Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Br J Cancer ; 115(6): 691-702, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27529514

ABSTRACT

BACKGROUND: To assess antivascular effects, and evaluate clinically translatable magnetic resonance imaging (MRI) biomarkers of tumour response in vivo, following treatment with vanucizumab, a bispecific human antibody against angiopoietin-2 (Ang-2) and vascular endothelial growth factor-A (VEGF-A). METHODS: Colo205 colon cancer xenografts were imaged before and 5 days after treatment with a single 10 mg kg(-1) dose of either vanucizumab, bevacizumab (anti-human VEGF-A), LC06 (anti-murine/human Ang-2) or omalizumab (anti-human IgE control). Volumetric response was assessed using T2-weighted MRI, and diffusion-weighted, dynamic contrast-enhanced (DCE) and susceptibility contrast MRI used to quantify tumour water diffusivity (apparent diffusion coefficient (ADC), × 10(6) mm(2) s(-1)), vascular perfusion/permeability (K(trans), min(-1)) and fractional blood volume (fBV, %) respectively. Pathological correlates were sought, and preliminary gene expression profiling performed. RESULTS: Treatment with vanucizumab, bevacizumab or LC06 induced a significant (P<0.01) cytolentic response compared with control. There was no significant change in tumour ADC in any treatment group. Uptake of Gd-DTPA was restricted to the tumour periphery in all post-treatment groups. A significant reduction in tumour K(trans) (P<0.05) and fBV (P<0.01) was determined 5 days after treatment with vanucizumab only. This was associated with a significant (P<0.05) reduction in Hoechst 33342 uptake compared with control. Gene expression profiling identified 20 human genes exclusively regulated by vanucizumab, 6 of which are known to be involved in vasculogenesis and angiogenesis. CONCLUSIONS: Vanucizumab is a promising antitumour and antiangiogenic treatment, whose antivascular activity can be monitored using DCE and susceptibility contrast MRI. Differential gene expression in vanucizumab-treated tumours is regulated by the combined effect of Ang-2 and VEGF-A inhibition.


Subject(s)
Adenocarcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Colonic Neoplasms/drug therapy , Gene Expression Profiling , Magnetic Resonance Imaging/methods , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Adenocarcinoma/blood supply , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Angiogenesis Inhibitors/immunology , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Bevacizumab/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/blood supply , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/pathology , DNA Replication/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoglobulin E/immunology , Mice , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/pathology , Omalizumab/therapeutic use , Tumor Burden , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays
2.
Cancer Invest ; 33(8): 378-86, 2015.
Article in English | MEDLINE | ID: mdl-26115098

ABSTRACT

The blockade of VEGF pathway has been clinically validated as an initial treatment for renal cell carcinoma (RCC). Angiopoietin-2 (Ang-2) has been indicated as a key regulator for angiogenesis escape. The effect of a novel bispecific antibody (A2V CrossMab) against both Ang-2 and VEGF was investigated in comparison with either factor. A2V CrossMab significantly reduced tumor volume, vessel density, and interstitial fluid pressure compared to either monotherapy of anti-VEGF or anti-Ang-2. Host-derived angiogenesis-related genes have been significantly down-regulated in A2V CrossMab group. These data demonstrate that A2V CrossMab has additive anti-tumor effect for the treatment of RCC.


Subject(s)
Angiopoietin-2/immunology , Antibodies, Bispecific/pharmacology , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/immunology , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/metabolism , Animals , Antibodies, Bispecific/immunology , Carcinoma, Renal Cell/genetics , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kidney Neoplasms/genetics , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
3.
Exp Dermatol ; 24(6): 424-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25776770

