TY - THES A1 - O'Rourke, Fiona T1 - Interactions of Bartonella henselae with myeloid angiogenic cells and consequences for pathological angiogenesis N2 - Vascular tumors associated with chronic B. henselae infections are unique examples of infection-associated pathological angiogenesis. The chaotic vascular architecture and prominent myeloid infiltrate of B. henselae induced vascular lesions show many similarities with malignant tumors. In human cancers infiltrating myeloid cells play a decisive role in tumor progression and vascularization. In particular, tumor associated macrophages (TAMs) transform the tumor microenvironment, drive tumor invasion and vascularization through secretion of pro-angiogenic and immune modulatory cytokines and participation in matrix remodeling processes. Myeloid angiogenic cells (MACs) are a subset of circulating myeloid progenitors with important roles in regenerative and pathological angiogenesis and a critical involvement in tumor vascularization. The phenotypic plasticity and importance of MACs in pathological angiogenic processes, position these cells as key potential players in B. henselae associated vascular tumor formation. To investigate the possible role of MACs in B henselae induced pathological angiogenesis, the objective of this study was to examine the interaction of B. henselae with MACs and determine how this may affect their angiogenic capacity. Building on previous work by MÓ“ndle (2005) this study has demonstrated that MACs are susceptible to infection with B. henselae and reside in intracellular vacuoles. As in endothelial cells, infection of MACs with B. henselae was associated with inhibition of apoptosis and activation of endogenous angiogenic programs including activation of the angiogenic transcription factor HIF-1. In addition to angiogenic re-programming on a molecular level B. henselae infection increases MAC functional angiogenic capacity. B. henselae infected MACs were found to integrate into growing endothelium and increase the rate of angiogenic sprouting in a paracrine manner. When cultured in a Matrigel capillary formation assay, infected MACs were also found to form networks of capillary-like structures that were stable over long periods of time. The B. henselae pathogenicity factor BadA was essential for the induction of this vascular mimicry phenotype as well as the activation of HIF-1 in infected MACs indicating that this factor may play an important role in MAC angiogenic re-programming. Examination of infected MACs via FACS analysis, cytospin immunohistochemistry and qRT-PCR revealed that endothelial differentiation does not play a role in the B. henselae induced pro-angiogenic phenotype. Instead, MACs were shown to be myeloid in phenotype displaying typical macrophage markers which were upregulated upon B. henselae infection and maintained over long-term culture. The increased angiogenic activity of B. henselae infected MACs was found to be associated with a broad phenotypic reprogramming in infected cells. In particular, gene expression programs related to angiogenesis, structural organization, apoptosis, sterol metabolism and immune regulation, were upregulated. Further examination of microarray gene expression profiles revealed that B. henselae infected MACs display a predominantly M2 anti-inflammatory macrophage activation status. Finally, examination of the paracrine microenvironment created by B. henselae infected MACs revealed a diverse cytokine secretion profile dominated by inflammatory-angiogenic cytokines and matrix remodeling elements and lacking expression of some of the most important cytokines involved in the expansion of the inflammatory response. This B. henselae induced activation status was demonstrated to be distinct from the general inflammatory response induced by E. coli LPS treatment. Comparison of B. henselae infected MACs to TAMs revealed many parallels in functional and phenotypic characteristics. Both TAMs and B. henselae infected MACs demonstrate increased angiogenic capacity, invasive, and immune modulatory phenotypes and the ability to participate in the formation of vascular mimicry phenotypes under angiogenic pressure. Furthermore, the pro-angiogenic paracrine microenvironment created by B. henselae infected MACs shows many similarities to the TAM-created tumor-microenvironment. In conclusion, these investigations have demonstrated that the infection of MACs with B. henselae results in the phenotypic re-programming towards TAM-like cells with increased pro-angiogenic, invasive and immune-modulatory qualities. The results of this study elucidate new aspects of B. henselae pathogenicity in myeloid cells and highlight the role of these cells as paracrine mediators of B. henselae induced vascular tumor formation. In addition, these findings demonstrate that manipulation of myeloid cells by pathogenic bacteria can contribute to microenvironmental regulation of pathological tissue growth and suggest parallels underlying bacterial infections and cancer. KW - Bartonella henselae KW - bacillary angiomatosis KW - myeloid angiogenic cells KW - bacterial infection KW - angiogenesis Y1 - 2015 UR - http://publikationen.ub.uni-frankfurt.de/frontdoor/index/index/docId/38652 UR - https://nbn-resolving.org/urn:nbn:de:hebis:30:3-386523 PB - Univ.-Bibliothek CY - Frankfurt am Main ER -