Literature
首页医源资料库在线期刊美国病理学杂志2007年第169卷第10期

Minocycline Modulates Neuroinflammation Independently of Its Antimicrobial Activity in Staphylococcus aureus-Induced Brain Abscess

来源:《美国病理学杂志》
摘要:【摘要】Minocyclineexertsbeneficialimmunemodulatoryeffectsinseveralnoninfectiousneurodegenerativediseasemodels。Minocyclinewasalsocapableofprotectingthebrainparenchymafromnecroticdamageasevidentbysignificantlysmallerabscessesinminocycline-treatedmice。Minocy......

点击显示 收起

【摘要】  Minocycline exerts beneficial immune modulatory effects in several noninfectious neurodegenerative disease models; however, its potential to influence the host immune response during central nervous system bacterial infections, such as brain abscess, has not yet been investigated. Using a minocycline-resistant strain of Staphylococcus aureus to dissect the antibiotic??s bacteriostatic versus immune modulatory effects in a mouse experimental brain abscess model, we found that minocycline significantly reduced mortality rates within the first 24 hours following bacterial exposure. This protection was associated with a transient decrease in the expression of several proinflammatory mediators, including interleukin-1ß and CCL2 (MCP-1). Minocycline was also capable of protecting the brain parenchyma from necrotic damage as evident by significantly smaller abscesses in minocycline-treated mice. In addition, minocycline exerted anti-inflammatory effects when administered as late as 3 days following S. aureus infection, which correlated with a significant decrease in brain abscess size. Finally, minocycline was capable of partially attenuating S. aureus-dependent microglial and astrocyte activation. Therefore, minocycline may afford additional therapeutic benefits extending beyond its antimicrobial activity for the treatment of central nervous system infectious diseases typified by a pathogenic inflammatory component through its ability to balance beneficial versus detrimental inflammation.
--------------------------------------------------------------------------------
Staphylococcus aureus is a frequently encountered organism among patients with brain abscess, an entity that represents a significant medical problem despite recent advances in diagnosis and therapy.1-3 Brain abscess has been reported to account for approximately 1 in 10,000 hospital admissions in the United States.2 The most common sources of brain abscess are direct or indirect cranial infection arising from the paranasal sinuses, middle ear, and teeth. Other routes include seeding of the brain from distant sites of systemic infection (ie, endocarditis or lung abscess) or penetrating trauma to the head.1,2 Even with recent advances made in imaging techniques, the clinical diagnosis of brain abscess is often delayed, because patients may initially present with nonspecific clinical signs. Another factor complicating diagnosis is the tendency of brain abscesses to appear similar to necrotic tumors on computed tomography and magnetic resonance imaging scans. If not detected early, an abscess has the potential to rupture into the ventricular space, a serious complication with an 80% mortality rate. The current treatment for brain abscess includes surgical drainage to relieve pressure and long-term (6 to 8 weeks) systemic antimicrobial therapy.4 Treatment can be complicated by the emergence of multidrug-resistant strains of S. aureus and decreased central nervous system (CNS) penetration of alternative drugs such as vancomycin.5,6 Patients surviving brain abscesses may experience any number of long-term complications including seizures, loss of mental acuity, and focal neurological defects that are lesion site-dependent.1,2 We have established a mouse experimental brain abscess model using S. aureus in which lesion sites are greatly exaggerated compared with the relatively localized area of initial bacterial growth.3,7-9 This phenomenon is also observed in human disease, where lesions can encompass a large portion of brain tissue, often spreading well beyond the initial focus of infection. Collectively, these sequelae are reminiscent of an overactive immune response. The continual production of bactericidal compounds by both resident glia and peripheral immune cells as well as amplification of cytokine networks and protease production can contribute to the loss of surrounding uninfected tissue. We propose that a balance exists between sufficient and overcompensatory responses to S. aureus in the CNS, which dictates the extent of parenchymal damage during brain abscess development. Indeed, we have found that S. aureus leads to the immediate and sustained expression of numerous proinflammatory mediators in the brain, indicative of a potent innate immune response.7,8,10,11
Brain abscesses develop in response to a parenchymal infection with pyogenic bacteria, beginning as a localized area of cerebritis and evolving into a suppurative lesion surrounded by a well-vascularized fibrotic capsule.1-3 Brain abscesses are typified by extensive edema and tissue necrosis and tend to localize at white-gray matter junctions where microcirculatory flow is poor.12,13 In addition to the sequential progression from cerebritis to necrosis during brain abscess evolution, the activation of resident glial cells and influx of peripheral leukocytes demonstrate temporal patterns.3,14,15 Specifically, microglial and astrocyte activation is evident immediately following the entry of bacteria into the CNS parenchyma and persists throughout abscess evolution.11,16 Neutrophils are the initial leukocyte subset to infiltrate developing abscesses and are observed as early as 12 hours following bacterial exposure.7,11 Macrophages and T cells are associated with lesions as they progress, with infiltrates generally more pronounced around days 3 and 7, respectively.11,14 Beginning at 7 to 10 days postinfection, a highly vascularized fibrotic wall forms around the necrotic milieu, effectively forming a barrier to contain the infection.11,17,18 It is important to note that the kinetics of cellular activation, influx, and bordering functions represent general time frames, and there is probably overlap between each of these processes during brain abscess evolution.
Although necessary for pathogen containment, excessive inflammation during the early phases of brain abscess development may have deleterious consequences on disease outcome through the induction of exaggerated edematous and/or necrotic responses.3,12,13 Similar detrimental effects have been implicated in the pathophysiology of bacterial meningitis, where the dysregulated production of free radicals, proinflammatory cytokines, and edema lead to significant neuron damage and associated mortality.19,20 Therefore, attenuating glial and peripheral immune cell activation without compromising bacterial containment may result in less damage to normal brain tissue and improvements in cognitive and neurological functions.
One attractive candidate to achieve this balance during the course of brain abscess development is minocycline. Minocycline is a second-generation tetracycline that effectively crosses the blood-brain barrier due to its small (495 d) and lipophilic nature.21-24 In susceptible bacterial strains, minocycline binds to the h34 ribosomal subunit, resulting in a conformational change that effectively interferes with protein synthesis.25,26 Minocycline has been shown to exert neuroprotective effects distinct from its bacteriostatic activity in animal models of cerebral ischemia,27-30 Parkinson??s and Huntington??s disease,31-34 Alzheimer??s disease,35,36 Down??s syndrome,37 amyotrophic lateral sclerosis,38,39 spinal cord injury,40-42 and experimental autoimmune encephalomyelitis, an animal model for multiple sclerosis.43-45 Minocycline is thought to modulate these diseases, in part, by inhibiting the production of potentially cytotoxic molecules by activated microglia.46 For example, minocycline has been shown to inhibit microglial production of NO, interleukin (IL)-1ß, tumor necrosis factor (TNF)-, and IL-6, which can impact neuron survival.47-49 In addition, minocycline may also protect CNS cells from apoptosis through inhibition of caspase-3 activity.31,41,42 Minocycline has also been shown to down-regulate signal transduction pathways such as nuclear factor-B and p38 mitogen-activated protein kinase,32,48,50,51 both of which play pivotal roles in controlling the expression of numerous proinflammatory genes. Tetracyclines have also been shown to inhibit production of the proinflammatory molecules TNF-,52,53 NO,54 and matrix metalloproteinases44,55 by leukocytes. However, it is also important to acknowledge the fact that other investigators have reported that minocycline has no impact or, in some cases, exacerbates neuronal injury in several CNS disease models,23,56-60 highlighting the need for further studies in this area to identify specific conditions in which minocycline therapy may exert beneficial effects.
Although a few recent reports have documented favorable effects of minocycline in various viral and parasitic infections,61-64 the consequences of this antimicrobial on modulating inflammatory responses in the context of CNS bacterial infections, such as brain abscess, have not yet been examined. The relative bacteriostatic versus immune modulatory effects of minocycline have been difficult to differentiate; however, in this study we have devised a strategy to discern between these two possibilities by engineering a S. aureus strain that is resistant to the bacteriostatic effects of minocycline. Our findings indicate that an overactive host response to infection can lead to the heightened expression of proinflammatory mediators and subsequent mortality during the initial stages of brain abscess, which can be attenuated by minocycline independently of its antibacterial activity. In addition, minocycline was capable of reducing ongoing inflammation, as both inflammatory mediator expression and brain abscess size were attenuated when minocycline treatment was initiated 3 days following S. aureus infection. Collectively, these results suggest that if not tightly regulated, the inflammatory response that ensues during brain abscess development can exert deleterious effects. Therefore, minocycline may represent an excellent candidate for brain abscess therapy through its ability to attenuate chronic inflammation and reduce the extent of parenchymal tissue damage that is characteristic of infection.