ABSTRACT

Primary cutaneous large B-cell lymphomas, leg type (PCLBCL/LT) are primary cutaneous B-cell lymphoma (PCBCL) with an intermediate prognosis. Therefore, antracycline-based polychemotherapy combined with rituximab has been recommended as first-line treatment. Yet, despite this regimen, the 5-year survival rate remains 50-66% only. Angiogenesis, the formation of a vascular network, is essential for the pathogenesis of nodal lymphomas. So far, no study has analysed angiogenesis and its key factors in PCLBCL/LT. The present study was aimed at characterizing angiogenesis in PCLBCL/LT to identify the angiogenic molecules as potential therapeutic targets. The intra-tumoral microvessel density (MVD) was assessed by immunohistochemical studies of CD20 and CD31. The MVD was higher in PCLBCL/LT compared with indolent PCBCL. Analyses of open-source microarray data showed correlation between the angiogenic molecule angiopoietin-2 (Ang-2) and pan-endothelial cell markers. ELISA studies determined a shift between Ang-2 and Ang-1 towards Ang-2 in the peripheral blood of PCLBCL/LT patients. Immunofluorescence costainings against the Ang receptor Tie2/angiogenic integrins/CD34 revealed that the vasculature in both aggressive and indolent PCBCL tumors harbours an endothelial cell subpopulation with reduced expression of Tie2. In contrast, the alternative Ang-2 binding partners, angiogenic integrins, are strongly expressed in PCBCL. In line with these findings, downstream targets of Ang-2-integrin signalling, that is phosphorylation of focal adhesion kinase at Tyr397, and sprouting angiogenesis are enhanced in PCLBCL/LT. Our data present Ang-2 as a promising therapeutic target and anti-angiogenic therapy as a new line in treatment of PCLBCL/LT as a hitherto intractable disease.


Subject(s)
Angiopoietin-2/metabolism , Lymphoma, B-Cell/metabolism , Neovascularization, Pathologic/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Angiopoietin-2/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Integrins/metabolism , Lymphoma, B-Cell/genetics , Microvessels/pathology , Phosphorylation , Signal Transduction/genetics , Signal Transduction/physiology , Skin Neoplasms/genetics
4.
J Dtsch Dermatol Ges ; 13(2): 125-36, 2015 Feb.
Article in English, German | MEDLINE | ID: mdl-25631130

ABSTRACT

Despite the development of novel therapies, the therapy of malignant melanoma remains challenging. Various studies have shown the vascular system to be pivotal for metastasis in melanoma. Consequently, the effect of various antiangiogenic therapies has been and is being investigated in preclinical and clinical trials. While most studies focus on inhibition of vascular endothelial growth factor (VEGF) signaling, others are aimed at determining the effect of multikinase inhibitors or the inhibition of angiogenic integrin activity. However, overall survival rates have not significantly improved in clinical trials with antiangiogenic agents. Resistance to anti-VEGF monotherapy has been observed in several studies, especially in malignant melanoma. Angiopoietin-2 (Ang-2) represents a promising candidate molecule for antiangiogenic therapy and the effect of Ang-2 inhibitors is currently being explored in first trials. In melanoma, Ang-2 has been shown to be a marker for metastasis formation and represents an interesting therapeutic target molecule. Future studies are required to analyze the effect of a combined approach, using anti-VEGF and anti-Ang-2, as therapy for malignant melanoma.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Melanoma/blood supply , Melanoma/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Angiopoietin-2/antagonists & inhibitors , Animals , Disease Progression , Drug Resistance, Neoplasm , Drug Therapy, Combination , Humans , Vascular Endothelial Growth Factor A/antagonists & inhibitors
5.
Proc Natl Acad Sci U S A ; 108(27): 11187-92, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21690412

ABSTRACT

We describe a generic approach to assemble correctly two heavy and two light chains, derived from two existing antibodies, to form human bivalent bispecific IgG antibodies without use of artificial linkers. Based on the knobs-into-holes technology that enables heterodimerization of the heavy chains, correct association of the light chains and their cognate heavy chains is achieved by exchange of heavy-chain and light-chain domains within the antigen binding fragment (Fab) of one half of the bispecific antibody. This "crossover" retains the antigen-binding affinity but makes the two arms so different that light-chain mispairing can no longer occur. Applying the three possible "CrossMab" formats, we generated bispecific antibodies against angiopoietin-2 (Ang-2) and vascular endothelial growth factor A (VEGF-A) and show that they can be produced by standard techniques, exhibit stabilities comparable to natural antibodies, and bind both targets simultaneously with unaltered affinity. Because of its superior side-product profile, the CrossMab(CH1-CL) was selected for in vivo profiling and showed potent antiangiogenic and antitumoral activity.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/chemistry , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Angiopoietin-2/immunology , Animals , Antibodies, Bispecific/metabolism , Antibody Affinity , Antibody Specificity , Cell Line , Cell Line, Tumor , Female , Humans , Immunoglobulin G/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Models, Molecular , Neovascularization, Physiologic , Protein Engineering , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Vascular Endothelial Growth Factor A/immunology
6.
Surg Neurol Int ; 14: 246, 2023.
Article in English | MEDLINE | ID: mdl-37560593

