Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Publication year range
1.
FASEB J ; 37(4): e22894, 2023 04.
Article in English | MEDLINE | ID: mdl-36961390

ABSTRACT

Regulation of vascular permeability to plasma is essential for tissue and organ homeostasis and is mediated by endothelial cell-to-cell junctions that tightly regulate the trafficking of molecules between blood and tissue. The single-pass transmembrane glycoprotein CD93 is upregulated in endothelial cells during angiogenesis and controls cytoskeletal dynamics. However, its role in maintaining homeostasis by regulating endothelial barrier function has not been elucidated yet. Here, we demonstrate that CD93 interacts with vascular endothelial (VE)-cadherin and limits its phosphorylation and turnover. CD93 deficiency in vitro and in vivo induces phosphorylation of VE-cadherin under basal conditions, displacing it from endothelial cell-cell contacts. Consistent with this, endothelial junctions are defective in CD93-/- mice, and the blood-brain barrier permeability is enhanced. Mechanistically, CD93 regulates VE-cadherin phosphorylation and turnover at endothelial junctions through the Rho/Rho kinase-dependent pathway. In conclusion, our results identify CD93 as a key regulator of VE-cadherin stability at endothelial junctions, opening up possibilities for therapeutic strategies directed to control vascular permeability.


Subject(s)
Cadherins , Endothelial Cells , Animals , Mice , Phosphorylation , Endothelial Cells/metabolism , Cadherins/genetics , Cadherins/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Capillary Permeability/physiology , Endothelium, Vascular/metabolism , Cells, Cultured , Adherens Junctions/metabolism
2.
Development ; 147(16)2020 08 24.
Article in English | MEDLINE | ID: mdl-32747434

ABSTRACT

Central nervous system (CNS) blood vessels contain a functional blood-brain barrier (BBB) that is necessary for neuronal survival and activity. Although Wnt/ß-catenin signaling is essential for BBB development, its downstream targets within the neurovasculature remain poorly understood. To identify targets of Wnt/ß-catenin signaling underlying BBB maturation, we performed a microarray analysis that identified Fgfbp1 as a novel Wnt/ß-catenin-regulated gene in mouse brain endothelial cells (mBECs). Fgfbp1 is expressed in the CNS endothelium and secreted into the vascular basement membrane during BBB formation. Endothelial genetic ablation of Fgfbp1 results in transient hypervascularization but delays BBB maturation in specific CNS regions, as evidenced by both upregulation of Plvap and increased tracer leakage across the neurovasculature due to reduced Wnt/ß-catenin activity. In addition, collagen IV deposition in the vascular basement membrane is reduced in mutant mice, leading to defective endothelial cell-pericyte interactions. Fgfbp1 is required cell-autonomously in mBECs to concentrate Wnt ligands near cell junctions and promote maturation of their barrier properties in vitro Thus, Fgfbp1 is a crucial extracellular matrix protein during BBB maturation that regulates cell-cell interactions and Wnt/ß-catenin activity.


Subject(s)
Blood-Brain Barrier/embryology , Collagen Type IV/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Collagen Type IV/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Pericytes/cytology , Pericytes/metabolism , beta Catenin/genetics
3.
Cancer Immunol Immunother ; 71(8): 2029-2040, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35018481

ABSTRACT

Galectin-1 (Gal1) is a glycan-binding protein that promotes tumor progression by several distinct mechanisms. Through direct binding to vascular endothelial growth factor (VEGF)-receptor 2, Gal1 is able to induce VEGF-like signaling, which contributes to tumor angiogenesis. Furthermore, several studies have demonstrated an immunosuppressive function of Gal1 through effects on both effector and regulatory T cells. Elevated Gal1 expression and secretion have been shown in many tumor types, and high Gal1 serum levels have been connected to poor prognosis in cancer patients. These findings suggest that therapeutic strategies directed against Gal1 would enable simultaneous targeting of angiogenesis, immune evasion and metastasis. In the current study, we have analyzed the potential of Gal1 as a cancer vaccine target. We show that it is possible to generate high anti-Gal1 antibody levels in mice immunized with a recombinant vaccine protein consisting of bacterial sequences fused to Gal1. Growth of Gal1 expressing melanomas was significantly impaired in the immunized mice compared to the control group. This was associated with improved perfusion of the tumor vasculature, as well as increased infiltration of macrophages and cytotoxic T cells (CTLs). The level of granzyme B, mainly originating from CTLs in our model, was significantly elevated in Gal1 vaccinated mice and correlated with a decrease in tumor burden. We conclude that vaccination against Gal1 is a promising pro-immunogenic approach for cancer therapy that could potentially enhance the effect of other immunotherapeutic strategies due to its ability to promote CTL influx in tumors.