【关键词】  minocycline modulates neuroinflammation independently antimicrobial activity staphylococcus aureus-induced



Materials and Methods


Bacterial Strains and Reagents


The minocycline-sensitive S. aureus strain RN6390 was used as previously described.7-10 A minocycline-resistant isogenic strain of RN6390 was constructed with a tetracycline resistance cassette (tetM), which also confers resistance to minocycline, stably inserted into the bacterial chromosome using the integration vector pCL84, which preferentially inserts into the S. aureus lipase gene.65 Chromosomal insertion of the tetM resistance cassette was confirmed by sequence analysis of chromosomal polymerase chain reaction (PCR) products and Southern blots. To determine whether the insertion of the tetM cassette led to any overt alterations in the S. aureustetM mutant compared with the parental strain RN6390, pulse field gel electrophoresis and sodium dodecyl sulfate-polyacrylamide gel electrophoresis of whole cell lysates and secreted proteins from several independent bacterial colonies were performed.


Minocycline-HCl was purchased from Sigma Chemical Co. (St. Louis, MO), and a 10 mg/ml stock solution was prepared in endotoxin-free H2O and stored in aliquots at C20??C until use. For animal injections, minocycline working stocks were prepared fresh daily in sterile endotoxin-free phosphate-buffered saline (pH 7.4).


Generation of Experimental Brain Abscesses


Brain abscesses were induced in 6- to 8-week-old C57BL/6 mice using a stereotaxic approach as previously described.8,9 For all studies, an equal number of age-matched males and females were used as previous work has established that both genders exhibit qualitatively similar inflammatory profiles following bacterial challenge.7-10 The animal use protocol has been approved by the University of Arkansas for Medical Sciences Institutional Animal Care and Use Committee and is in accord with the National Institutes of Health guidelines for the use of rodents.