ABSTRACT

Background: Cerebral edema (CE) and intracranial hypertension (IHT) are complications of numerous neurological pathologies. However, the study of CE and noninvasive methods to predict IHT remains rudimentary. This study aims to identify in traumatic brain injury (TBI) patients the relationship between the volume of the lateral ventricles and the parameters of the noninvasive intracranial pressure waveform (nICPW). Methods: This is an analytical, descriptive, and cross-sectional study with nonsurgical TBI patients. The monitoring of nICPW was performed with a mechanical strain gauge, and the volumetry of the lateral ventricles was calculated using the free 3D Slicer software, both during the acute phase of the injury. The linear model of fixed and random mixed effects with Gamma was used to calculate the influence of nICPW parameters (P2/P1 and time-to-peak [TTP]) values on volumetry. Results: Considering only the fixed effects of the sample, there was P = 0.727 (95% CI [-0.653; 0.364]) for the relationship between P2/P1 and volumetry and 0.727 (95% CI [-1.657; 1.305]) for TTP and volumetry. Considering the fixed and random effects, there was P = 8.5e-10 (95% CI [-0.759; 0.355]) for the relationship between P2/P1 and volumetry and 8.5e-10 (95% CI [-2.001; 0.274]) for TTP and volumetry. Conclusion: The present study with TBI patients found association between nICPW parameters and the volume of the lateral ventricles in the 1st days after injury.

7.
J Bacteriol ; 194(11): 2894-903, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22447903

ABSTRACT

The anaerobic metabolism of indoleacetate (indole-3-acetic acid [IAA]) in the denitrifying betaproteobacterium Azoarcus evansii was studied. The strain oxidized IAA completely and grew with a generation time of 10 h. Enzyme activities that transformed IAA were present in the soluble cell fraction of IAA-grown cells but were 10-fold downregulated in cells grown on 2-aminobenzoate or benzoate. The transformation of IAA did not require molecular oxygen but required electron acceptors like NAD(+) or artificial dyes. The first products identified were the enol and keto forms of 2-oxo-IAA. Later, polar products were observed, which could not yet be identified. The first steps likely consist of the anaerobic hydroxylation of the N-heterocyclic pyrrole ring to the enol form of 2-oxo-IAA, which is catalyzed by a molybdenum cofactor-containing dehydrogenase. This step is probably followed by the hydrolytic ring opening of the keto form, which is catalyzed by a hydantoinase-like enzyme. A comparison of the proteome of IAA- and benzoate-grown cells identified IAA-induced proteins. Owing to the high similarity of A. evansii with strain EbN1, whose genome is known, we identified a cluster of 14 genes that code for IAA-induced proteins involved in the early steps of IAA metabolism. These genes include a molybdenum cofactor-dependent dehydrogenase of the xanthine oxidase/aldehyde dehydrogenase family, a hydantoinase, a coenzyme A (CoA) ligase, a CoA transferase, a coenzyme B(12)-dependent mutase, an acyl-CoA dehydrogenase, a fusion protein of an enoyl-CoA hydratase and a 3-hydroxyacyl-CoA dehydrogenase, a beta-ketothiolase, and a periplasmic substrate binding protein for ABC transport as well as a transcriptional regulator of the GntR family. Five predicted enzymes form or act on CoA thioesters, indicating that soon after the initial oxidation of IAA and possibly ring opening, CoA thioesters are formed, and the carbon skeleton is rearranged, followed by a CoA-dependent thiolytic release of another CoA thioester. We propose a scheme of an anaerobic IAA metabolic pathway that ultimately leads to 2-aminobenzoyl-CoA or benzoyl-CoA.


