsies. AMA0825 reduced TGFß1-induced activation of myocardin-related transcription factor (MRTF) and p38 mitogen-activated protein kinase (MAPK), down-regulating matrix metalloproteinases, collagen, and IL6 secretion from fibroblasts. In these cells, ROCK inhibition potentiated autophagy, which was required for the observed reduction in collagen and IL6 production. AMA0825 did not affect pro-inflammatory cytokine secretion from other ROCK-positive cell types, corroborating the selective in vivo effect on fibrosis. CONCLUSIONS: Local ROCK inhibition prevents and reverses intestinal fibrosis by diminishing MRTF and p38 MAPK activation and increasing autophagy in fibroblasts. Overall, our results show that local ROCK inhibition is promising for counteracting fibrosis as an add-on therapy for CD.


Subject(s)
Ileum/drug effects , Inflammatory Bowel Diseases/prevention & control , Intestinal Obstruction/prevention & control , Myofibroblasts/drug effects , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Adoptive Transfer , Animals , Autophagy/drug effects , Case-Control Studies , Collagen/metabolism , Dextran Sulfate , Disease Models, Animal , Enzyme Activation , Fibrosis , Humans , Ileum/enzymology , Ileum/immunology , Ileum/pathology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/pathology , Interleukin-6/metabolism , Intestinal Obstruction/chemically induced , Intestinal Obstruction/enzymology , Intestinal Obstruction/pathology , Male , Matrix Metalloproteinases/metabolism , Mice, Inbred C57BL , Myofibroblasts/enzymology , Myofibroblasts/immunology , Myofibroblasts/pathology , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Time Factors , Tissue Culture Techniques , p38 Mitogen-Activated Protein Kinases/metabolism , rho-Associated Kinases/metabolism
4.
Bioorg Med Chem Lett ; 25(18): 4005-10, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26233434

ABSTRACT

Inhibitors of LIM kinases are considered of interest for several indications, including elevated intraocular pressure (IOP), cancer, or infection by HIV-1. LX-7101 (Lexicon Pharmaceuticals) was advanced to Phase-I clinical trials as an IOP-lowering agent for treatment of glaucoma. We here discuss the design, synthesis and evaluation of LIMK inhibitors based on a pyrrolopyrimidine scaffold, which represent close analogs of LX-7101. Exploration of structure-activity relationships revealed that many of such compounds, including LX-7101, cause potent inhibition of LIMK1 and LIMK2, and also ROCK2 and PKA. Molecular variations around the various structural elements of LX-7101 were attempted. Substitution on position 6 of the pyrrolopyrimidine scaffold led to the identification of LX-7101 analogs displaying good selectivity versus ROCK, PKA and Akt.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Drug Design , Lim Kinases/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , HeLa Cells , Humans , Lim Kinases/metabolism , Models, Molecular , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , rho-Associated Kinases/metabolism
5.
Bioorg Med Chem Lett ; 24(18): 4594-4597, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25149511

ABSTRACT

PDE4 inhibitors are of high interest for treatment of a wide range of inflammatory or autoimmune diseases. Their potential however has not yet been realized due to target-associated side effects, resulting in a low therapeutic window. We herein report the design, synthesis and evaluation of novel PDE4 inhibitors containing a γ-lactone structure. Such molecules are designed to undergo metabolic inactivation when entering circulation, thereby limiting systemic exposure and reducing the risk for side effects. The resulting inhibitors were highly active on both PDE4B1 and PDE4D2 and underwent rapid degradation in human plasma by paraoxonase 1. In contrast, their metabolites displayed markedly reduced permeability and/or on-target activity.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Aminopyridines/blood , Aminopyridines/chemistry , Benzamides/blood , Benzamides/chemistry , Cyclopropanes/blood , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Dose-Response Relationship, Drug , Humans , Hydrolysis , Molecular Structure , Phosphodiesterase 4 Inhibitors/blood , Phosphodiesterase 4 Inhibitors/chemistry , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 23(23): 6442-6, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24119557