For evaluating the potential therapeutic effects of minocycline during brain abscess development, mice were subjected to either one of two treatment paradigms. In the first, the effects of minocycline when administered before S. aureus exposure were examined. For these experiments, mice received once daily intraperitoneal (i.p.) injections of minocycline beginning 1 day before S. aureus infection and continuing until study termination. In the second treatment paradigm, minocycline was evaluated for its ability to modulate ongoing CNS inflammation. For these studies, minocycline was administered 3 days following S. aureus infection, a time point when bacterial burdens have nearly peaked.7,11 After this initial treatment, minocycline injections continued daily until the termination of the study. Minocycline was initially examined at three concentrations (2, 10, and 50 mg/kg/day); however, in subsequent studies one dose was selected (50 mg/kg/day), which was determined to exert optimal effects on the ensuing host CNS antibacterial response. For all experiments, one group of animals received minocycline treatment alone (50 mg/kg/day) to assess any effects of the drug independently of S. aureus infection. Animals receiving 50 mg/kg/day minocycline alone did not demonstrate any overt adverse effects to the drug (ie, no significant alterations in body weight). The doses of minocycline tested are representative of concentrations used in previously published reports of disparate CNS neurodegenerative models,46,66 and although they represent concentrations well above those used in humans, this is offset by the extremely short half-life of minocycline in mice (2 to 3 hours) versus humans (15 to 18 hours).22,66,67


Simultaneous Collection of RNA, Protein, and Quantitation of Viable Bacteria from Brain Abscesses


Brain abscesses were visualized by the stab wound created during injections, sectioned within 2 to 3 mm on all sides, and homogenized in PBS supplemented with protease and RNase inhibitors. Subsequently, RNA, protein, and bacterial titers were obtained as previously described.9 It is not feasible to dissect away necrotic from viable brain tissue while collecting samples for analysis, because these microscopic borders are not easily discernable by the naked eye. Although we do not specify between viable and necrotic tissue when collecting brain abscesses, we propose that this does not introduce artifacts in the results obtained. For example, our results demonstrate that minocycline treatment reduced brain abscess severity as typified by smaller lesions, which translates into a greater amount of surviving tissue. Yet in this case, the expression of several proinflammatory mediators was diminished. If necrotic tissue greatly impacts the results obtained, we would have expected a reduction in cytokine production in brain abscesses of vehicle-treated animals, which was not observed.


Quantitation of Brain Abscess Size


At the indicated time points postinfection, minocycline- and vehicle-treated mice were perfused transcardially with PBS to eliminate leukocytes from the vasculature, whereupon brains were removed and immediately flash-frozen on dry ice. Before cryostat sectioning, brain tissues were embedded in optimal cutting temperature medium, and serial 10-µm sections were made throughout the entire lesioned tissue and stained with hematoxylin and eosin to demarcate abscess margins. Evaluation of serial sections throughout the affected brain parenchyma ensured that the largest abscess cross-sectional area would be identified for comparisons of lesion size. Abscess area (reported as square millimeters) was calculated using the MetaMorph image analysis program (Universal Imaging Corporation, Downingtown, PA).


Evaluation of Tissue Viability Using Nitro Blue Tetrazolium (NBT) Staining


To better demarcate the border between living and nonviable tissue during the early stages of brain abscess development, NBT staining of fresh frozen tissue sections was performed.68 The intensity of staining is dictated by the amount of mitochondrial diaphorase enzymes that were present in viable cells at the time of tissue collection, which metabolize the conversion of NBT into a blue formazan product. The loss of staining is indicative of the degree of cell damage/injury. In brief, 10-µm cryostat sections were fixed for 10 minutes in ice-cold acetone and air-dried. Tissues were subsequently incubated for 40 minutes at room temperature with 200 µl/slide NBT development solution (0.2 mol/L Tris-HCl, pH 7.1, containing 0.25 mg/ml NBT, 2 mg/ml NADH, both from Sigma Chemical Co., and 1 mg/ml MgCl2). To terminate the reaction, slides were rinsed in double distilled water, dehydrated, and coverslipped using Permount mounting medium.