Subject(s)
Azoarcus/metabolism , Indoleacetic Acids/metabolism , Anaerobiosis , Azoarcus/enzymology , Azoarcus/genetics , Azoarcus/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Metabolic Networks and Pathways , Molecular Sequence Data
8.
Front Public Health ; 10: 948205, 2022.
Article in English | MEDLINE | ID: mdl-36111186

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a highly contagious disease that has claimed the lives of millions of people worldwide in the last 2 years. Because of the disease's rapid spread, it is critical to diagnose it at an early stage in order to reduce the rate of spread. The images of the lungs are used to diagnose this infection. In the last 2 years, many studies have been introduced to help with the diagnosis of COVID-19 from chest X-Ray images. Because all researchers are looking for a quick method to diagnose this virus, deep learning-based computer controlled techniques are more suitable as a second opinion for radiologists. In this article, we look at the issue of multisource fusion and redundant features. We proposed a CNN-LSTM and improved max value features optimization framework for COVID-19 classification to address these issues. The original images are acquired and the contrast is increased using a combination of filtering algorithms in the proposed architecture. The dataset is then augmented to increase its size, which is then used to train two deep learning networks called Modified EfficientNet B0 and CNN-LSTM. Both networks are built from scratch and extract information from the deep layers. Following the extraction of features, the serial based maximum value fusion technique is proposed to combine the best information of both deep models. However, a few redundant information is also noted; therefore, an improved max value based moth flame optimization algorithm is proposed. Through this algorithm, the best features are selected and finally classified through machine learning classifiers. The experimental process was conducted on three publically available datasets and achieved improved accuracy than the existing techniques. Moreover, the classifiers based comparison is also conducted and the cubic support vector machine gives better accuracy.


Subject(s)
COVID-19 , Deep Learning , Moths , Animals , Humans , Neural Networks, Computer , X-Rays
9.
J Biol Chem ; 285(31): 23842-9, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20519501

ABSTRACT

The angiopoietins (Ang-1 and Ang-2) have been identified as agonistic and antagonistic ligands of the endothelial receptor tyrosine kinase Tie2, respectively. Both ligands have been demonstrated to induce translocation of Tie2 to cell-cell junctions. However, only Ang-1 induces Tie2-dependent Akt activation and subsequent survival signaling and endothelial quiescence. Ang-2 interferes negatively with Ang-1/Tie2 signaling, thereby antagonizing the Ang-1/Tie2 axis. Here, we show that both Ang-1 and Ang-2 recruit beta3 integrins to Tie2. This co-localization is most prominent in cell-cell junctions. However, only Ang-2 stimulation resulted in complex formation among Tie2, alphavbeta3 integrin, and focal adhesion kinase as evidenced by co-immunoprecipitation experiments. Focal adhesion kinase was phosphorylated in the FAT domain at Ser(910) upon Ang-2 stimulation and the adaptor proteins p130Cas and talin dissociated from alphavbeta3 integrin. The alphavbeta3 integrin was internalized, ubiquitinylated, and gated toward lysosomes. Taken together, the experiments define Tie2/alphavbeta3 integrin association-induced integrin internalization and degradation as mechanistic consequences of endothelial Ang-2 stimulation.


Subject(s)
Angiopoietin-2/metabolism , Endothelial Cells/cytology , Integrin alphaVbeta3/metabolism , Cell Communication , Cell Line , Cell Movement , Endothelium, Vascular/cytology , Humans , Immunoprecipitation , Lysosomes/metabolism , Models, Biological , Phosphorylation , Receptor, TIE-2/metabolism , Signal Transduction , Ubiquitin/chemistry
10.
Microcirculation ; 18(7): 598-607, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21851472

ABSTRACT

OBJECTIVE: Angiogenesis, a critical contributor to ocular as well as neoplastic diseases, is stimulated by endothelial production of angiopoietin-2 (Ang2). Our objective was to determine the requirement of ocular angiogenesis for Ang2 in animal models of disease. METHODS: We developed and compared the effect of a novel human Ang2 antibody with a pan-angiopoietin strategy on angiogenesis in ocular angiogenesis in animal models of oxygen-induced retinopathy, and laser photocoagulation and confirmed its efficacy in xenografted human colorectal tumors. RESULTS: Human anti-Ang2 and anti-angiopoietin1(Ang1)/Ang2 antibodies blocked colorectal carcinoma growth in immuno-compromised mice (p < 0.001, n = 6). Injection of 1 µg of Ang2 or Ang2/Ang1 antibody-inhibited angiogenesis in models of retinal (p < 0.001, n = 6), and choroidal neovascularization (p < 0.001, n = 11-13 per group) to levels similar to that with anti-VEGF antibodies. There was no difference between Ang2 specific and Ang1/Ang2 bi-specific antibodies. In vitro, Ang2 antibodies showed no cytotoxicity and did not inhibit endothelial cell migration or proliferation. CONCLUSION: Thus, human Ang2 antibodies are potentially therapeutic agents for ocular neovascularization in models of retinal and choroidal neovascularization, in the absence of VEGF inhibition.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Neovascularization, Pathologic/drug therapy , Ribonuclease, Pancreatic/antagonists & inhibitors , Animals , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Models, Animal , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Retinal Diseases/chemically induced , Retinal Diseases/drug therapy , Retinal Diseases/pathology , Transplantation, Heterologous
11.
Surg Neurol Int ; 12: 493, 2021.
Article in English | MEDLINE | ID: mdl-34754543