ABSTRACT

Clinical development of ROCK inhibitors has so far been limited by systemic or local ROCK-associated side effects. A soft drug approach, which involves predictable metabolic inactivation of an active compound to a nontoxic metabolite, could represent an attractive way to obtain ROCK inhibitors with improved tolerability. We herein report the design and synthesis of a new series of soft ROCK inhibitors structurally related to the ROCK inhibitor Y-27632. These inhibitors contain carboxylic ester moieties which allow inactivation by esterases. While the parent esters display strong activity in enzymatic (ROCK2) and cellular (MLC phosphorylation) assays, their corresponding carboxylic acid metabolites have negligible functional activity. Compound 32 combined strong efficacy (ROCK2 IC50=2.5 nM) with rapid inactivation in plasma (t1/2 <5'). Compound 32 also demonstrated in vivo efficacy when evaluated as an IOP-lowering agent in ocular normotensive New-Zealand White rabbits, without ocular side effects.


Subject(s)
Benzoates/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Animals , Benzoates/chemistry , Models, Molecular , Phosphorylation , Pyridines/chemistry , Pyridines/pharmacology , Rabbits
7.
J Immunol ; 184(6): 3164-73, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20154208

ABSTRACT

Vascular adhesion protein-1 (VAP-1) is an endothelial, cell surface-expressed oxidase involved in leukocyte traffic. The adhesive function of VAP-1 can be blocked by anti-VAP-1 Abs and small-molecule inhibitors. However, the effects of VAP-1 blockade on antitumor immunity and tumor progression are unknown. In this paper, we used anti-VAP-1 mAbs and small-molecule inhibitors of VAP-1 in B16 melanoma and EL-4 lymphoma tumor models in C57BL/6 mice. Leukocyte accumulation into tumors and neoangiogenesis were evaluated by immunohistochemistry, flow cytometry, and intravital videomicroscopy. We found that both anti-VAP-1 Abs and VAP-1 inhibitors reduced the number of leukocytes in the tumors, but they targeted partially different leukocyte subpopulations. Anti-VAP-1 Abs selectively inhibited infiltration of CD8-positive lymphocytes into tumors and had no effect on accumulation of myeloid cells into tumors. In contrast, the VAP-1 inhibitors significantly reduced only the number of proangiogenic Gr-1(+)CD11b(+) myeloid cells in melanomas and lymphomas. Blocking of VAP-1 by either means left tumor homing of regulatory T cells and type 2 immune-suppressing monocytes/macrophages intact. Notably, VAP-1 inhibitors, but not anti-VAP-1 Abs, retarded the growth of melanomas and lymphomas and reduced tumor neoangiogenesis. The VAP-1 inhibitors also reduced the binding of Gr-1(+) myeloid cells to the tumor vasculature. We conclude that tumors use the catalytic activity of VAP-1 to recruit myeloid cells into tumors and to support tumor progression. Small-molecule VAP-1 inhibitors therefore might be a potential new tool for immunotherapy of tumors.


Subject(s)
Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Amine Oxidase (Copper-Containing)/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/immunology , Cell Migration Inhibition/immunology , Growth Inhibitors/therapeutic use , Lymphoma, T-Cell/immunology , Melanoma, Experimental/immunology , Myeloid Cells/immunology , Myeloid Cells/pathology , Adjuvants, Immunologic/therapeutic use , Allylamine/analogs & derivatives , Allylamine/therapeutic use , Amine Oxidase (Copper-Containing)/biosynthesis , Animals , Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Cell Adhesion Molecules/biosynthesis , Cell Line, Tumor , Cell Migration Inhibition/drug effects , Enzyme Inhibitors/therapeutic use , Female , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/therapy , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Semicarbazides/therapeutic use
8.
Mol Cancer ; 9: 231, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20813052