Subject(s)
Cancer Vaccines , Galectin 1 , Melanoma , Tumor Burden , Animals , Cancer Vaccines/immunology , Galectin 1/metabolism , Melanoma/therapy , Mice , Neovascularization, Pathologic , T-Lymphocytes, Cytotoxic/metabolism , Vaccination
4.
Cell Mol Life Sci ; 77(9): 1745-1770, 2020 May.
Article in English | MEDLINE | ID: mdl-31690961

ABSTRACT

Tumor vascularization occurs through several distinct biological processes, which not only vary between tumor type and anatomic location, but also occur simultaneously within the same cancer tissue. These processes are orchestrated by a range of secreted factors and signaling pathways and can involve participation of non-endothelial cells, such as progenitors or cancer stem cells. Anti-angiogenic therapies using either antibodies or tyrosine kinase inhibitors have been approved to treat several types of cancer. However, the benefit of treatment has so far been modest, some patients not responding at all and others acquiring resistance. It is becoming increasingly clear that blocking tumors from accessing the circulation is not an easy task to accomplish. Tumor vessel functionality and gene expression often differ vastly when comparing different cancer subtypes, and vessel phenotype can be markedly heterogeneous within a single tumor. Here, we summarize the current understanding of cellular and molecular mechanisms involved in tumor angiogenesis and discuss challenges and opportunities associated with vascular targeting.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Animals , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Signal Transduction
5.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830297

ABSTRACT

During angiogenesis, cell adhesion molecules expressed on the endothelial cell surface promote the growth and survival of newly forming vessels. Hence, elucidation of the signaling pathways activated by cell-to-matrix adhesion may assist in the discovery of new targets to be used in antiangiogenic therapy. In proliferating endothelial cells, the single-pass transmembrane glycoprotein CD93 has recently emerged as an important endothelial cell adhesion molecule regulating vascular maturation. In this study, we unveil a signaling pathway triggered by CD93 that regulates actin cytoskeletal dynamics responsible of endothelial cell adhesion. We show that the Src-dependent phosphorylation of CD93 and the adaptor protein Cbl leads to the recruitment of Crk, which works as a downstream integrator in the CD93-mediated signaling. Moreover, confocal microscopy analysis of FRET-based biosensors shows that CD93 drives the coordinated activation of Rac1 and RhoA at the cell edge of spreading cells, thus promoting the establishment of cell polarity and adhesion required for cell motility.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , Receptors, Complement/metabolism , Signal Transduction/genetics , rhoA GTP-Binding Protein/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Polarity/genetics , Cells, Cultured , Humans , Membrane Glycoproteins/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-crk/metabolism , RNA Interference , Receptors, Complement/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
6.
BMC Cancer ; 20(1): 339, 2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32321460

ABSTRACT

BACKGROUND: Patients with metastatic renal cell cancer (mRCC) are commonly treated with the tyrosine kinase inhibitor sunitinib, which blocks signalling from vascular endothelial growth factor (VEGF) - and platelet-derived growth factor-receptors, inhibiting development of new blood vessels. There are currently no predictive markers available to select patients who will gain from this treatment. Epidermal growth factor, latrophilin and seven transmembrane domain-containing protein 1 (ELTD1) is up-regulated in tumor endothelial cells in many types of cancer and may be a putative predictive biomarker due to its association with ongoing angiogenesis. METHODS: ELTD1, CD34 and VEGF receptor 2 (VEGFR2) expressions were analysed in tumor vessels of renal cancer tissues from 139 patients with mRCC using immunohistochemistry. Ninety-nine patients were treated with sunitinib as the first or second-line therapy. Early toxicity, leading to the termination of the treatment, eliminated 22 patients from the analyses. The remaining (n = 77) patients were included in the current study. In an additional analysis, 53 sorafenib treated patients were evaluated. RESULTS: Patients with high ELTD1 expression in the tumor vasculature experienced a significantly better progression free survival (PFS) with sunitinib treatment as compared to patients with low ELTD1 expression (8 versus 5.5 months, respectively). The expression level of CD34 and VEGFR2 showed no correlation to sunitinib response. In sorafenib treated patients, no association with ELTD1 expression and PFS/OS was found. CONCLUSIONS: Our results identify tumor vessel ELTD1 expression as a positive predictive marker for sunitinib-treatment in patients suffering from mRCC. The negative results in the sorafenib treated group supports ELTD1 being a pure predictive and not a prognostic marker for sunitinib therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/secondary , Endothelial Cells/metabolism , Kidney Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , Sunitinib/therapeutic use , Adult , Aged , Antigens, CD34/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Female , Follow-Up Studies , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Eur J Nucl Med Mol Imaging ; 46(3): 569-579, 2019 03.
Article in English | MEDLINE | ID: mdl-30109401