ABSTRACT

BACKGROUND: Intracranial pressure (ICP) monitoring has been variously explored as a diagnostic and therapeutic modality in many pathological conditions leading neurological injury. This monitoring standardly depends on an invasive procedure such as cranial or lumbar catheterization. The gold standard for ICP monitoring is through an intraventricular catheter, but this invasive technique is associated with certain risks such as haemorrhage and infection. (1) Also, it is a high-cost procedure and consequently not available in a variety of underprivileged places and clinical situations in which intracranial hypertension is prevalent (3). An accurate non-invasive and low-priced method to measure elevated ICP would therefore be desirable. Under these circumstances, Brazilian scientists developed a non-invasive method for intracranial pressure monitoring (ICP-NI), which uses an electric resistance extensometer that measures micro deformations of the skull and transforms it into an electrical signal. In this case report, the authors describe a pediatrician patient with the diagnosis of idiopathic intracranial hypertension who was successfully submitted to a lumbar puncture under monitorization with this device. CASE DESCRIPTION: 7 year old girl with progressive symptoms that lead to the diagnosis of idiopathic intracranial hypertension. The patient was submitted to a lumbar punction with continuous non-invasive ICP monitoring. CONCLUSION: Estimating ICP (non-invasive) from LP monitoring (invasive) often reflect inaccurate ICP results, and affects negatively on IIH diagnosis and a non-invasive diagnostic method could reduce the requirement for invasive approaches, improving patient health outcomes.

12.
Clin Cancer Res ; 15(4): 1384-92, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19228739

ABSTRACT

PURPOSE: The blood vessel-destabilizing Tie2 ligand angiopoietin-2 (Ang-2) acts in concert with the vascular endothelial growth factor/vascular endothelial growth factor receptor system to control vessel assembly during tumor progression. We hypothesized that circulating soluble Ang-2 (sAng-2) may be involved in melanoma progression. EXPERIMENTAL DESIGN: Serum samples (n=98) from melanoma patients (American Joint Committee on Cancer stages I-IV), biopsies of corresponding patients, and human melanoma cell lines were analyzed for expression of Ang-2 and S100beta. Multiple sera of a subcohort of 33 patients were tested during progression from stage III to IV. Small interfering RNA-based loss-of-function experiments were done to assess effects of Ang-2 on melanoma cells. RESULTS: Circulating levels of sAng-2 correlate with tumor progression in melanoma patients (P<0.0001) and patient survival (P=0.007). Analysis of serum samples during the transition from stage III to IV identified an increase of sAng-2 up to 400%. Comparative analyses revealed a 56% superiority of sAng-2 as predictive marker over the established marker S100beta. Immunohistochemistry and reverse transcription-PCR confirmed the prominent expression of Ang-2 by tumor-associated endothelial cells but identified Ang-2 also as a secreted product of melanoma cells themselves. Corresponding cellular experiments revealed that human melanoma-isolated tumor cells were Tie2 positive and that Ang-2 acted as an autocrine regulator of melanoma cell migration and invasion. CONCLUSIONS: The experiments establish sAng-2 as a biomarker of melanoma progression and metastasis correlating with tumor load and overall survival. The identification of an autocrine angiopoietin/Tie loop controlling melanoma migration and invasion warrants further functional experiments and validate the angiopoietin/Tie system as a promising therapeutic target for human melanomas.


