scholarly journals Bartonella henselae Persistence within Mesenchymal Stromal Cells Enhances Endothelial Cells Activation and Infectability Amplifying the Angiogenic Process.

2021 ◽  
Author(s):  
Scutera Sara ◽  
Mitola Stefania ◽  
Sparti Rosaria ◽  
Salvi Valentina ◽  
Grillo Elisabetta ◽  
...  

Some bacterial pathogens can manipulate the angiogenic response, suppressing or inducing it for their own ends. In humans, B. henselae is associated with cat-scratch disease and vasculoproliferative disorders such as bacillary angiomatosis and bacillary peliosis. Although endothelial cells (ECs) support the pathogenesis of Bartonella , the mechanisms by which Bartonella induces EC activation are not completely clear, as well as the possible contribution of other cells recruited at the site of infection. Mesenchymal stromal cells (MSCs) are endowed with angiogenic potential and play a dual role in infections exerting antimicrobial properties but also acting as a shelter for pathogens. Here we delved into the role of MSCs as reservoir of Bartonella and modulator of EC functions. B. henselae readily infected MSCs and survived in perinuclear bound vacuoles for up to 8 days. Infection enhanced MSC proliferation and the expression of EGFR, TLR2 and NOD1, proteins that are involved in bacterial internalization and cytokine production. Secretome analysis revealed that infected MSCs secreted higher levels of the proangiogenic factors VEGF, FGF-7, MMP-9, PIGF, serpin E1, TSP-1, uPA, IL-6, PDGF-D, CCL5 and CXCL8. Supernatants from B. henselae -infected MSCs increased the susceptibility of ECs to B. henselae infection and enhanced EC proliferation, invasion and reorganization in tube-like structures. Altogether, these results candidate MSCs as a still underestimated niche for B. henselae persistent infection and reveal a MSC-EC crosstalk that may contribute to exacerbate bacterial-induced angiogenesis and granuloma formation.

2020 ◽  
Author(s):  
Sara Scutera ◽  
Stefania Mitola ◽  
Rosaria Sparti ◽  
Giorgia Piersigilli ◽  
Elisabetta Grillo ◽  
...  

Abstract Background B henselae is in humans the aetiologic agent of cat-scratch disease and of the vasculoproliferative disorders bacillary angiomatosis and bacillary peliosis. Although endothelial cells are crucial in the pathogenesis other cell types function as reservoir and contribute to pathological angiogenesis. Among them, mesenchymal stromal cells (MSCs) can sense pathogens and mount an appropriate cytokine/chemokine response through different Pattern Recognition Receptors (PRRs). MSCs exert direct antimicrobial effector function but may also shelter bacteria such as M. tuberculosis. Methods Adipose-derived MSCs were infected with B. henselae and analyzed for bacterial persistence by gentamicin protection assay, immunohistochemistry and immunofluorescence. Involvement of PRRs in bacterial infection was evaluated through gene and protein expression analysis. The effect of infection on MSC proliferation, apoptosis and release of soluble factors was assessed. The role of infected-MSC conditioned medium in promoting Bartonella infection of endothelial cells and angiogenesis was demonstrated using respectively gentamicin protection assay and different pro-angiogenic assays including spheroid, wound healing and morphogenesis. Results B. henselae can readily infect MSCs and survive in perinuclear bound vacuoles for up to 8 days. Bartonella infection stimulates MSC proliferation and upregulation of EGFR and of the two pattern recognition receptors (PRRs) TLR2 and NOD1. Specific inhibition of EGFR reduces bacterial internalization and treatment with anti-TLR2 neutralizing antibody or EGFR/NOD1 inhibitors significantly downmodulates CXCL8 production. Secretome analysis shows that, in addition to CXCL8, infected MSCs secrete higher levels of the proangiogenic factors VEGF, FGF-7, MMP-9, PIGF, serpin E1, TSP-1, uPA, IL-6, CCL5 and PDGF-D. Importantly, supernatants from B. henselae-infected MSCs increase the susceptibility of ECs to B. henselae infection while enhancing EC proangiogenic potential. Conclusions Altogether, these findings indicate that MSCs constitute a novel niche for B. henselae, which favors the persistence of vascular proliferative disorders.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2425-2425
Author(s):  
Felicia Ciuculescu ◽  
Melanie Giesen ◽  
Erika Deak ◽  
Erhard Seifried ◽  
Tanja Nicole Hartmann ◽  
...  

Abstract Mesenchymal Stromal Cells (MSCs) are being developed as a cellular therapeutic, and used clinically used for induction of immunomodulation and suppression of inflammation after application via the intravenous route. However, relatively little is known on how MSCs interact with the vessel wall to induce tissue-specific egress. To elucidate potentially underlying mechanisms, we analyzed human bone-marrow derived MSCs in parallel plate flow chambers and characterized their interaction with endothelial cells or immobilized endothelial ligands as to tethering and rolling, sudden arrest, adhesion strengthening, and transendothelial migration. Flow cytometric analysis of MSCs confirmed the known expression predominantly of integrins α4, α5 and β1, and revealed only low detectable levels of a range of chemokine receptors including CCR1, 5, 6, 7 and CXCR1, 4 and 5. To investigate a potential role of chemokines in the adhesion process of MSCs, we first seeded MSCs without shear onto immobilized recombinant (r) VCAM-Fc fusion protein without or with co-immobilized chemokines in parallel plate flow chambers for 3 min, and subsequently started flow by stepwise increasing shear stress from 0.35 to 15 dynes/cmE2. Co-immobilization of various chemokines including CCL15/HCC-2, a known activator of CCR1 and 3, CCL20/LARC (of CCR6), CCL19/ELC (of CCR7), CXCL8/IL-8 (of CXCR1/2), CXCL12/SDF-1 (of CXCR4) and CXCL13/BCA-1 (of CXCR5) with rVCAM-1 increased shear-resistant binding of MSCs by up to four-fold compared to controls on rVCAM alone, with only moderate increases through CXCL8/IL-8 and CXCL12/SDF-1 and strongest effects through CCL19/ELC and CCL20/LARC. The reverse was observed in hematopoetic progenitor cells, which responded best to CXCL12/SDF-1. No or little adhesion of MSCs to rICAM-1 was observed under analogous conditions. We next chose to study CXCL12/SDF-1 and CCL19/ELC in more detail on intact endothelial cells. MSCs were flushed through parallel plate flow chambers on Human Umbilical Vein Endothelial Cells (HUVECs) at an initial shear stress of 0.35 or 0.5 dyn/cmE2 for 1 min followed by an elevated shear stress of 5 dyn/cmE2 for further 10 min. We found that rolling of MSCs was increased after pre-treatment of HUVECs with TNF-α but only few spontaneous arrests were obsereved under these conditions. However, when HUVECs were additionally overlaid with CXCL12/SDF-1 or CCL19/ELC, MSCs arrested firmly on the endothelium. Pretreatment of MSCs with function-blocking anti-CD44 or anti-VLA-4 antibodies, or pre-treatment of HUVECs with anti-E-selectin or anti-VCAM-1 antibodies partially (anti-CD44, -E-selectin) or completely (anti-VLA-4, -VCAM-1) suppressed arrest of MSCs. Analysis of adhesion strengthening on HUVECs as well as on immobilized rVCAM-1 confirmed the mediation of shear-resistant binding of MSCs by CXCL12/SDF-1 and CCL19/ELC. Pre-incubation of MSCs with pertussis toxin also suppressed both, sudden arrest and firm adhesion, indicating a role of signalling through G protein coupled receptor via Gαi. Finally, MSCs were able to transmigrate chemokine-coated HUVECs but not untreated. We conclude that MSCs respond to individual chemokines with different efficiencies to induce arrest and firm adhesion before their transendothelial migration. MSCs were found highly responsive to chemokines which physiologically act on lymphocytes to enter secondary lymphoid organs or sites of inflammation. This points to a preference of MSCs to follow lymphocyte egress pathways and reach sites of inflammation to induce immunomodulation and –suppression.


2001 ◽  
Vol 69 (8) ◽  
pp. 5088-5097 ◽  
Author(s):  
Oliver Fuhrmann ◽  
Mardjan Arvand ◽  
Alexander Göhler ◽  
Michael Schmid ◽  
Matthias Krüll ◽  
...  

ABSTRACT The endothelium is a specific target for Bartonella henselae, and endothelial cell infection represents an important step in the pathogenesis of cat scratch disease and bacillary angiomatosis. Mechanisms of Bartonella-endothelial cell interaction as well as signaling pathways involved in target cell activation were analyzed. B. henselae strain Berlin-1, isolated from bacillary angiomatosis lesions of a human immunodeficiency virus-infected patient, potently stimulated human umbilical cord vein endothelial cells (HUVEC), as determined by NF-κB activation and enhanced adhesion molecule expression. These effects were accompanied by increased PMN rolling on and adhesion to infected endothelial cell monolayers, as measured in a parallel-plate flow chamber assay. Monoclonal antibodies against E-selectin significantly reduced PMN rolling and adhesion. In our hands, B. henselae Berlin-1 was substantially more active than the typing strain B. henselae ATCC 49882. E-selectin and ICAM-1 upregulation occurred for up to 9 days, as verified by Northern blotting and cell surface enzyme-linked immunosorbent assay. Induction of adhesion molecules was mediated via NF-κB activation and could be blocked by a specific NF-κB inhibitor. Additional studies indicated that B. henselae-induced effects did not require living bacteria or Bartonella lipopolysaccharides. Exposure of HUVEC to purified B. henselae outer membrane proteins (OMPs), however, reproduced all aspects of endothelial cell activation. In conclusion, B. henselae, the causative agent of cat scratch disease and bacillary angiomatosis, infects and activates endothelial cells. B. henselae OMPs are sufficient to induce NF-κB activation and adhesion molecule expression followed by enhanced rolling and adhesion of leukocytes. These observations identify important new properties of B. henselae, demonstrating its capacity to initiate a cascade of events culminating in a proinflammatory phenotype of infected endothelial cells.


2015 ◽  
Vol 39 (10) ◽  
pp. 1099-1110 ◽  
Author(s):  
Iordanis Pelagiadis ◽  
Eftichia Stiakaki ◽  
Christianna Choulaki ◽  
Maria Kalmanti ◽  
Helen Dimitriou

2004 ◽  
Vol 53 (12) ◽  
pp. 1221-1227 ◽  
Author(s):  
Christine M Litwin ◽  
Joel M Johnson ◽  
Thomas B Martins

Bartonella henselae is a recently recognized pathogenic bacterium associated with cat-scratch disease, bacillary angiomatosis and bacillary peliosis. A recombinant clone expressing an immunoreactive antigen of B. henselae was isolated by screening a genomic DNA cosmid library by Western blotting with sera pooled from patients positive for B. henselae IgG antibodies by indirect immunofluorescence (IFA). The deduced amino acid sequence of the 43.7 kDa encoded protein was found to be 76.3 % identical to the dihydrolipoamide succinyltransferase enzyme (SucB) of Brucella melitensis. SucB has been shown to be an immunogenic protein during infections by Brucella melitensis, Coxiella burnetii and Bartonella vinsonii. The agreement between reactivity with a recombinant SucB fusion protein on immunoblot analysis and the results obtained by IFA was 55 % for IFA-positive sera and 88 % for IFA-negative sera. Cross-reactivity was observed with sera from patients with antibodies against Brucella melitensis, Mycoplasma pneumoniae, Francisella tularensis, Coxiella burnetii and Rickettsia typhi.


2021 ◽  
Author(s):  
Charlotte Sarre ◽  
Rafael Contreras Lopez ◽  
Nitirut Nerpernpisooth ◽  
Christian Barrere ◽  
Sarah Bahraoui ◽  
...  

Abstract Background: Mesenchymal Stromal Cells (MSC) have been widely used for their therapeutic properties in many clinical applications including myocardial infarction. Despite promising preclinical results and evidences of safety and efficacy in phases I/ II, inconsistencies in phase III trials have been reported. In a previous study, we have shown using MSC derived from the bone marrow of PPARβ/δ (Peroxisome proliferator-activated receptors β/δ) knockout mice that the acute cardioprotective properties of MSC during the first hour of reperfusion are PPARβ/δ-dependent but not related to the anti-inflammatory effect of MSC. However, the role of the modulation of PPARβ/δ expression on MSC cardioprotective and anti-apoptotic properties has never been investigated. Objectives: The aim of this study was to investigate the role of PPARβ/δ modulation (inhibition or activation) in MSC therapeutic properties in vitro and ex vivo in an experimental model of myocardial infarction.Methods and results: Naïve MSC and MSC pharmacologically activated or inhibited for PPARβ/δ were challenged with H202. Through specific DNA fragmentation quantification and qRT-PCR experiments, we evidenced in vitro an increased resistance to oxidative stress in MSC pre-treated by the PPARβ/δ agonist GW0742 versus naïve MSC. In addition, PPARβ/δ-priming allowed to reveal the anti-apoptotic effect of MSC on co-cultured cardiomyocytes. When injected during reperfusion in an ex vivo heart model of myocardial infarction, PPARβ/δ-primed MSC at a dose of 3.75x105 MSC/heart provided the same cardioprotective efficiency than 7.5x105 naïve MSC, identified as the optimal dose in our model. These enhanced short-term cardioprotective effects were associated with an increase in both anti-apoptotic effects and the number of MSC detected in the left ventricular wall at 1 hour of reperfusion. By contrast, inhibition of PPARβ/δ before their administration in post-ischemic hearts during reperfusion decreased their cardioprotective effects. Conclusion: Altogether these results revealed that PPARβ/δ-primed MSC exhibit an increased resistance to oxidative stress and enhanced anti-apoptotic properties on cardiac cells in vitro. PPARβ/δ-priming appears as an innovative strategy to enhance the cardioprotective effects of MSC and to decrease the injected doses. These results could be of major interest to improve MSC efficacy for the cardioprotection of injured myocardium in AMI patients.


Stroke ◽  
2018 ◽  
Vol 49 (Suppl_1) ◽  
Author(s):  
Kaavya Giridhar ◽  
Nikunj Satani ◽  
Bing Yang ◽  
Songmi Lee ◽  
Xiaopei Xi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document