ABSTRACT

PURPOSE: Oligodendrogliomas are heterogeneous tumors in terms of imaging appearance, and a deeper understanding of the histopathological tumor characteristics in correlation to imaging parameters is needed. We used PET-to-MRI-to-histology co-registration with the aim of studying intra-tumoral 11C-methionine (MET) uptake in relation to tumor perfusion and the protein expression of histological cell markers in corresponding areas. METHODS: Consecutive histological sections of four tumors covering the entire en bloc-removed tumor were immunostained with antibodies against IDH1-mutated protein (tumor cells), Ki67 (proliferating cells), and CD34 (blood vessels). Software was developed for anatomical landmarks-based co-registration of subsequent histological images, which were overlaid on corresponding MET PET scans and MRI perfusion maps. Regions of interest (ROIs) on PET were selected throughout the entire tumor volume, covering hot spot areas, areas adjacent to hot spots, and tumor borders with infiltrating zone. Tumor-to-normal tissue (T/N) ratios of MET uptake and mean relative cerebral blood volume (rCBV) were measured in the ROIs and protein expression of histological cell markers was quantified in corresponding regions. Statistical correlations were calculated between MET uptake, rCBV, and quantified protein expression. RESULTS: A total of 84 ROIs were selected in four oligodendrogliomas. A significant correlation (p < 0.05) between MET uptake and tumor cell density was demonstrated in all tumors separately. In two tumors, MET correlated with the density of proliferating cells and vessel cell density. There were no significant correlations between MET uptake and rCBV, and between rCBV and histological cell markers. CONCLUSIONS: The MET uptake in hot spots, outside hotspots, and in infiltrating tumor edges unanimously reflects tumor cell density. The correlation between MET uptake and vessel density and density of proliferating cells is less stringent in infiltrating tumor edges and is probably more susceptible to artifacts caused by larger blood vessels surrounding the tumor. Although based on a limited number of samples, this study provides histological proof for MET as an indicator of tumor cell density and for the lack of statistically significant correlations between rCBV and histological cell markers in oligodendrogliomas.


Subject(s)
Magnetic Resonance Imaging , Multimodal Imaging , Oligodendroglioma/diagnostic imaging , Oligodendroglioma/pathology , Positron-Emission Tomography , Adult , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Oligodendroglioma/surgery , Tumor Burden
8.
Cell Commun Signal ; 17(1): 55, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138217

ABSTRACT

BACKGROUND: In the endothelium, the single-pass membrane protein CD93, through its interaction with the extracellular matrix protein Multimerin-2, activates signaling pathways that are critical for vascular development and angiogenesis. Trafficking of adhesion molecules through endosomal compartments modulates their signaling output. However, the mechanistic basis coordinating CD93 recycling and its implications for endothelial cell (EC) function remain elusive. METHODS: Human umbilical vein ECs (HUVECs) and human dermal blood ECs (HDBEC) were used in this study. Fluorescence confocal microscopy was employed to follow CD93 retrieval, recycling, and protein colocalization in spreading cells. To better define CD93 trafficking, drug treatments and transfected chimeric wild type and mutant CD93 proteins were used. The scratch assay was used to evaluate cell migration. Gene silencing strategies, flow citometry, and quantification of migratory capability were used to determine the role of Rab5c during CD93 recycling to the cell surface. RESULTS: Here, we identify the recycling pathway of CD93 following EC adhesion and migration. We show that the cytoplasmic domain of CD93, by its interaction with Moesin and F-actin, is instrumental for CD93 retrieval in adhering and migrating cells and that aberrant endosomal trafficking of CD93 prevents its localization at the leading edge of migration. Moreover, the small GTPase Rab5c turns out to be a key component of the molecular machinery that is able to drive CD93 recycling to the EC surface. Finally, in the Rab5c endosomal compartment CD93 forms a complex with Multimerin-2 and active ß1 integrin, which is recycled back to the basolaterally-polarized cell surface by clathrin-independent endocytosis. CONCLUSIONS: Our findings, focusing on the pro-angiogenic receptor CD93, unveil the mechanisms of its polarized trafficking during EC adhesion and migration, opening novel therapeutic opportunities for angiogenic diseases.


Subject(s)
Blood Proteins/metabolism , Cell Adhesion , Cell Movement , Integrin beta1/metabolism , Membrane Glycoproteins/metabolism , Receptors, Complement/metabolism , rab5 GTP-Binding Proteins/metabolism , Cells, Cultured , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans
9.
Semin Cancer Biol ; 45: 23-35, 2017 08.
Article in English | MEDLINE | ID: mdl-28257957

ABSTRACT

Cell therapy is an advanced form of cancer immunotherapy that has had remarkable clinical progress in the past decade in the search for cure of cancer. Most success has been achieved for chimeric antigen receptor (CAR) T-cells where CAR T-cells targeting CD19 show very high complete response rates for patients with refractory acute B-cell acute lymphoblastic leukemia (ALL) and are close to approval for this indication. CD19 CAR T-cells are also effective against B-cell chronic lymphoblastic leukemia (CLL) and B-cell lymphomas. Although encouraging, CAR T-cells have not yet proven clinically effective for solid tumors. This is mainly due to the lack of specific and homogenously expressed targets to direct the T-cells against and a hostile immunosuppressive tumor microenvironment in solid tumors. Cancer vaccines based on dendritic cells (DC) are also making progress although clinical efficacy is still lacking. The likelihood of success is however increasing now when individual tumors can be sequences and patient-specific neoepitopes identified. Neoepitopes and/or neoantigens can then be included in patient-based DC vaccines. This review discusses recent advancements of DC vaccines and CAR T-cells with emphasis on the cancer-immunity cycle, and current efforts to design novel cell therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunity/drug effects , Neoplasms/immunology , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell- and Tissue-Based Therapy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Design , Genetic Engineering , Genetic Therapy , Humans , Immunity/genetics , Immunomodulation/drug effects , Immunomodulation/genetics , Immunotherapy/adverse effects , Immunotherapy/methods , Neoplasms/genetics , Neoplasms/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
10.
Angiogenesis ; 21(3): 425-532, 2018 08.
Article in English | MEDLINE | ID: mdl-29766399

ABSTRACT

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Subject(s)
Biological Assay/methods , Neoplasms , Neovascularization, Pathologic , Animals , Biological Assay/instrumentation , Guidelines as Topic , Humans , Mice , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
11.
Eur J Immunol ; 47(2): 385-393, 2017 02.
Article in English | MEDLINE | ID: mdl-27873300

ABSTRACT

Checkpoint blockade of CTLA-4 results in long-lasting survival benefits in metastatic cancer patients. However, patients treated with CTLA-4 blockade have suffered from immune-related adverse events, most likely due to the breadth of the induced T-cell activation. Here, we investigated the efficacy of a local low-dose anti-CTLA-4 administration for treatment of subcutaneous or orthotopic murine bladder 49 (MB49) bladder carcinoma in C57BL/6 mice. When MB49 tumors were grown s.c., peritumoral (p.t.) injection of anti-CTLA-4 treatment was equally effective as intravenous or s.c. (nontumor bearing flank) administration. Notably, p.t. injection was associated with lower circulating antibody levels and decreased IL-6 serum levels as compared to systemic treatment. Ultrasound-guided intratumoral anti-CTLA-4 antibody treatment of orthotopically growing MB49 tumors resulted in tumor regression, with more than tenfold reduction in systemic antibody levels as compared to i.v. or s.c. administration, in line with the compartmentally restrained nature of the bladder. Local anti-CTLA-4 therapy in combination with anti-PD-1 therapy resulted in complete responses, superior to each therapy alone. In addition, p.t. anti-CTLA-4 therapy was potentiated by depletion of regulatory T cells. Our results demonstrate that local anti-CTLA-4 antibody therapy is equally effective as systemic administration, but reduces systemic antibody levels and cytokine release, and enhances the response to anti-PD1 therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , CTLA-4 Antigen/immunology , Growth Inhibitors/therapeutic use , Immunotherapy/methods , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Urinary Bladder Neoplasms/therapy , Animals , Cell Line, Tumor , Cytokines/metabolism , Drug Therapy, Combination , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms, Experimental , Urinary Bladder Neoplasms/immunology
13.
BMC Cancer ; 16(1): 904, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27863473

ABSTRACT

BACKGROUND: Anillin (ANLN), an actin-binding protein required for cytokinesis, has recently been presented as part of a prognostic marker panel in breast cancer. The objective of the current study was to further explore the prognostic and functional value of ANLN as a single biomarker in breast cancer. METHODS: Immunohistochemical assessment of ANLN protein expression was performed in two well characterized breast cancer cohorts (n = 484) with long-term clinical follow-up data and the results were further validated at the mRNA level in a publicly available transcriptomics dataset. The functional relevance of ANLN was investigated in two breast cancer cell lines using RNA interference. RESULTS: High nuclear fraction of ANLN in breast tumor cells was significantly associated with large tumor size, high histological grade, high proliferation rate, hormone receptor negative tumors and poor prognosis in both examined cohorts. Multivariable analysis showed that the association between ANLN and survival was significantly independent of age in cohort I and significantly independent of proliferation, as assessed by Ki-67 expression in tumor cells, age, tumor size, ER and PR status, HER2 status and nodal status in cohort II. Analysis of ANLN mRNA expression confirmed that high expression of ANLN was significantly correlated to poor overall survival in breast cancer patients. Consistent with the role of ANLN during cytokinesis, transient knock-down of ANLN protein expression in breast cancer cell lines resulted in an increase of senescent cells and an accumulation of cells in the G2/M phase of the cell cycle with altered cell morphology including large, poly-nucleated cells. Moreover, ANLN siRNA knockdown also resulted in decreased expression of cyclins D1, A2 and B1. CONCLUSIONS: ANLN expression in breast cancer cells plays an important role during cell division and a high fraction of nuclear ANLN expression in tumor cells is correlated to poor prognosis in breast cancer patients, independent of Ki-67, tumor size, hormone receptor status, HER2 status, nodal status and age.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Cycle , Ki-67 Antigen/metabolism , Microfilament Proteins/metabolism , Adult , Biomarkers, Tumor/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cohort Studies , Cyclins/genetics , Cyclins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Microfilament Proteins/genetics , Microscopy, Confocal , Middle Aged , Prognosis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
14.
FASEB J ; 29(1): 227-38, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25361735

ABSTRACT

Antiangiogenic treatment targeting the vascular endothelial growth factor (VEGF) signaling pathway is in clinical use, but its effect on vascular function and the tumor microenvironment is poorly understood. Here, we investigate cross-talk between VEGF and proinflammatory TNF-α signaling in endothelial cells and its impact on leukocyte recruitment. We found that cotreatment with VEGF decreased TNF-α-induced Jurkat cell adhesion to human microvascular endothelial cells by 40%. This was associated with inhibition of TNF-α-mediated regulation of 86 genes, including 2 T-lymphocyte-attracting chemokines, CXCL10 and CXCL11 [TNF-α concentration 1 ng/ml; 50% inhibition/inhibitory concentration (IC50) VEGF, 3 ng/ml]. Notably, VEGF directly suppressed TNF-α-induced gene expression through negative cross-talk with the NF-κB-signaling pathway, leading to an early decrease in IFN regulatory factor 1 (IRF-1) expression and reduced phosphorylation of signal transducer and activator of transcription 1 (p-Stat1) at later times. Inhibition of VEGF signaling in B16 melanoma tumor-bearing mice by sunitinib treatment resulted in up-regulation of CXCL10 and CXCL11 in tumor vessels, accompanied by up to 18-fold increased infiltration of CD3(+) T-lymphocytes in B16 tumors. Our results demonstrate a novel role of VEGF in negative regulation of NF-κB signaling and endothelial activation in the tumor microenvironment and provide evidence that pharmacological inhibition of VEGF signaling enhances T-lymphocyte recruitment through up-regulation of chemokines CXCL10 and CXCL11.


Subject(s)
Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , NF-kappa B/metabolism , Tumor Microenvironment/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Adhesion , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Jurkat Cells , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Microfluidic Analytical Techniques , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
15.
Biochem Soc Trans ; 42(6): 1647-52, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25399584

ABSTRACT

Glioblastoma is characterized by microvascular proliferation and a highly abnormal dysfunctional vasculature. The glioblastoma vessels differ significantly from normal brain vessels morphologically, functionally and molecularly. The present review provides a brief overview of the current understanding of the formation, functional abnormalities and specific gene expression of glioblastoma vessels and the consequences of vascular abnormalization for the tumour microenvironment.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Glioblastoma/blood supply , Glioblastoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Brain Neoplasms/pathology , Cell Hypoxia , Gene Expression , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/pathology , Tumor Microenvironment
16.
Histopathology ; 64(3): 365-79, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24410805

ABSTRACT

AIMS: In 2010, four subtypes (classical, proneural, mesenchymal, and neural) of glioblastoma multiforme (GBM) were defined by molecular genetic analyses. The objective of this study was to assess whether gliomas, independently of the type and grade, could be subdivided into protein-based subtypes. METHODS AND RESULTS: A tissue microarray (TMA) approach was applied to incorporate tissue samples of low-grade and high-grade gliomas into five TMAs. High expression levels of epidermal growth factor receptor (EGFR), CD44, c-MER proto-oncogene tyrosine kinase (MERTK), platelet-derived growth factor receptor α, p53, oligodendrocyte transcription factor 2 (OLIG2) and isocitrate dehydrogenase 1 with the R132H mutation were assessed using immunohistochemistry (IHC). Glioma could be subdivided into four subtypes by IHC. The majority of the low-grade gliomas were of the proneural subtype, i.e. high p53 expression (63% of grade II). The classical subtype, with high EGFR and low p53 expression, was most common in GBMs (39%), followed by the proneural (29%) and mesenchymal (with high CD44 and MERTK expression) (29%) subtypes, a frequency that is in line with previously published data based on molecular genetics. CONCLUSIONS: Assessment of the expression of the five proteins EGFR, CD44, MERTK, p53 and OLIG2 is sufficient for subtyping gliomas, and can be recommended for implementation in clinical practice for both low-grade and high-grade gliomas.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Adult , Aged , Aged, 80 and over , Algorithms , Astrocytoma/classification , Astrocytoma/metabolism , Astrocytoma/pathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/classification , ErbB Receptors/metabolism , Female , Glioblastoma/classification , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/classification , Humans , Hyaluronan Receptors/metabolism , Immunohistochemistry , Male , Middle Aged , Neoplasm Grading/methods , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglioma/classification , Oligodendroglioma/metabolism , Oligodendroglioma/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Retrospective Studies , Tissue Array Analysis , Tumor Suppressor Protein p53/metabolism , World Health Organization , Young Adult , c-Mer Tyrosine Kinase
17.
FASEB J ; 27(1): 151-62, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23033322

ABSTRACT

The molecular chaperone αB-crystallin has emerged as a target for cancer therapy due to its expression in human tumors and its role in regulating tumor angiogenesis. αB-crystallin also reduces neuroinflammation, but its role in other inflammatory conditions has not been investigated. Here, we examined whether αB-crystallin regulates inflammation associated with tumors and ischemia. We found that CD45(+) leukocyte infiltration is 3-fold increased in tumors and ischemic myocardium in αB-crystallin-deficient mice. Notably, αB-crystallin is prominently expressed in CD11b(+) Gr-1(+) immature myeloid cells (IMCs), known as regulators of angiogenesis and immune responses, while lymphocytes and mature granulocytes show low αB-crystallin expression. αB-Crystallin deficiency results in a 3-fold higher accumulation of CD11b(+) Gr-1(+) IMCs in tumors and a significant rise in CD11b(+) Gr-1(+) IMCs in spleen and bone marrow. Similarly, we noted a 2-fold increase in CD11b(+) Gr-1(+) IMCs in chronically inflamed livers in αB-crystallin-deficient mice. The effect of αB-crystallin on IMC accumulation is limited to pathological conditions, as CD11b(+) Gr-1(+) IMCs are not elevated in naive mice. Through ex vivo differentiation of CD11b(+) Gr-1(+) cells, we provide evidence that αB-crystallin regulates systemic expansion of IMCs through a cell-intrinsic mechanism. Our study suggests a key role of αB-crystallin in limiting expansion of CD11b(+) Gr-1(+) IMCs in diverse pathological conditions.


Subject(s)
Bone Marrow Cells/immunology , CD11b Antigen/immunology , Crystallins/physiology , Teratocarcinoma/pathology , Animals , Base Sequence , Cell Differentiation , DNA Primers , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Teratocarcinoma/immunology
18.
Cancer Cell ; 42(3): 328-330, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38366590

ABSTRACT

Brain metastases are clinically challenging due to the unique brain microenvironment. In this issue of Cancer Cell, Bejarano et al. use transcriptional profiling and data integration to shed light on the molecular and cellular composition of the vasculature in brain metastases, identifying CD276 as an immunomodulatory target for therapy.


Subject(s)
Brain Neoplasms , Humans , Brain Neoplasms/secondary , Immunomodulation , Tumor Microenvironment , B7 Antigens
19.
JCI Insight ; 9(7)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441970

ABSTRACT

Compromised vascular integrity facilitates extravasation of cancer cells and promotes metastatic dissemination. CD93 has emerged as a target for antiangiogenic therapy, but its importance for vascular integrity in metastatic cancers has not been evaluated. Here, we demonstrate that CD93 participates in maintaining the endothelial barrier and reducing metastatic dissemination. Primary melanoma growth was hampered in CD93-/- mice, but metastatic dissemination was increased and associated with disruption of adherens and tight junctions in tumor endothelial cells and elevated expression of matrix metalloprotease 9 at the metastatic site. CD93 directly interacted with vascular endothelial growth factor receptor 2 (VEGFR2) and its absence led to VEGF-induced hyperphosphorylation of VEGFR2 in endothelial cells. Antagonistic anti-VEGFR2 antibody therapy rescued endothelial barrier function and reduced the metastatic burden in CD93-/- mice to wild-type levels. These findings reveal a key role of CD93 in maintaining vascular integrity, which has implications for pathological angiogenesis and endothelial barrier function in metastatic cancer.


Subject(s)
Endothelial Cells , Neoplasms , Animals , Mice , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Angiogenesis ; 16(4): 975-83, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23929007

ABSTRACT

αB-crystallin is a small heat shock protein, which has pro-angiogenic properties by increasing survival of endothelial cells and secretion of vascular endothelial growth factor A. Here we demonstrate an additional role of αB-crystallin in regulating vascular function, through enhancing tumor necrosis factor α (TNF-α) induced expression of endothelial adhesion molecules involved in leukocyte recruitment. Ectopic expression of αB-crystallin in endothelial cells increases the level of E-selectin expression in response to TNF-α, and enhances leukocyte-endothelial interaction in vitro. Conversely, TNF-α-induced expression of intercellular adhesion molecule 1, vascular cell adhesion molecule 1 and E-selectin is markedly inhibited in endothelial cells isolated from αB-crystallin-deficient mice. This is associated with elevated levels of IκB in αB-crystallin deficient cells and incomplete degradation upon TNF-α stimulation. Consistent with this, endothelial adhesion molecule expression is reduced in inflamed vessels of αB-crystallin deficient mice, and leukocyte rolling velocity is increased. Our data identify αB-crystallin as a new regulator of leukocyte recruitment, by enhancing pro-inflammatory nuclear factor κ B-signaling and endothelial adhesion molecule expression during endothelial activation.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Endothelial Cells/cytology , Leukocyte Rolling/physiology , Leukocytes/cytology , NF-kappa B/metabolism , alpha-Crystallin B Chain/physiology , alpha-Crystallins/deficiency , Active Transport, Cell Nucleus , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/genetics , Cell Line , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , I-kappa B Proteins/biosynthesis , I-kappa B Proteins/genetics , Inflammation , Jurkat Cells , Male , Mice , Microvessels/cytology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transduction, Genetic , Tumor Necrosis Factor-alpha/physiology , Up-Regulation , alpha-Crystallin B Chain/genetics , alpha-Crystallins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL