U.S. flag

An official website of the United States government

Display Settings:

Items per page

PMC Full-Text Search Results

Items: 15

1.
Figure 9

Figure 9. A subset of biofilm-associated macrophages express arginase-1. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Tissues surrounding biofilm-infected catheters were collected from wild type mice at day 7 following S. aureus infection and subjected to immunofluorescence staining for the macrophage-specific marker Iba-1 (red) and arginase-1 (green) to determine co-localization patterns by confocal microscopy (arrows, 20X magnification). The position of the biofilm-infected catheter is denoted by asterisks. Results are representative of 6 individual animals.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
2.
Figure 7

Figure 7. Macrophage migration towards S. aureus biofilms in vivo is limited. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Tissues surrounding biofilm-infected catheters were collected from wild type mice at day 7 following S. aureus infection and subjected to immunofluorescence staining with Iba-1 to identify macrophages (red) and DAPI to demarcate nuclei (blue) by confocal microscopy (20X magnification). Asterisks indicate the location of infected catheters and the border between biofilm and surrounding host tissue is denoted by dashed lines. Results are representative of 6 individual animals.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
3.
Figure 13

Figure 13. Macrophages exposed to mature S. aureus biofilm exhibit a dystrophic morphology. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Primary macrophages labeled with CellTracker orange (CTO; yellow-orange) were exposed to 4 or 6 day-old USA300 LAC-GFP biofilms (green) for a 24 h period, whereupon visualization of macrophage morphology was evaluated by confocal microscopy (63X magnification, 1 μm slice). Macrophages infiltrating immature (4 day-old) biofilms retained a typical rounded morphology, whereas macrophages infiltrating mature biofilms (6 day-old) often exhibited a dystrophic ghost-like morphology with minimal retention of CTO (arrows). Results are representative of six independent experiments.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
4.
Figure 14

Figure 14. Macrophage viability is dictated by invasion into the biofilm proper. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

(A) 4 day-old USA300 LAC-GFP biofilms (average height ~ 33 μm) were incubated with (B) primary macrophages labeled with CellTracker orange (yellow-orange) for a 24 h period, whereupon macrophage viability was determined by uptake of the live/dead stain TOTO3 (purple; C). The majority of dead (TOTO3+) macrophages are within 34 μm of the biofilm proper (p < 0.05) and a composite image of all staining is shown (D). Bottom; significant differences between the percentages of TOTO3+ macrophages based on location within the biofilm proper are denoted with an asterisk (*, p < 0.05; n = 218 cells).

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
5.
Figure 15

Figure 15. S. aureus biofilms skew macrophage responses to favor bacterial persistence. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Despite the presence of TLR2 and TLR9 ligands within the biofilm (lipoproteins/PGN and eDNA, respectively), these traditional pattern recognition receptors to not impart macrophage responsiveness or bacterial clearance during biofilm infection (depicted by red “x” marks). Likewise, the expression of iNOS is reduced following biofilm formation concomitant with an increase in arginase-1. Collectively, these changes shift the immune response away from a bactericidal pathway and likely contribute to biofilm persistence.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
6.
Figure 10

Figure 10. S. aureus biofilms skew macrophage gene expression towards an alternatively activated (M2) phenotype. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Bone marrow-derived macrophages from GFP Tg mice were incubated with 6 day-old biofilms or planktonic bacteria for 2 h, whereupon viable macrophages were purified by FACS and RNA immediately isolated for qRT-PCR analysis. Gene expression levels in macrophages exposed to S. aureus biofilms were calculated after normalizing signals against GAPDH and are presented as the fold-change relative to macrophages incubated with planktonic bacteria. Results represent the mean ± SEM of four independent experiments (*, p < 0.05; **, p < 0.001).

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
7.
Figure 3

Figure 3. Visualization of S. aureus biofilm infection using IVIS. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Wild type (WT), TLR2 knockout (KO), and TLR9 KO mice were infected with 5 × 105 CFU of USA300 LAC::lux either in the lumen of surgically implanted catheters to establish biofilm infection or subcutaneously in the absence of any indwelling device. At the indicated time points post-infection, mice were subjected to IVIS imaging to visualize the extent of biofilm or subcutaneous infection. Images are presented from one mouse per group that was imaged at all time points to map the progression of biofilm infection.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
8.
Figure 12

Figure 12. Macrophages recovered from S. aureus biofilm infections harbor viable intracellular bacteria. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Viable macrophages (F4/80+, 7-AAD) were recovered from tissues surrounding biofilm infections at day 7 post-infection by FACS, whereupon gentamicin protection assays were performed to evaluate the presence of viable intracellular bacteria. Controls included sorted macrophages without any antibiotic treatment (to assess extra- and intracellular bacteria) and macrophages treated with rifampicin, which is capable of penetrating the eukaryotic cell membrane. Results are expressed as the number of viable S. aureus (CFU per 103 biofilm-associated macrophages) and represent the mean ± SEM of five independent experiments.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
9.
Figure 5

Figure 5. S. aureus biofilm attenuates inflammatory mediator expression compared to the wound healing response elicited following sterile catheter implantation. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Tissues surrounding S. aureus infected or sterile catheters from wild type mice were collected and homogenized at the indicated time points to obtain supernatants to quantitate differences in TNF-α (A), IL-1β (B), CXCL2 (C), or CCL2 (D) expression by ELISA. Results were normalized to the amount of total protein recovered to correct for differences in tissue sampling size. Significant differences in inflammatory mediator expression between biofilm infected versus sterile catheters are denoted by asterisks (*, p < 0.05). Results are presented from individual animals combined from three independent experiments.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
10.
Figure 2

Figure 2. Evidence of catheter-associated biofilm growth in vivo. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Catheters were isolated from wild type mice at day 10 following S. aureus infection and processed for SEM analysis. The smooth surface at the periphery of the images in A and B represents the catheter with biofilm visible on the internal face. (A), 200X magnification demonstrating the irregular undulating pattern of the biofilm surface (arrows indicate tower structures); (B), higher magnification of a tower from the image shown in (A) revealing a predominantly hollow interior with numerous cocci at the margins (800X magnification). The image has been pseudocolored to highlight S. aureus (gold) and presumably matrix material (green), which likely aggregated during the SEM dehydration process, from the remaining biofilm structure (grey; 800X magnification). The smooth surface at the upper right represents the catheter (salmon) with biofilm visible on the internal face. (C), 3000X magnification of a S. aureus cluster within the tower depicted in (B).

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
11.
Figure 1

Figure 1. Characterization of S. aureus localization during biofilm development. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Catheters and surrounding tissues were isolated from wild type mice at day 10 following S. aureus infection and subjected to Gram-staining (A and B) or SEM (C). There was evidence of bacterial growth (purple) within the catheter lumen (A) as well as host tissue immediately surrounding the catheter (B, arrow). Insets in panels A & B depict 10X magnification with red rectangles indicating the area enlarged at 40X magnification and asterisks denoting the original location of infected catheters that were non-adherent to glass slides. A 400X magnification SEM image (C) has been pseudocolored to highlight S. aureus biofilm (blue) at the interface between the catheter (grey) and surrounding host tissue (purple). Host cells located at a distance from the biofilm are indicated by arrows.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
12.
Figure 11

Figure 11. Macrophages actively phagocytize planktonic S. aureus but not biofilm-associated bacteria. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

(A) Primary macrophages labeled with CellTracker orange (orange) were exposed to 4 day-old USA300 LAC-GFP biofilms or planktonic bacteria (green) for a 24 h period, whereupon visualization of intracellular bacteria was evaluated by confocal microscopy (63X magnification, 1 μm slice). Biofilm-associated macrophages exhibited little evidence of phagocytosis, whereas intracellular bacteria were readily detectable in macrophages incubated with planktonic organisms. Results are representative of six independent experiments. In (B), four and six day-old USA300 LAC-GFP static biofilms were mechanically disrupted by triturating and incubated with macrophages for 1 h. Planktonic bacteria were included as a positive control. Fluorescent microscopy of cytospin preparations shows the ability of macrophages to phagocytize planktonic (left) as well as disassociated bacteria from four day-(center) and six day-old (right) biofilms. Results are representative of three independent experiments.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
13.
Figure 6

Figure 6. S. aureus biofilms elicit exaggerated macrophage infiltrates compared to abscesses. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Wild type mice were infected with 5 × 105 CFU of USA300 LAC::lux either in the lumen of surgically implanted catheters or subcutaneously in the absence of any indwelling device to establish biofilm and abscess infections, respectively. Animals were sacrificed at days 3, 7, or 14 following S. aureus exposure, whereupon tissues surrounding infected catheters or s.c. injection sites were collected to quantitate macrophage infiltrates by FACS. Results are expressed as the percent of F4/80+ macrophages after correction for isotype control staining and represent the mean ± SEM of three independent experiments. Significant differences in macrophage infiltration into biofilm versus abscess infections are denoted by asterisks (*, p < 0.01).

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
14.
Figure 8

Figure 8. S. aureus biofilms repress iNOS and augment arginase-1 expression. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Wild type mice were infected with 5 × 105 CFU of USA300 LAC::lux either in the lumen of surgically implanted catheters or subcutaneously in the absence of any indwelling device to establish biofilm and abscess infections, respectively. Animals were sacrificed at day 7 following S. aureus exposure, whereupon tissues surrounding infected catheters or s.c. injection sites were collected and subjected to immunofluorescence staining with arginase-1 (red) and DAPI to identify nuclei (blue) by confocal microscopy (20X magnification). Stars represent the location of material remaining from the catheter lumen and the diamonds denote the necrotic abscess core. The border between biofilm and surrounding host tissue is depicted by arrows. Results are representative of 6 individual animals.

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.
15.
Figure 4

Figure 4. Infection with an equivalent S. aureus dose leads to the establishment of catheter-related biofilm infection but rapid clearance from subcutaneous sites. From: Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo.

Wild type (WT), TLR2 knockout (KO), and TLR9 KO mice were infected with 5 × 105 CFU of USA300 LAC::lux either in the lumen of surgically implanted catheters (cath) to establish biofilm infection or subcutaneously (sc) in the absence of any indwelling device. Animals were sacrificed at the indicated days following S. aureus infection, whereupon host tissues surrounding infected catheters or s.c. injection sites were homogenized to quantitate bacterial burdens. Results are expressed as the number of CFU per mg host tissue to correct for differences in tissue sampling size. Significant differences in bacterial burdens between biofilm-infected versus s.c. injected mice are denoted by asterisks (*, p < 0.001).

Lance R. Thurlow, et al. J Immunol. ;186(11):6585-6596.

Display Settings:

Items per page

Supplemental Content

Recent activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...
Support Center