ABSTRACT

BACKGROUND: Disorganized angiogenesis is associated with several pathologies, including cancer. The identification of new genes that control tumor neovascularization can provide novel insights for future anti-cancer therapies. Sprouty1 (SPRY1), an inhibitor of the MAPK pathway, might be one of these new genes. We identified SPRY1 by comparing the transcriptomes of untreated endothelial cells with those of endothelial cells treated by the angiostatic agent 16 K prolactin (16 K hPRL). In the present study, we aimed to explore the potential function of SPRY1 in angiogenesis. RESULTS: We confirmed 16 K hPRL induced up-regulation of SPRY1 in primary endothelial cells. In addition, we demonstrated the positive SPRY1 regulation in a chimeric mouse model of human colon carcinoma in which 16 K hPRL treatment was shown to delay tumor growth. Expression profiling by qRT-PCR with species-specific primers revealed that induction of SPRY1 expression by 16 K hPRL occurs only in the (murine) endothelial compartment and not in the (human) tumor compartment. The regulation of SPRY1 expression was NF-κB dependent. Partial SPRY1 knockdown by RNA interference protected endothelial cells from apoptosis as well as increased endothelial cell proliferation, migration, capillary network formation, and adhesion to extracellular matrix proteins. SPRY1 knockdown was also shown to affect the expression of cyclinD1 and p21 both involved in cell-cycle regulation. These findings are discussed in relation to the role of SPRY1 as an inhibitor of ERK/MAPK signaling and to a possible explanation of its effect on cell proliferation. CONCLUSIONS: Taken together, these results suggest that SPRY1 is an endogenous angiogenesis inhibitor.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Neovascularization, Pathologic/metabolism , Peptide Fragments/metabolism , Phosphoproteins/metabolism , Prolactin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blotting, Western , Cattle , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Female , HCT116 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Peptide Fragments/pharmacology , Phosphoproteins/genetics , Prolactin/pharmacology , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
9.
Blood ; 112(13): 4940-7, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18515660

ABSTRACT

Interleukin-21 (IL-21) is a recently described immunoregulatory cytokine. It has been identified as a very potent immunotherapeutic agent in several cancer types in animal models, and clinical studies are ongoing. IL-21 belongs to the type I cytokine family of which other members, ie, IL-2, IL-15, and IL-4, have been shown to exert activities on vascular endothelial cells (ECs). We hypothesized that IL-21, in addition to inducing the antitumor immune response, also inhibits tumor angiogenesis. In vitro experiments showed a decrease of proliferation and sprouting of activated ECs after IL-21 treatment. We found that the IL-21 receptor is expressed on vascular ECs. Furthermore, in vivo studies in the chorioallantoic membrane of the chick embryo and in mouse tumors demonstrated that IL-21 treatment disturbs vessel architecture and negatively affects vessel outgrowth. Our results also confirm the earlier suggested angiostatic potential of IL-2 in vitro and in vivo. The angiostatic effect of IL-21 is confirmed by the decrease in expression of angiogenesis-related genes. Interestingly, IL-21 treatment of ECs leads to a decrease of Stat3 phosphorylation. Our research shows that IL-21 is a very powerful antitumor compound that combines the induction of an effective antitumor immune response with inhibition of tumor angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Interleukins/pharmacology , Animals , Antineoplastic Agents , Aorta/cytology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Humans , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Receptors, Interleukin-21 , STAT3 Transcription Factor/metabolism
10.
Angiogenesis ; 12(1): 17-24, 2009.
Article in English | MEDLINE | ID: mdl-19067197

ABSTRACT

Molecular imaging of angiogenesis is urgently needed for diagnostic purposes such as early detection, monitoring of (angiostatic) therapy and individualized therapy. Multimodality molecular imaging is a promising and refined technique to study tumor angiogenesis, which has so far been largely unexplored due to the lack of suitable multimodal contrast agents. Here, we report on the application of a novel alphavbeta3-specific quantum dot-based nanoparticle, which has been optimized for both optical and magnetic resonance detection of tumor angiogenesis. Upon intravenous injection of RGD-pQDs in tumor-bearing mice, intravital microscopy allowed the detection of angiogenically activated endothelium at cellular resolution with a small scanning window and limited penetration depth, while magnetic resonance imaging was used to visualize angiogenesis at anatomical resolution throughout the entire tumor. Fluorescence imaging allowed whole-body investigation of angiogenic activity. Using these quantum dots and the aforementioned imaging modalities, the angiogenic tumor vasculature was readily detected with the highest angiogenic activity occurring in the periphery of the tumor. This nanoparticle may be employed for multimodality imaging of a variety of diseases that are accompanied by activation of endothelial cells. Furthermore, the current technology might be developed for molecular imaging of other pathophysiological processes.


Subject(s)
Diagnostic Imaging , Integrin alphaVbeta3/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Quantum Dots , Animals , Chickens , Chorioallantoic Membrane/blood supply , Magnetic Resonance Imaging , Mice , Microscopy, Fluorescence , Oligopeptides/metabolism
11.
Biochim Biophys Acta ; 1776(2): 160-74, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17888580

ABSTRACT

In spite of a gradual improvement of its therapy, cancer is still a deadly disease for millions of patients. Immunotherapy is one of promising treatment strategies, but several mechanisms counteract the development of a proper anti-tumor immune response and the formation of an effective inflammatory infiltrate. One of the difficult hurdles is the hampered recruitment of leukocytes from the blood into the tumor site. It has been demonstrated that tumor cells evolved mechanisms to escape immunity, based on down regulation of endothelial adhesion molecules. This paper reviews the endothelial cell adhesion molecules that mediate leukocyte recruitment and the regulation of them during tumor development. In addition, an overview will be given of the translational development and clinical application of the specific composition of adhesion molecules on tumor endothelium, in diagnosis and therapy.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Endothelium, Vascular/metabolism , Leukocytes/immunology , Neoplasms/blood supply , Neoplasms/immunology , Biomarkers/analysis , Cell Adhesion , Cell Movement , Drug Delivery Systems , Endothelium, Vascular/cytology , Humans , Leukocyte Rolling , Lymphocytes, Tumor-Infiltrating , Models, Biological , Tumor Escape
12.
Biochem Biophys Res Commun ; 368(2): 364-7, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18237547

ABSTRACT

We report the generation of a transgenic Tie2-GFP athymic nude mouse, carrying green fluorescent blood vessels throughout the body. This transgenic mouse is a tool for studies in vascular biology, and is especially of interest for imaging of tumor angiogenesis and the study of anti-angiogenesis strategies in (human) xenografts. Intravital microscopy identified the presence of blood conducting structures that are not lined by endothelial cells. Dedifferentiation of aggressive tumor cells can lead to acquisition of endothelial characteristics. This process of tumor cell plasticity, also referred to as vasculogenic mimicry, has been suggested to contribute to the circulatory system in a tumor. In plastic EW7 Ewing sarcoma tumors in these Tie2-GFP mice, we observed blood flow in both regular blood vessels and non-fluorescent tumor cell-lined channels, visualizing in vivo hemodynamics in vasculogenic channels. These results demonstrate that the transgenic Tie2-GFP athymic mouse model is a valuable tool for vascular biology research.


Subject(s)
Disease Models, Animal , Microscopy, Fluorescence/methods , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptor, TIE-2/metabolism , Animals , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Molecular Probe Techniques , Neoplasms/blood supply , Receptor, TIE-2/genetics
13.
Bioconjug Chem ; 19(12): 2471-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19035793

ABSTRACT

Silica particles as a nanoparticulate carrier material for contrast agents have received considerable attention the past few years, since the material holds great promise for biomedical applications. A key feature for successful application of this material in vivo is biocompatibility, which may be significantly improved by appropriate surface modification. In this study, we report a novel strategy to coat silica particles with a dense monolayer of paramagnetic and PEGylated lipids. The silica nanoparticles carry a quantum dot in their center and are made target-specific by the conjugation of multiple alphavbeta3-integrin-specific RGD-peptides. We demonstrate their specific uptake by endothelial cells in vitro using fluorescence microscopy, quantitative fluorescence imaging, and magnetic resonance imaging. The lipid-coated silica particles introduced here represent a new platform for nanoparticulate multimodality contrast agents.


Subject(s)
Contrast Media/chemistry , Fluorescence , Lipids/chemistry , Magnetics , Quantum Dots , Silicon Dioxide/chemistry , Animals , Cattle , Cell Line , Gadolinium DTPA/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Imaging , Polyethylene Glycols/chemistry , Surface Properties
14.
Mol Endocrinol ; 21(6): 1422-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17405903

ABSTRACT

The 16-kDa N-terminal fragment of human prolactin (16K hPRL) is a potent angiostatic factor that inhibits tumor growth in mouse models. Using microarray experiments, we have dissected how the endothelial-cell genome responds to 16K hPRL treatment. We found 216 genes that show regulation by 16K hPRL, of which a large proportion turned out to be associated with the process of immunity. 16K hPRL induces expression of various chemokines and endothelial adhesion molecules. These expressions, under the control of nuclear factor-kappaB, result in an enhanced leukocyte-endothelial cell interaction. Furthermore, analysis of B16-F10 tumor tissues reveals a higher expression of adhesion molecules (intercellular adhesion molecule 1, vascular cell adhesion molecule 1, or E-selectin) in endothelial cells and a significantly higher number of infiltrated leukocytes within the tumor treated with 16K hPRL compared with the untreated ones. In conclusion, this study describes a new antitumor mechanism of 16K hPRL. Because cellular immunity against tumor cells is a crucial step in therapy, the discovery that treatment with 16K hPRL overcomes tumor-induced anergy may become important for therapeutic perspectives.


Subject(s)
Angiostatic Proteins/pharmacology , Clonal Anergy/drug effects , Endothelium, Vascular/drug effects , Gene Expression/drug effects , NF-kappa B/physiology , Peptide Fragments/pharmacology , Prolactin/pharmacology , Animals , Cell Adhesion , Clonal Anergy/genetics , Endothelium, Vascular/immunology , Humans , Leukocytes/drug effects , Leukocytes/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , NF-kappa B/genetics , Oligonucleotide Array Sequence Analysis , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology
15.
Cancer Res ; 66(22): 10770-7, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108113

ABSTRACT

Tumors can escape from immunity by repressing leukocyte adhesion molecule expression on tumor endothelial cells and by rendering endothelial cells unresponsive to inflammatory activation. This endothelial cell anergy is induced by angiogenic growth factors and results in reduced leukocyte-vessel wall interactions, thereby attenuating infiltration of leukocytes into the tumor. This report describes a novel mechanism of endothelial cell anergy regulation. We recently reported that DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors have angiostatic activity. Here, we studied whether epigenetic mechanisms regulate this angiogenesis-mediated escape from immunity. We found that DNMT inhibitors 5-aza-2'-deoxycytidine and zebularine, as well as HDAC inhibitor trichostatin A, reexpressed intercellular adhesion molecule-1 (ICAM-1) on tumor-conditioned endothelial cells in vitro, resulting in restored leukocyte-endothelial cell adhesion. In addition, treatment with DNMT or HDAC inhibitors in vivo also restored ICAM-1 expression on tumor endothelial cells from two different mouse tumor models. Furthermore, leukocyte-vessel wall interactions in mouse tumors were increased by these compounds, as measured by intravital microscopy, resulting in enhanced leukocyte infiltration. We show that ICAM-1 down-regulation in tumor endothelial cells is associated with ICAM-1 promoter histone H3 deacetylation and loss of histone H3 Lys(4) methylation but not with DNA hypermethylation. In conclusion, our data show that ICAM-1 is epigenetically silenced in tumor endothelial cells by promoter histone modifications, which can be overcome by DNMT and HDAC inhibitors, suggesting a new molecular mechanism based on which novel therapeutic approaches for cancer can be pursued.


Subject(s)
Endothelial Cells/physiology , Histones/genetics , Intercellular Adhesion Molecule-1/genetics , Acetylation , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Clonal Anergy , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Down-Regulation , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epigenesis, Genetic , Gene Silencing , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Leukocytes/pathology , Melanoma, Experimental/blood , Melanoma, Experimental/pathology , Methylation , Mice , Mice, Inbred C57BL
16.
FASEB J ; 20(6): 621-30, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581970

ABSTRACT

Tumor escape from immunity, as well as the failure of several anti-cancer vaccination and cellular immunotherapy approaches, is suggested to be due to the angiogenesis-mediated suppression of endothelial cell (EC) adhesion molecules involved in leukocyte-vessel wall interactions. We hypothesized that inhibition of angiogenesis would overcome this escape from immunity. We investigated this in vivo by means of intravital microscopy and ex vivo by immunohistochemistry in two mouse tumor models. Angiogenesis inhibitors anginex, endostatin, and angiostatin, and the chemotherapeutic agent paclitaxel were found to significantly stimulate leukocyte-vessel wall interactions by circumvention of EC anergy in vivo, i.e., by the up-regulation of endothelial adhesion molecules in tumor vessels. This was confirmed by in vitro studies of cultured EC at the protein and mRNA levels. The new angiostatic designer peptide anginex was most potent at overcoming EC anergy; the enhanced leukocyte-vessel interactions led to an increase in the numbers of tumor infiltrating leukocytes. While anginex inhibited tumor growth and microvessel density significantly, the amount of infiltrated leukocytes (CD45), as well as the number of CD8+ cytotoxic T lymphocytes, was enhanced markedly. The current results suggest that immunotherapy strategies can be improved by combination with anti-angiogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Endothelium/cytology , Leukocytes/cytology , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Angiostatins/pharmacology , Animals , Cell Line, Tumor , Clonal Anergy , Cyclohexanes , Cyclophosphamide/pharmacology , Down-Regulation , Endostatins/pharmacology , Endothelial Cells , Endothelium/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/metabolism , Mice , O-(Chloroacetylcarbamoyl)fumagillol , Paclitaxel/pharmacology , Peptides , Proteins/pharmacology , Sesquiterpenes/pharmacology , Tumor Necrosis Factor-alpha , Vascular Cell Adhesion Molecule-1/metabolism
17.
Cancer Res ; 65(24): 11520-8, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16357161

ABSTRACT

A striking feature of Ewing sarcoma is the presence of blood lakes lined by tumor cells. The significance of these structures, if any, is unknown. Here, we report that the extent of blood lakes correlates with poor clinical outcomes, whereas variables of angiogenesis do not. We also show that Ewing sarcoma cells form vessel-like tubes in vitro and express genes associated with vasculogenic mimicry. In tumor models, we show that there is blood flow through the blood lakes, suggesting that these structures in Ewing sarcoma contribute to the circulation. Furthermore, we present evidence that reduced oxygen tension may be instrumental in tube formation by plastic tumor cells. The abundant presence of these vasculogenic structures, in contrast to other tumor types, makes Ewing sarcoma the ideal model system to study these phenomena. The results suggest that optimal tumor treatment may require targeting of these structures in combination with prevention of angiogenesis.


Subject(s)
Cell Hypoxia , Microcirculation/pathology , Neovascularization, Pathologic , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/physiopathology , Adolescent , Adult , Animals , Bone Neoplasms/blood supply , Bone Neoplasms/physiopathology , Child , Child, Preschool , Collagen/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , Middle Aged , Phenotype , Tumor Cells, Cultured/transplantation
18.
Curr Eye Res ; 42(2): 260-272, 2017 02.
Article in English | MEDLINE | ID: mdl-27399806

ABSTRACT

PURPOSE: Diabetic retinopathy (DR) is characterized by an early stage of inflammation and vessel leakage, and an advanced vasoproliferative stage. Also, neurodegeneration might play an important role in disease pathogenesis. The aim of this study was to investigate the effect of the Rho kinase (ROCK) inhibitor, AMA0428, on these processes. METHODS: The response to ROCK inhibition by AMA0428 (1 µg) was studied in vivo using the murine model for streptozotocin (STZ)-induced diabetes, focusing on early non-proliferative DR features and the oxygen-induced retinopathy (OIR) model to investigate proliferative DR. Intravitreal (IVT) administration of AMA0428 was compared with murine anti-VEGF-R2 antibody (DC101, 6.2 µg) and placebo (H2O/PEG; 1C8). Outcome was assessed by analyzing leukostasis using fluorescein isothiocyanate coupled concanavalin A (FITC-ConA) and vessel leakage (bovine serum albumin conjugated with fluorescein isothiocyanate; FITC-BSA)/neovascularization and neurodegeneration by immunohistological approaches (hematoxylin and eosin (H&E), terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL), Brn3a). ELISA and Western blotting were employed to unravel the consequences of ROCK inhibition (1 µM AMA0428) on myosin phosphatase target protein (MYPT)-1 phosphorylation, endothelial nitric oxide synthase (eNOS) phosphorylation, and vascular endothelial growth factor (VEGF) levels in retinas of diabetic mice, on NF-κß activity and ICAM-1 expression in endothelial cells (ECs). RESULTS: In vivo, AMA0428 significantly reduced vessel leakage and neovascularization, respectively, in the STZ and OIR model, comparable to DC101 therapy. Additionally, the ROCK inhibitor decreased neurodegeneration in both models and inhibited leukostasis by 30% (p < 0.05) in the STZ model (p < 0.05), while DC101 had no positive effect on the outcome of these latter processes. ROCK activity was upregulated in the diabetic retina and AMA0428 administration resulted in decreased phospho-MYPT-1, enhanced phospho-eNOS, and reduced VEGF levels. In vitro, AMA0428 interfered with NF-κß activity, thereby inhibiting ICAM-1 expression in ECs. CONCLUSIONS: Targeting ROCK with AMA0428 effectively attenuated outcome in an early DR model (STZ) and a late vasoproliferative retinopathy model (OIR). These findings make AMA0428 a promising candidate with an additional anti-inflammatory and neuroprotective benefit for DR patients, as compared with anti-VEGF treatment.


Subject(s)
Diabetic Retinopathy/drug therapy , Enzyme Inhibitors/pharmacology , Retina/pathology , rho-Associated Kinases/antagonists & inhibitors , Animals , Blotting, Western , Cells, Cultured , Diabetes Mellitus, Experimental , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Progression , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Male , Mice , NF-kappa B/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
Prog Brain Res ; 220: 283-97, 2015.
Article in English | MEDLINE | ID: mdl-26497796

ABSTRACT

PURPOSE: First, to elucidate the effect of Rho kinase inhibitor, AMA0526, on Human Tenon Fibroblast (HTF) proliferation and transdifferentiation to myofibroblasts. Second, the effects of ROCK inhibition on the wound healing process and surgical outcome were investigated in a rabbit model of glaucoma filtration surgery. METHODS: After exposure of HTF to AMA0526 (0.1-25 µM), a water-soluble tetrazolium salt-1 assay and caspase 3/7 activity assay were used to assess its effect on cell proliferation and to elucidate any toxic effects, respectively. Immunohistochemistry of α-smooth muscle actin expression was used to investigate fibroblast-to-myofibroblast differentiation induced by transforming growth factor-beta 1 (TGF-ß1) in the presence or absence of the ROCK inhibitor. The effect of topical treatment was studied in a rabbit model of glaucoma filtration surgery. Treatment outcome was studied by performing intraocular pressure (IOP) measurements and clinical investigation of the bleb area and survival. Immunohistological analysis for inflammation (CD45), angiogenesis (CD31), and collagen I was performed at day 8, 14, and 30 after surgery (n=5/time point). Separate control groups treated with vehicle were used as control. RESULTS: In vitro results showed that AMA0526 dose dependently inhibited proliferation of HTF (P<0.05) without the induction of caspase 3/7 activity. Incubation of HTF with the AMA0526 inhibited TGF-ß1 induced fibroblast-to-myofibroblast differentiation. In the rabbit model, topical treatment significantly improved surgical outcome. Compared to vehicle-treated eyes, AMA0526 resulted in increased bleb area (P<0.0001) and prolonged survival (P=0.0025). IOP remained significantly lower throughout the course of the experiment in the AMA0526 group (P<0.0001). Histological evaluation revealed that blebs treated with the ROCK inhibitor were characterized by reduced inflammation, angiogenesis, and collagen deposition at the site of filtration surgery (P<0.05). CONCLUSIONS: AMA0526 had profound effects on HTF proliferation and myofibroblast transition and improved glaucoma filtration surgery outcome by interfering at different levels of the wound healing process. Therefore, these data indicate that ROCK inhibitors may be considered as more physiological agents which specifically target the wound healing process to improve the outcome of glaucoma surgery.


Subject(s)
Enzyme Inhibitors/therapeutic use , Filtering Surgery/adverse effects , Glaucoma/drug therapy , Glaucoma/surgery , Animals , Antigens, CD/metabolism , Caspases/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Postoperative Complications/prevention & control , Rabbits , Statistics, Nonparametric , Time Factors , Vimentin/metabolism
20.
Cornea ; 34(9): 1120-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26075454

ABSTRACT

PURPOSE: To investigate the effect of AMA0526, a specific inhibitor of rho-associated protein kinase (ROCK), on corneal neovascularization (NV) and scarring in different in vitro and in vivo experimental models. METHODS: The effect of AMA0526 on cell viability, proliferation, and migration of human umbilical vein endothelial cells was determined. Its in vivo topical effect on NV was investigated in the corneal micropocket mouse model (bevacizumab as a control). The vessel length, clock hours, and NV area were measured on photographs. The effect of AMA0526 on pathological wound healing was investigated in the alkali burn mouse model (dexamethasone as a control). Corneas were scored for corneal opacity (CO) and NV after burn injury. Immunohistochemistry was performed to study inflammation, blood vessel density, and collagen III deposition after 7 days. RESULTS: ROCK inhibition significantly inhibited vascular endothelial cell proliferation and migration in vitro in a dose-dependent manner. In the micropocket model, NV was significantly reduced by AMA0526 (37% reduction, P < 0.05) comparable to bevacizumab. CO and NV were reduced after AMA0526, compared with the vehicle (P < 0.05 at all time points from day 3) after chemical burn. AMA0526 resulted in decreased inflammatory cell infiltration (26% reduction, P < 0.01), angiogenesis (47% reduction, P < 0.01), and collagen III deposition (27% reduction, P = 0.009) in the alkali burn model. AMA0526 administration showed results similar to those of dexamethasone with an additional antifibrotic effect. CONCLUSIONS: The ROCK inhibitor, AMA0526, efficiently inhibited angiogenesis in vitro, reduced CO and NV, and controlled the complete process of wound healing in vivo. These results warrant further investigation of the therapeutic potential of AMA0526 for corneal NV and scarring.


Subject(s)
Burns, Chemical/prevention & control , Corneal Opacity/prevention & control , Enzyme Inhibitors/pharmacology , Eye Burns/chemically induced , Neovascularization, Pathologic/prevention & control , Protein Kinase Inhibitors/pharmacology , Wound Healing/drug effects , rho-Associated Kinases/antagonists & inhibitors , Animals , Burns, Chemical/enzymology , Burns, Chemical/etiology , Cell Movement/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Cells, Cultured , Collagen Type III/metabolism , Corneal Opacity/chemically induced , Corneal Opacity/enzymology , Dexamethasone/pharmacology , Disease Models, Animal , Drug Combinations , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Eye Burns/enzymology , Glucocorticoids/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/etiology , Sodium Hydroxide
SELECTION OF CITATIONS
SEARCH DETAIL