Subject(s)
Angiopoietin-2/blood , Melanoma/blood , Skin Neoplasms/blood , Adult , Aged , Biomarkers, Tumor/blood , Cells, Cultured , Disease Progression , Endothelial Cells/chemistry , Female , Humans , Male , Melanoma/mortality , Melanoma/pathology , Melanoma/secondary , Middle Aged , Nerve Growth Factors/blood , Receptor, TIE-2/analysis , Receptor, TIE-2/physiology , S100 Calcium Binding Protein beta Subunit , S100 Proteins/blood , Skin Neoplasms/mortality , Skin Neoplasms/pathology
13.
Cancer Res ; 80(12): 2586-2598, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32303578

ABSTRACT

The angiopoietin (Angpt)-TIE signaling pathway controls vascular maturation and maintains the quiescent phenotype of resting vasculature. The contextual agonistic and antagonistic Tie2 ligand ANGPT2 is believed to be exclusively produced by endothelial cells, disrupting constitutive ANGPT1-TIE2 signaling to destabilize the microvasculature during pathologic disorders like inflammation and cancer. However, scattered reports have also portrayed tumor cells as a source of ANGPT2. Employing ISH-based detection of ANGPT2, we found strong tumor cell expression of ANGPT2 in a subset of patients with melanoma. Comparative analysis of biopsies revealed a higher fraction of ANGPT2-expressing tumor cells in metastatic versus primary sites. Tumor cell-expressed Angpt2 was dispensable for primary tumor growth, yet in-depth analysis of primary tumors revealed enhanced intratumoral necrosis upon silencing of tumor cell Angpt2 expression in the absence of significant immune and vascular alterations. Global transcriptional profiling of Angpt2-deficient tumor cells identified perturbations in redox homeostasis and an increased response to cellular oxidative stress. Ultrastructural analyses illustrated a significant increase of dysfunctional mitochondria in Angpt2-silenced tumor cells, thereby resulting in enhanced reactive oxygen species (ROS) production and downstream MAPK stress signaling. Functionally, enhanced ROS in Angpt2-silenced tumor cells reduced colonization potential in vitro and in vivo. Taken together, these findings uncover the hitherto unappreciated role of tumor cell-expressed ANGPT2 as an autocrine-positive regulator of metastatic colonization and validate ANGPT2 as a therapeutic target for a well-defined subset of patients with melanoma. SIGNIFICANCE: This study reveals that tumor cells can be a source of ANGPT2 in the tumor microenvironment and that tumor cell-derived ANGPT2 augments metastatic colonization by protecting tumor cells from oxidative stress.


Subject(s)
Angiopoietin-2/metabolism , Melanoma/secondary , Nevus/pathology , Skin Neoplasms/pathology , Angiopoietin-2/genetics , Animals , Autocrine Communication , Biopsy , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Female , Gene Expression Profiling , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Kaplan-Meier Estimate , MAP Kinase Signaling System , Melanoma/mortality , Mice , Reactive Oxygen Species/metabolism , Skin/pathology , Skin Neoplasms/mortality , Tissue Array Analysis , Tumor Microenvironment
14.
iScience ; 23(9): 101517, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32927263

ABSTRACT

Structural mutants of p53 induce global p53 protein destabilization and misfolding, followed by p53 protein aggregation. First evidence indicates that p53 can be part of protein condensates and that p53 aggregation potentially transitions through a condensate-like state. We show condensate-like states of fluorescently labeled structural mutant p53 in the nucleus of living cancer cells. We furthermore identified small molecule compounds that interact with the p53 protein and lead to dissolution of p53 structural mutant condensates. The same compounds lead to condensation of a fluorescently tagged p53 DNA-binding mutant, indicating that the identified compounds differentially alter p53 condensation behavior depending on the type of p53 mutation. In contrast to p53 aggregation inhibitors, these compounds are active on p53 condensates and do not lead to mutant p53 reactivation. Taken together our study provides evidence for structural mutant p53 condensation in living cells and tools to modulate this process.

15.
Angiogenesis ; 12(2): 125-37, 2009.
Article in English | MEDLINE | ID: mdl-19449109

ABSTRACT

The Angiopoietin/Tie system acts as a vascular specific ligand/receptor system to control endothelial cell survival and vascular maturation. The Angiopoietin family includes four ligands (Angiopoietin-1, Angiopoietin-2 and Angiopoietin-3/4) and two corresponding tyrosine kinase receptors (Tie1 and Tie2). Ang-1 and Ang-2 are specific ligands of Tie2 binding the receptor with similar affinity. Tie2 activation promotes vessel assembly and maturation by mediating survival signals for endothelial cells and regulating the recruitment of mural cells. Ang-1 acts in a paracrine agonistic manner inducing Tie2 phosphorylation and subsequent vessel stabilization. In contrast, Ang-2 is produced by endothelial cells and acts as an autocrine antagonist of Ang-1-mediated Tie2 activation. Ang-2 thereby primes the vascular endothelium to exogenous cytokines and induces vascular destabilization at higher concentrations. Ang-2 is strongly expressed in the vasculature of many tumors and it has been suggested that Ang-2 may act synergistically with other cytokines such as vascular endothelial growth factor to promote tumor-associated angiogenesis and tumor progression. The better mechanistic understanding of the Ang/Tie system is gradually paving the way toward the rationale exploitation of this vascular signaling system as a therapeutic target for neoplastic and non-neoplastic diseases.


Subject(s)
Angiopoietins/metabolism , Blood Vessels/embryology , Blood Vessels/metabolism , Morphogenesis , Animals , Blood Vessels/pathology , Humans , Ligands , Receptors, TIE/chemistry , Receptors, TIE/metabolism , Signal Transduction
16.
MAbs ; 8(3): 562-73, 2016.
Article in English | MEDLINE | ID: mdl-26864324

ABSTRACT

Vascular endothelial growth factor (VEGF)-A blockade has been validated clinically as a treatment for human cancers. Angiopoietin-2 (Ang-2) is a key regulator of blood vessel remodeling and maturation. In tumors, Ang-2 is up-regulated and an unfavorable prognostic factor. Recent data demonstrated that Ang-2 inhibition mediates anti-tumoral effects. We generated a tetravalent bispecific antibody (Ang-2-VEGF-TAvi6) targeting VEGF-A with 2 arms based on bevacizumab (Avastin®), and targeting Ang-2 with 2 arms based on a novel anti-Ang-2 antibody (LC06). The two Ang-2-targeting single-chain variable fragments are disulfide-stabilized and fused to the C-terminus of the heavy chain of bevacizumab. Treatment with Ang-2-VEGF-A-TAvi6 led to a complete abrogation of angiogenesis in the cornea micropocket assay. Metastatic spread and tumor growth of subcutaneous, orthotopic and anti-VEGF-A resistant tumors were also efficiently inhibited. These data further establish Ang-2-VEGF bispecific antibodies as a promising anti-angiogenic, anti-metastatic and anti-tumor agent for the treatment of cancer.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Antibodies, Bispecific , Antibodies, Neoplasm , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neoplasm Metastasis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays
17.
J R Soc Interface ; 12(110): 0546, 2015 09 06.
Article in English | MEDLINE | ID: mdl-26289655

ABSTRACT

Angiogenesis, the process by which new vessels form from existing ones, plays an important role in many developmental processes and pathological conditions. We study angiogenesis in the context of a highly controllable experimental environment: the cornea micropocket assay. Using a multidisciplinary approach that combines experiments, image processing and analysis, and mathematical modelling, we aim to provide mechanistic insight into the action of two angiogenic factors, vascular endothelial growth factor A (VEGF-A) and basic fibroblast growth factor (bFGF). We use image analysis techniques to extract quantitative data, which are both spatially and temporally resolved, from experimental images, and we develop a mathematical model, in which the corneal vasculature evolves in response to both VEGF-A and bFGF. The experimental data are used for model parametrization, while the mathematical model is used to assess the utility of the cornea micropocket assay and to characterize proposed synergies between VEGF-A and bFGF.


Subject(s)
Corneal Neovascularization , Fibroblast Growth Factor 2/metabolism , Models, Cardiovascular , Vascular Endothelial Growth Factor A/metabolism , Animals , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C
18.
Cancer Cell ; 26(6): 880-895, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25490450

ABSTRACT

Antiangiogenic tumor therapy has failed in the adjuvant setting. Here we show that inhibition of the Tie2 ligand angiopoietin-2 (Ang2) effectively blocks metastatic growth in preclinical mouse models of postsurgical adjuvant therapy. Ang2 antibody treatment combines well with low-dose metronomic chemotherapy (LDMC) in settings in which maximum-dose chemotherapy does not prove effective. Mechanistically, Ang2 blockade could be linked to quenching the inflammatory and angiogenic response of endothelial cells (ECs) in the metastatic niche. Reduced EC adhesion molecule and chemokine expression inhibits the recruitment of tumor-promoting CCR2(+)Tie2(-) metastasis-associated macrophages. Moreover, LDMC contributes to therapeutic efficacy by inhibiting the recruitment of protumorigenic bone marrow-derived myeloid cells. Collectively, these data provide a rationale for mechanism-guided adjuvant tumor therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/drug therapy , Adjuvants, Pharmaceutic/administration & dosage , Adjuvants, Pharmaceutic/adverse effects , Administration, Metronomic , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Animals , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cell Line, Tumor , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mammary Neoplasms, Experimental , Maximum Tolerated Dose , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Xenograft Model Antitumor Assays
19.
Science ; 343(6169): 416-9, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24458641

ABSTRACT

Liver regeneration requires spatially and temporally precisely coordinated proliferation of the two major hepatic cell populations, hepatocytes and liver sinusoidal endothelial cells (LSECs), to reconstitute liver structure and function. The underlying mechanisms of this complex molecular cross-talk remain elusive. Here, we show that the expression of Angiopoietin-2 (Ang2) in LSECs is dynamically regulated after partial hepatectomy. During the early inductive phase of liver regeneration, Ang2 down-regulation leads to reduced LSEC transforming growth factor-ß1 production, enabling hepatocyte proliferation by releasing an angiocrine proliferative brake. During the later angiogenic phase of liver regeneration, recovery of endothelial Ang2 expression enables regenerative angiogenesis by controlling LSEC vascular endothelial growth factor receptor 2 expression. The data establish LSECs as a dynamic rheostat of liver regeneration, spatiotemporally orchestrating hepatocyte and LSEC proliferation through angiocrine- and autocrine-acting Ang2, respectively.


Subject(s)
Angiopoietin-2/metabolism , Cell Proliferation , Endothelium, Vascular/metabolism , Hepatocytes/physiology , Liver Regeneration/physiology , Angiopoietin-2/genetics , Animals , Hepatectomy , Hepatocytes/cytology , Liver Regeneration/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Transforming Growth Factor beta/metabolism
20.
PLoS One ; 8(8): e70459, 2013.
Article in English | MEDLINE | ID: mdl-23940579

ABSTRACT

Genetic experiments (loss-of-function and gain-of-function) have established the role of Angiopoietin/Tie ligand/receptor tyrosine kinase system as a regulator of vessel maturation and quiescence. Angiopoietin-2 (Ang-2) acts on Tie2-expressing resting endothelial cells as an antagonistic ligand to negatively interfere with the vessel stabilizing effects of constitutive Ang-1/Tie-2 signaling. Ang-2 thereby controls the vascular response to inflammation-inducing as well as angiogenesis-inducing cytokines. This study was aimed at assessing the role of Ang-2 as an autocrine (i.e. endothelial-derived) regulator of rapid vascular responses (within minutes) caused by permeability-inducing agents. Employing two independent in vivo assays to quantitatively assess vascular leakage (tracheal microsphere assay, 1-5 min and Miles assay, 20 min), the immediate vascular response to histamine, bradykinin and VEGF was analyzed in Ang-2-deficient (Ang-2(-/-)) mice. In comparison to the wild type control mice, the Ang2(-/-) mice demonstrated a significantly attenuated response. The Ang-2(-/-) phenotype was rescued by systemic administration (paracrine) of an adenovirus encoding Ang-2. Furthermore, cytokine-induced intracellular calcium influx was impaired in Ang-2(-/-) endothelioma cells, consistent with reduced phospholipase activation in vivo. Additionally, recombinant human Ang-2 (rhAng-2) alone was unable to induce vascular leakage. In summary, we report here in a definite genetic setting that Ang-2 is critical for multiple vascular permeability-inducing cytokines.


Subject(s)
Angiopoietin-2/metabolism , Cytokines/pharmacology , Angiopoietin-2/genetics , Animals , Blotting, Western , Bradykinin/genetics , Bradykinin/metabolism , Capillary Permeability/drug effects , Capillary Permeability/genetics , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells , Lung/cytology , Lung/metabolism , Mice , Mice, Knockout , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL