Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Breast Cancer Res Treat ; 200(2): 293-304, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37222874

ABSTRACT

PURPOSE: Angiogenesis is crucial for tumor growth and is one of the hallmarks of cancer. In this study, we analyzed microvessel density, vessel median size, and perivascular a-SMA expression as prognostic biomarkers in breast cancer. METHODS: Dual IHC staining was performed where alpha-SMA antibodies were used together with antibodies against the endothelial cell marker CD34. Digital images of stainings were analyzed to extract quantitative data on vessel density, vessel size, and perivascular alpha-SMA status. RESULTS: The analyses in the discovery cohort (n = 108) revealed a statistically significant relationship between large vessel size and shorter disease-specific survival (p = 0.007, log-rank test; p = 0.01, HR 3.1; 95% CI 1.3-7.4, Cox-regression analyses). Subset analyses indicated that the survival association of vessel size was strengthened in ER + breast cancer. To consolidate these findings, additional analyses were performed on a validation cohort (n = 267) where an association between large vessel size and reduced survival was also detected in ER + breast cancer (p = 0.016, log-rank test; p = 0.02; HR 2.3, 95% CI 1.1-4.7, Cox-regression analyses). CONCLUSION: Alpha-SMA/CD34 dual-IHC staining revealed breast cancer heterogeneity regarding vessel size, vessel density, and perivascular a-SMA status. Large vessel size was linked to shorter survival in ER + breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Receptors, Estrogen/metabolism , Prognosis , Biomarkers, Tumor/metabolism
2.
J Pathol Clin Res ; 6(1): 69-82, 2020 01.
Article in English | MEDLINE | ID: mdl-31605508

ABSTRACT

Cancer-associated fibroblasts are essential modifiers of the tumor microenvironment. The collagen-binding integrin α11ß1 has been proposed to be upregulated in a pro-tumorigenic subtype of cancer-associated fibroblasts. Here, we analyzed the expression and clinical relevance of integrin α11ß1 in a large breast cancer series using a novel antibody against the human integrin α11 chain. Several novel monoclonal antibodies against the integrin α11 subunit were tested for use on formalin-fixed paraffin-embedded tissues, and Ab 210F4B6A4 was eventually selected to investigate the immunohistochemical expression in 392 breast cancers using whole sections. mRNA data from METABRIC and co-expression patterns of integrin α11 in relation to αSMA and cytokeratin-14 were also investigated. Integrin α11 was expressed to varying degrees in spindle-shaped cells in the stroma of 99% of invasive breast carcinomas. Integrin α11 co-localized with αSMA in stromal cells, and with αSMA and cytokeratin-14 in breast myoepithelium. High stromal integrin α11 expression (66% of cases) was associated with aggressive breast cancer features such as high histologic grade, increased tumor cell proliferation, ER negativity, HER2 positivity, and triple-negative phenotype, but was not associated with breast cancer specific survival at protein or mRNA levels. In conclusion, high stromal integrin α11 expression was associated with aggressive breast cancer phenotypes.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Integrin alpha Chains/biosynthesis , Aged , Antibodies, Monoclonal , Carcinoma/pathology , Female , Humans , Integrin alpha Chains/analysis , Integrins/analysis , Integrins/biosynthesis , Middle Aged , Phenotype , Receptors, Collagen/analysis , Receptors, Collagen/biosynthesis
3.
J Pathol Clin Res ; 4(2): 114-123, 2018 04.
Article in English | MEDLINE | ID: mdl-29665322

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cell carcinoma, and anti-angiogenic treatment is currently first line therapy for metastatic ccRCC (mccRCC). Response rates and duration of response show considerable variation, and adverse events have a major influence on patient quality of life. The need for predictive biomarkers to select responders to receptor tyrosine kinase inhibitors upfront is urgent. We investigated the predictive value of immunohistochemical biomarkers associated with angiogenesis and systemic inflammation in mccRCC. Forty-six patients with metastatic or non-resectable ccRCC treated with sunitinib were included. Metastatic and/or primary tumour tissue was stained by immunohistochemistry for selected markers related to angiogenesis [vascular endothelial growth factor A (VEGF-A), VEGF receptor 2 (VEGFR2), platelet-derived growth factor receptor ß (PDGFRß), and heat shock protein 27 (HSP27)] and immune responses [Interleukin 6 receptor α (IL6Rα), interleukin-6 (IL6), and jagged1 (JAG1)]. The predictive potential of the candidate markers was assessed by correlations with response rates (RECIST). In addition, progression free survival (PFS) and overall survival (OS) were analysed. Low tumour cell expression of IL6Rα was significantly associated with improved response to sunitinib (Fisher's exact test, p = 0.03), but not with PFS or OS. Median/high expression of IL6Rα showed significant association with median/high expression of VEGF-A and HSP27. Furthermore, low expression of IL6 was significantly associated with improved PFS, but not OS or response rates. High expression of IL6 was significantly associated with high expression of JAG1, VEGF-A, VEGFR2, and PDGFRß. Loss of tumour cell expression of IL6Rα in mccRCC patients treated with sunitinib predicts improved treatment response, and might represent a candidate predictive marker.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Sunitinib/administration & dosage , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/pathology , Female , Humans , Immunohistochemistry , Interleukin-6/metabolism , Kidney Neoplasms/diagnosis , Kidney Neoplasms/pathology , Male , Middle Aged , Neovascularization, Pathologic , Prognosis , Prospective Studies , Receptors, Interleukin-6/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
J Immunol ; 198(8): 3318-3325, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28258201

ABSTRACT

IL-33, required for viral clearance by cytotoxic T cells, is generally expressed in vascular endothelial cells in healthy human tissues. We discovered that endothelial IL-33 expression was stimulated as a response to adenoviral transduction. This response was dependent on MRE11, a sensor of DNA damage that can also be activated by adenoviral DNA, and on IRF1, a transcriptional regulator of cellular responses to viral invasion and DNA damage. Accordingly, we observed that endothelial cells responded to adenoviral DNA by phosphorylation of ATM and CHK2 and that depletion or inhibition of MRE11, but not depletion of ATM, abrogated IL-33 stimulation. In conclusion, we show that adenoviral transduction stimulates IL-33 expression in endothelial cells in a manner that is dependent on the DNA-binding protein MRE11 and the antiviral factor IRF1 but not on downstream DNA damage response signaling.


Subject(s)
Adenoviridae Infections/immunology , DNA Damage/immunology , Human Umbilical Vein Endothelial Cells/immunology , Interleukin-33/immunology , Adenoviridae , Adenoviridae Infections/metabolism , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoblotting , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-1/metabolism , Interleukin-33/biosynthesis , MRE11 Homologue Protein , Polymerase Chain Reaction , Transfection
5.
Arterioscler Thromb Vasc Biol ; 33(2): e47-55, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23162017

ABSTRACT

OBJECTIVE: Interleukin (IL)-33 is a nuclear protein that is released from stressed or damaged cells to act as an alarmin. We investigated the effects of IL-33 on endothelial cells, using the prototype IL-1 family member, IL-1ß, as a reference. METHODS AND RESULTS: Human umbilical vein endothelial cells were stimulated with IL-33 or IL-1ß, showing highly similar phosphorylation of signaling molecules, induction of adhesion molecules, and transcription profiles. However, intradermally injected IL-33 elicited significantly less proinflammatory endothelial activation when compared with IL-1ß and led us to observe that quiescent endothelial cells (ppRb(low)p27(high)) were strikingly resistant to IL-33. Accordingly, the IL-33 receptor was preferentially expressed in nonquiescent cells of low-density cultures, corresponding to selective induction of adhesion molecules and chemokines. Multiparameter phosphoflow cytometry confirmed that signaling driven by IL-33 was stronger in nonquiescent cells. Manipulation of nuclear IL-33 expression by siRNA or adenoviral transduction revealed no functional link between nuclear, endogenous IL-33, and exogenous IL-33 responsiveness. CONCLUSIONS: In contrast to other inflammatory cytokines, IL-33 selectively targets nonquiescent endothelial cells. By this novel concept, quiescent cells may remain nonresponsive to a proinflammatory stimulus that concomitantly triggers a powerful response in cells that have been released from contact inhibition.


Subject(s)
Cell Proliferation , Dermatitis/immunology , Endothelial Cells/immunology , Inflammation Mediators/metabolism , Interleukins/metabolism , Skin/blood supply , Adenoviridae/genetics , Animals , Biopsy , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dermatitis/pathology , E-Selectin/metabolism , Endothelial Cells/pathology , Female , Flow Cytometry , Genetic Vectors , Human Umbilical Vein Endothelial Cells/immunology , Humans , Interleukin-1beta/metabolism , Interleukin-33 , Interleukins/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neovascularization, Physiologic , Phosphorylation , RNA Interference , Receptors, Interleukin/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Transcription, Genetic , Transduction, Genetic , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Am J Pathol ; 181(3): 1099-111, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22809957

ABSTRACT

The molecular mechanisms that drive expression of the alarmin interleukin-33 (IL-33) in endothelial cells are unknown. Because nuclear IL-33 is a marker of endothelial cell quiescence (corroborated in this study by coexpression of cyclin-dependent kinase inhibitor p27(Kip1)), we hypothesized that Notch signaling might be involved in regulating IL-33 expression. Activation of Notch1 by immobilized Notch ligands was sufficient to induce nuclear IL-33 expression in cultured endothelial cells. Conversely, IL-33 expression was inhibited by the γ-secretase inhibitor DAPT or by inhibiting the function of Dll4, Jagged1, Notch1, or the canonical Notch transcription factor RBP-Jκ. Insensitivity to cycloheximide indicated that IL-33 was a direct target of Notch signaling, well in line with the identification of several conserved RBP-Jκ binding sites in the IL33 gene. The in vivo expression of Dll4 but not of Jagged1 was well correlated with expression of IL-33 in quiescent vessels, and subcutaneous injection of DAPT in healthy skin reduced IL-33 expression, indicating that Notch signaling was involved. On the other hand, loss of IL-33 during angiogenesis occurred despite sustained Dll4 and Notch1 expression, suggesting that other signals may override the IL-33-driving signal in this context. Taken together, our data demonstrate that endothelial nuclear IL-33 is induced by Notch and that Dll4 may be the dominant ligand responsible for this signaling in vivo.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Interleukins/metabolism , Receptor, Notch1/metabolism , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Binding Sites , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dipeptides/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Female , Genetic Loci/genetics , Genome, Human/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-33 , Interleukins/genetics , Jagged-1 Protein , Male , Membrane Proteins/metabolism , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Protein Binding/drug effects , Rats , Rats, Wistar , Receptor, Notch1/antagonists & inhibitors , Serrate-Jagged Proteins , Signal Transduction/drug effects , Signal Transduction/genetics , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...