Immunofluorescence Staining of Astrocytes and Microglia/Macrophages


The effects of minocycline on astrocyte and microglia/macrophage activation during brain abscess development were evaluated by immunofluorescence staining using glial fibrillary acidic protein (GFAP) and F4/80, respectively, and confocal microscopy. Brain abscess tissues were collected at the indicated time points following minocycline treatment, and 10-µm cryostat sections were mounted onto SuperFrost Plus slides (Fisher Scientific, Houston, TX), air-dried, and stored at C80??C until use. To initiate staining, slides were equilibrated at room temperature for 15 minutes and fixed in ice-cold methanol. Following numerous rinses in PBS, tissues were incubated with PBS/10% normal donkey serum to minimize nonspecific staining. Brain abscess tissues were incubated with either primary anti-rat GFAP (Zymed, South San Francisco, CA) or F4/80 (Serotec, Raleigh, NC) antibodies overnight at 4??C in a humidified chamber. Following numerous rinses in PBS, tissues were incubated with either a donkey anti-rat IgG fluorescein isothiocyanate conjugate (for GFAP) or biotinylated donkey anti-rat IgG antibody (for F4/80; both from Jackson Immunoresearch, West Grove, PA) for 1 hour at room temperature. F4/80 expression was visualized by the addition of a streptavidin-Alexa Fluor 568 conjugate (Molecular Probes, Eugene, OR). On completion of the staining protocol, slides were coverslipped using the Prolong antifade reagent (Molecular Probes) and sealed using nail polish. Slides were imaged using a Zeiss laser scanning confocal microscope (LSM 510; Carl Zeiss Microimaging Inc., Thornwood, NY), and fluorescein isothiocyanate was excited with a 488-nm argon laser to visualize GFAP immunoreactivity, and Alexa Fluor 568 was excited with a 561-nm diode-pumped solid-state laser to demonstrate F4/80 expression, with images collected using the appropriate emissions. The confocal pinhole was set to obtain an optical section thickness of 1.6 µm. Specific staining of antibodies was confirmed by the absence of fluorescence signal following incubation of brain abscess tissues with secondary antibodies alone (data not shown).


Preparation of Primary Microglia and Astrocyte Cultures


Primary microglia and astrocytes were isolated from neonatal C57BL/6 mice (postnatal days 2 to 4) as previously described.69 The purity of microglia and astrocyte cultures, as determined by immunohistochemical staining using antibodies against CD11b (BD PharMingen, San Jose, CA) and GFAP (Dako Corp., Carpinteria, CA), was routinely greater than 95 and 90%, respectively.


To investigate the effects of minocycline on glial activation, primary microglia and astrocytes were seeded into 96-well plates at 2 x 105 or 1 x 105 cells/well, respectively, and incubated overnight. The following day, cells were treated with various doses of minocycline for 1 hour before stimulation with either heat-inactivated S. aureus or 10 µg/ml peptidoglycan (PGN; InvivoGen, San Diego, CA). Cell-conditioned supernatants were collected at 24 hours following bacterial exposure, whereupon NO and cytokine/chemokine production was evaluated as described below. The doses of bacterial stimuli used were based on our previous studies that established optimal cytokine responses without any evidence of toxicity.70,71


Nitrite Assay


Nitrite (a stable reaction product of NO) levels were determined by adding 50 µl of astrocyte-conditioned culture medium with 50 µl of Griess reagent (0.1% naphthylethylenediamine dihydrochloride, 1% sulfanilamide, and 2.5% phosphoric acid; all from Sigma Chemical Co.) in a 96-well plate. The absorbance at 550 nm was measured on a plate reader (Spectra Max 190; Molecular Devices, Sunnyvale, CA), and nitrite concentrations were calculated using a standard curve with sodium nitrite (NaNO2; Sigma Chemical Co.; level of sensitivity 0.4 µmol/L).


Cell Viability Assays


The effects of minocycline on microglia and astrocyte viability were evaluated using a standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) (Sigma) assay based on the mitochondrial conversion of MTT into formazan crystals as previously described.72


Protein Extraction and Western Blotting


Protein extracts were prepared from primary microglia or astrocytes as previously described.72,73 The effects of minocycline on glial Toll-like receptor 2 (TLR2) protein expression were evaluated by Western blot analysis. Blots were probed using a goat anti-mouse TLR2 antibody (R&D Systems, Minneapolis, MN) followed by a donkey anti-goat IgGChorseradish peroxidase secondary antibody (Jackson Immunoresearch). Blots were developed using the Immobilon Western substrate (Millipore, Billerica, MA) and visualized by exposure to X-ray film. Subsequently, membranes were stripped and re-probed with a rabbit anti-actin polyclonal antibody (Sigma Chemical Co.) for normalization.


Quantitative Real-Time RT-PCR (qRT-PCR)


Total RNA from brain abscesses was isolated using the TRIzol reagent and treated with DNase1 (both from Invitrogen) before use in qRT-PCR studies. The experimental procedure was performed as previously described.73 IL-1ß, TNF-, and glyceraldehyde-3-phosphate dehydrogenase primers and TAMRA TaqMan probes were designed69,74 and synthesized by Applied Biosystems (ABI, Foster City, CA). An ABI Assays-on-Demand TaqMan kit was used to examine CCL2 expression.


Enzyme-Linked Immunosorbent Assay (ELISA)


Murine TNF-, IL-1ß, and CCL2 (OptiEIA; BD PharMingen) and CXCL2 (MIP-2, DuoSet; R&D Systems) were quantified in brain abscess homogenates using commercial ELISA kits. Results were normalized to the amount of total protein extracted from tissues to correct for differences in sampling size as previously described.8,11


Statistics


Significant differences between experimental groups were determined using the unpaired Student??s t-test at the 95% confidence interval with Sigma Stat (SPSS Science, Chicago, IL). This analysis was determined to be most appropriate since, although we are evaluating changes in proinflammatory mediator expression over time, repeated measurements are not made on the same animal (mice are sacrificed to collect abscess homogenates at each time point), precluding analysis of variance and post hoc analysis of the data.


In this study, we performed a minimum of three independent replicates of each study to confirm the results obtained. The reporting of our results as representative of "x" number of independent experiments was required, because it is difficult to achieve identical bacterial burdens in mice between independent brain abscess studies with distinct bacterial preparations. As a result, the absolute concentrations of the various proinflammatory mediators detected within brain abscesses of minocycline-treated and control mice differed between individual experiments; however, the trends were consistent. This required us to report results from a single experiment where at least four or five mice per time point per group were analyzed and statistical analysis conducted.


Results


Minocycline Inhibits S. aureus Replication in Brain Abscesses Concomitant with Attenuated Proinflammatory Mediator Expression


Initial experiments established that the S. aureus strain used in our experimental model, RN6390, was sensitive to the antibiotic effects of minocycline (data not shown). To examine whether minocycline affected the acute phase of brain abscess development in this susceptible strain, the antibiotic was administered daily to animals beginning one day before intracerebral S. aureus infection and continuing until termination of the study. Minocycline treatment led to a dose-dependent reduction in bacterial burdens (Figure 1) that coincided with significant decreases in the expression of several proinflammatory mediators that we have previously determined to be pivotal during the acute stage of brain abscess including IL-1ß, TNF-, and CCL2 (Figure 2) .8 The effects of minocycline on proinflammatory mediator expression were probably related to its ability to reduce bacterial burdens and/or modulate signaling pathways important for the induction of cytokines/chemokines including nuclear factor-B or p38 mitogen-activated protein kinase.47,51


Figure 1. Minocycline significantly reduces bacterial burdens in experimental brain abscesses. Mice (nine/group) received i.p. injections of PBS or minocycline (50, 10, or 2 mg/kg/day) 1 day before intracerebral injections with the minocycline-sensitive S. aureus strain RN6390 and daily until the experiment was terminated. Animals were sacrificed at days 1 (five/group) and 3 (four/group) following S. aureus exposure for quantitation of viable bacteria within abscesses. Significant differences between infected PBS- and minocycline-treated animals are denoted with asterisks (*P < 0.05, **P < 0.001). Results are representative of five independent experiments.


Figure 2. Minocycline attenuates proinflammatory cytokine and chemokine expression in experimental brain abscesses. Mice (nine/group) received i.p. injections of PBS or minocycline (50, 10, or 2 mg/kg/day) 1 day before intracerebral injections with the minocycline-sensitive S. aureus strain RN6390 and daily until the experiment was terminated. Abscess homogenates were prepared at days 1 (five/group) and 3 (four/group) following S. aureus exposure for quantitation of IL-1ß (A), TNF- (B), and CCL2 (C) expression by ELISA. Cytokine/chemokine concentrations were normalized to the amount of total protein recovered to correct for differences in tissue sampling size. Significant differences between infected PBS- and minocycline-treated animals are denoted with asterisks (*P < 0.05, **P < 0.001). Results are representative of five independent experiments.


Minocycline Exhibits Anti-Inflammatory Effects Independently of Its Bacteriostatic Activity


Because we were interested in evaluating the potential anti-inflammatory effects of minocycline in the brain abscess model independently of its antibiotic properties, we generated an isogenic minocycline-resistant strain of S. aureus RN6390 (referred to hereafter as S. aureustetM). It was important to create a minocycline mutant isogenic to the parental strain RN6390 rather than use an unrelated S. aureus isolate that is naturally resistant to minocycline, because we have extensively characterized the host immune response to S. aureus RN6390 in the brain abscess model.7-10,16 To determine whether the insertion of the minocycline resistance cassette tetM resulted in any dramatic changes in phenotype compared with the parental strain RN6390, pulse field gel electrophoresis and sodium dodecyl sulfate-polyacrylamide gel electrophoresis of whole cell lysates and secreted proteins from several independent clones of both the S. aureustetM mutant and parental strains were performed. Pulse field gel electrophoresis revealed the loss of a larger SmaI fragment in the S. aureustetM mutant with the concomitant appearance of two smaller fragments (Figure 3A) . This finding can be explained by the fact that the pCL84 vector used to insert the tetM gene into the bacterial chromosome contains SmaI sites. Evaluation of whole cell lysates and secreted proteins from both the parental and S. aureustetM strains in vitro revealed that protein expression was nearly identical between the two strains, except for the presence of a faint unique band in supernatants from the latter (Figure 3B) . Collectively, these results indicate that the introduction of the tetM cassette into the bacterial chromosome did not lead to dramatic alterations in protein expression. In vitro sensitivity of the S. aureustetM strain to minocycline was evaluated, and the minimum inhibitory concentration neared 5 µg/ml (data not shown), in close agreement with the level of resistance previously reported for the tetM gene.75


Figure 3. Characterization of the S. aureustetM strain. A: Pulse field gel electrophoresis (PFGE) of the parental S. aureus strain RN6390 and its isogenic tetM mutants. Chromosomal DNAs from three individual parental and mutant isolates were digested with SmaI and subjected to PFGE. Because the vector pCL84 that was used to insert the tetM gene into the lipase gene of the bacterial chromosome contains SmaI sites, this resulted in a loss of a larger fragment (solid arrow) and the generation of two smaller fragments (blocked arrows). B: Coomassie blue-stained gels depicting the array of proteins in bacterial cell lysates and conditioned supernatants. Overall, the results demonstrate that the in vitro protein expression profiles are similar between the parental RN6390 and S. aureustetM strains except for the presence of a faint protein band in the latter (arrow). The data presented were derived from separate colonies of parental or mutant strains that were propagated in culture independently.


To determine whether minocycline exhibited any immune modulatory effects independently of its antimicrobial activity, mice were treated with various doses of minocycline beginning 1 day before S. aureustetM infection. Minocycline treatment unexpectedly afforded a significant dose-dependent protection from mortality within the first day following S. aureustetM exposure (Figure 4A) . Importantly, these effects were irrespective of the bacteriostatic properties of minocycline, because bacterial burdens between minocycline- and vehicle-treated mice were not significantly different (Figure 4B) .


Figure 4. Minocycline exerts protective effects in brain abscesses independently of its antimicrobial activity. Mice received i.p. injections of PBS or minocycline (50, 10, or 2 mg/kg/day) one day before and on the day of intracerebral injection with a minocycline-resistant S. aureus strain (S. aureustetM). Differences in mortality rates (A) and bacterial burdens (B) between infected minocycline- and vehicle-treated mice are shown at 24 hours postinfection. Mortality rates represent the mean ?? SEM of four independent experiments (n = 16 to 24 mice per treatment group), whereas bacterial titer results are representative of four independent experiments (mean ?? SD from one individual experiment is presented; n = 4 or 5 mice per group). Significant differences between infected PBS- and minocycline-treated animals are denoted with asterisks (*P < 0.05, **P < 0.001).


Minocycline Transiently Attenuates Proinflammatory Mediator Expression in Acute Brain Abscesses


The improved survival rates of minocycline-treated mice were expected to coincide with reduced levels of select proinflammatory molecules. However, no alterations in IL-1ß, TNF-, CXCL2, or CCL2 expression were observed in minocycline-treated mice at 24 hours following infection (data not shown). Based on its relatively short half-life in the mouse, we considered the possibility that minocycline induced transient changes in inflammatory mediator expression that returned to baseline once therapeutic antibiotic levels had subsided because of the continued presence of high bacterial burdens in the CNS parenchyma. Therefore, we examined the expression of several proinflammatory mediators immediately following S. aureustetM infection (ie, 3, 6, and 12 hours). It is important to note that minocycline was administered immediately before bacteria on the day of infection, such that the amount of drug in the CNS was expected to remain at significant levels, at least during the initial time points examined. Because this analysis occurred at such early intervals postinfection, changes in proinflammatory mediator expression were examined by qRT-PCR, because protein levels had not yet accumulated to concentrations detectable by traditional ELISAs. The expression of several proinflammatory cytokines/chemokines, including IL-1ß and CCL2, was significantly decreased by minocycline in a dose-dependent manner at 3 and 6 hours following S. aureustetM infection (Figure 5, A and B) . However, these effects were transient, with proinflammatory mediator expression returning to levels observed in vehicle-treated animals within 12 hours postinfection, in agreement with the extremely short half-life of minocycline in mice. Importantly, the observed decreases in proinflammatory mediator expression were independent of the antibiotic activity of minocycline, because pathogen burdens were not statistically different between the various treatment groups (Figure 5C) .


Figure 5. Minocycline attenuates the early CNS cytokine/chemokine response independently of its bacteriostatic effects. Mice (n = 4/group) received i.p. injections of PBS or minocycline (50, 10, or 2 mg/kg/day) 1 day before and on the day of intracerebral injection with a minocycline-resistant S. aureus strain (S. aureustetM). Animals were sacrificed at 3, 6, or 12 hours following S. aureustetM exposure, whereupon IL-1ß (A) and CCL2 (B) expression was evaluated by qRT-PCR. Bacterial burdens were also determined (C) to verify that any effects of minocycline were not the result of altered bacterial replication. Significant differences between S. aureustetM-infected mice treated with vehicle (PBS) versus minocycline are denoted with asterisks (*P < 0.05, **P < 0.001). Results are representative of three independent experiments.


Minocycline Reduces Brain Abscess Size


The finding that minocycline was capable of transiently attenuating proinflammatory mediator expression in brain abscesses led us to consider that the degree of tissue injury may be less severe in these animals. The extent of brain abscess involvement was determined by performing hematoxylin and eosin stains on serial tissue sections prepared throughout the entire lesion to identify the largest abscess cross-sectional area. As shown in Figure 6 , minocycline pretreatment led to a significant sparing of brain parenchyma from necrotic damage, as evident by the fact that abscess sizes were dramatically reduced in minocycline-treated mice. To better demarcate the borders between necrotic tissue and viable peri-abscess parenchyma at the early time points examined following S. aureus infection, tissue viability stains with NBT were performed. As shown in Figure 7 , minocycline-treated animals exhibited less necrotic changes compared with vehicle-treated mice.


Figure 6. Minocycline treatment reduces brain abscess size. Mice (n = 10 mice/group) received i.p. injections of PBS or minocycline (50 mg/kg) 1 day before and on the day of intracerebral injection with a minocycline-resistant S. aureus strain (S. aureustetM). Animals were sacrificed at 12 or 24 hours following bacterial exposure, whereupon brain tissues were flash-frozen on dry ice for subsequent cryostat sectioning. Serial sections were prepared throughout the entire abscess to ensure that the maximal cross-sectional area was identified and stained with hematoxylin and eosin to demarcate the extent of tissue damage. In A, a series of microscopic images (magnification, x4) were assembled for each individual vehicle- or minocycline-treated animal to demonstrate the extent of abscess formation. Abscess margins (including cerebritis) are demarcated with dotted lines. In B, abscess area (mm2; mean ?? SD) was quantitated using the MetaMorph image analysis program by measuring the largest lesion size for each tissue specimen. Significant differences in brain abscess size between vehicle- or minocycline-treated mice are denoted with asterisks (*P < 0.05). Results are representative of two independent experiments.


Figure 7. Minocycline reduces necrotic changes during the early phases of brain abscess formation. Mice (n = 10 mice/group) received i.p. injections of PBS or minocycline (50 mg/kg) 1 day before and on the day of intracerebral injection with a minocycline-resistant S. aureus strain (S. aureustetM). Animals were sacrificed at 12 or 24 hours following bacterial exposure, whereupon brain tissues were flash-frozen on dry ice for subsequent cryostat sectioning. Serial sections were prepared throughout the entire abscess to ensure that the maximal cross-sectional area was identified and incubated with the vital dye NBT as described in Materials and Methods to demarcate the border between necrotic and viable peri-abscess parenchyma. In A, a series of microscopic images (magnification, x4) were assembled for each individual vehicle- or minocycline-treated animal to demonstrate the extent of necrosis. The margins between necrotic and viable tissue are demarcated with arrowheads. In B, representative high magnification fields are provided to facilitate the identification of the transition from viable (basophilic) to necrotic (pale eosinophilic to no color) tissue. Results are representative of tissues from four to six individual animals per treatment group.


To determine whether the decrease in proinflammatory mediator expression and brain abscess size observed following minocycline treatment correlated with alterations in astrocyte activation, GFAP reactivity was evaluated by immunofluorescence staining in the peri-abscess area. The degree of reactive astrocytosis progressively increased over time (Figure 8) ; however, minocycline treatment had no effect on either the relative number or degree of astrocyte activation along the abscess margins. We also attempted to evaluate the effects of minocycline on microglial/macrophage activation by F4/80 staining; however, sufficient signal was not observed at these early time points following S. aureus infection (data not shown).


Figure 8. Minocycline does not influence astrocyte activation during the early stage of brain abscess development. Mice (n = 10 mice/group) received i.p. injections of PBS or minocycline (50 mg/kg) 1 day before and on the day of intracerebral injection with a minocycline-resistant S. aureus strain (S. aureustetM). Animals were sacrificed at 12 or 24 hours following bacterial exposure, whereupon brain tissues were flash-frozen on dry ice for subsequent cryostat sectioning. Ten-micrometer-thick brain abscess sections were subjected to immunofluorescence staining using GFAP (green) and imaged by confocal microscopy. GFAP immunoreactivity is shown along the peri-abscess area, where the dark areas represent necrotic regions. Results presented are representative of a minimum of four individual mice per time point.


Minocycline Is Capable of Selectively Modulating Pre-Existing Inflammation in Brain Abscesses


Although our studies had demonstrated that minocycline attenuated proinflammatory mediator expression and brain abscess size in a pretreatment paradigm, this strategy is not reflective of a plausible clinical approach to modulate infections. Therefore, we next determined whether minocycline could affect established inflammation during brain abscess evolution. In these studies, mice received single daily doses of minocycline beginning at day 3 postinfection. Animals were sacrificed at 1 and 3 days following minocycline treatment for analysis, which corresponded to days 4 and 6 postinfection, respectively. Similar to what was observed in the pretreatment paradigm, minocycline significantly attenuated the expression of several proinflammatory mediators (Figure 9) as well as brain abscess size (Figure 10) . The extent of abscess-associated edema observed in minocycline-treated mice was notably reduced compared with vehicle-treated animals and was most dramatic at day 3 following minocycline administration (Figure 9A) . Interestingly, maximal protection of brain parenchyma was observed when mice received three consecutive daily doses of minocycline, whereas a single dose was not as effective. This result is not unexpected given the fact that at this stage, brain abscesses are typified by a vigorous, progressive inflammatory response, making it difficult for a single minocycline dose to exert any demonstrable effects. Examination of astrocyte and microglial/macrophage activation by immunofluorescence staining with GFAP and F4/80, respectively, did not reveal any significant differences following minocycline treatment, although the relative degree of immunoreactivity of both cell populations in the peri-abscess area progressively increased over time (Figure 11) , as we have previously demonstrated.11 Collectively, these results indicate that minocycline could represent a potential therapeutic agent even in conditions of pre-existing inflammation to attenuate bystander destruction of surrounding normal brain parenchyma.


Figure 9. Delayed minocycline administration is still capable of attenuating proinflammatory mediator expression. Mice (n = 4 or 5 per group) received i.p. injections of PBS or minocycline (50 mg/kg/day) beginning at 3 days following an intracerebral injection with the minocycline-resistant S. aureus strain (S. aureustetM), with treatment continuing daily until termination of the experiment. Animals were sacrificed at either 1 or 3 days after minocycline treatment (corresponding to days 4 and 6 postinfection), whereupon IL-1ß (A) and CXCL2 (B) expression were evaluated by ELISA and normalized to the amount of total protein recovered from abscesses to correct for differences in tissue sampling size. Bacterial burdens were also determined (C) to verify that any inhibitory effects of minocycline were not the result of reduced bacterial replication. Significant differences between S. aureustetM-infected mice treated with vehicle (PBS) versus minocycline are denoted with asterisks (*P < 0.05). Results are representative of three independent experiments.


Figure 10. Minocycline is capable of significantly reducing brain abscess size in the face of established infection. Mice (n = 6/group) received i.p. injections of PBS or minocycline (50 mg/kg/day) beginning at 3 days following an intracerebral injection with the minocycline-resistant S. aureus strain (S. aureustetM), with treatment continuing daily until termination of the experiment. Animals were sacrificed at either 1 or 3 days after minocycline treatment (corresponding to days 4 and 6 postinfection), whereupon brain tissues were flash-frozen on dry ice for subsequent cryostat sectioning. Serial sections were prepared throughout the entire abscess to ensure that the maximal cross-sectional area was identified and stained with hematoxylin and eosin to demarcate the extent of tissue damage. In A, a series of microscopic images (magnification, x4) were assembled for each individual vehicle- or minocycline-treated animal to demonstrate the extent of abscess formation and edema/necrosis. Abscess margins (including cerebritis) are demarcated with dotted lines. In B, abscess area (mm2; mean ?? SD) was quantitated using the MetaMorph image analysis program by measuring the largest lesion size for each tissue specimen. Significant differences in brain abscess size between vehicle- or minocycline-treated mice are denoted with asterisks (*P < 0.05, **P < 0.001). Results are representative of two independent experiments.


Figure 11. Minocycline does not influence astrocyte or microglial/macrophage activation when administered subsequent to infection. Mice (n = 6/group) received i.p. injections of PBS or minocycline (50 mg/kg/day) beginning at 3 days following an intracerebral injection with the minocycline-resistant S. aureus strain (S. aureustetM), with treatment continuing daily until termination of the experiment. Animals were sacrificed at either 1 or 3 days after minocycline treatment (corresponding to days 4 and 6 postinfection), whereupon brain tissues were flash-frozen on dry ice for subsequent cryostat sectioning. Ten-micrometer-thick brain abscess sections were subjected to immunofluorescence staining using GFAP (A; green) or F4/80 (B; red) to identify astrocytes and microglia/macrophages, respectively, and imaged by confocal microscopy. GFAP and F4/80 immunoreactivity is shown along the peri-abscess area, where the dark areas represent necrotic regions. Results presented are representative of a minimum of four independent mice per time point.


Minocycline Selectively Attenuates Proinflammatory Mediator Production in S. aureus- and PGN-Activated Glia


Microglia are the resident innate immune cells in the CNS parenchyma and are uniquely poised to immediately respond to bacterial infections by producing numerous cytokines and chemokines.76-78 The neuroprotective effects of minocycline have been attributed, in part, to its ability to attenuate microglial activation both in vivo and in vitro.31,33,43,48,49 However, the ability of minocycline to modulate microglial responses to a prevalent CNS pathogen, such as S. aureus, has not yet been investigated. Both S. aureus and its cell wall product, PGN, induced microglial activation typified by the release of IL-1ß and CXCL2 (Figure 12) . Pretreatment of microglia with minocycline for 1 hour before the addition of bacterial stimuli significantly attenuated IL-1ß and CXCL2 expression, although considerable levels of each mediator remained (Figure 12, A and B) . The reduction in proinflammatory mediator expression following minocycline treatment was not the result of overt cell death, because cell viability assays revealed that minocycline was not toxic to primary microglia at any of the doses examined (Figure 12C) . Interestingly, the inhibitory effects of minocycline were not universal, as the compound did not have any significant effects in regulating the expression of IL-12 p70 or IL-6 in response to bacterial stimulation (data not shown).


Figure 12. Minocycline selectively inhibits S. aureus- and PGN-induced proinflammatory mediator expression in microglia. Primary microglia were pre-treated with the indicated concentrations of minocycline for 1 hour before stimulation with heat-inactivated S. aureus (107 cfu/well) or PGN (10 µg/ml). As a control to determine whether minocycline alone exerts any effects on microglial responses, cells were also subjected to treatment with the highest dose of minocycline examined (ie, 50 µg/ml) in the absence of bacterial stimulation. Supernatants were collected at 24 hours following S. aureus or PGN exposure, whereupon IL-1ß (A) and CXCL2 (B) release were quantitated by ELISAs. Microglial viability was assessed using a standard MTT assay and the raw OD570 readings are reported (mean ?? SD; C). Significant differences between microglia exposed to S. aureus or PGN only versus cells pretreated with the various concentrations of minocycline + S. aureus or PGN are denoted with asterisks (*P < 0.01, **P < 0.001). Results are representative of six independent experiments.


Astrocytes represent the most numerous cell type in the brain and are recognized as one of the principal sources of CNS-derived chemokines.79,80 To our knowledge, the immunoregulatory effects of minocycline on astrocytic responses have not yet been investigated. Similar to what was observed with microglia, minocycline was capable of significantly attenuating S. aureus-dependent astrocyte activation in terms of NO and IL-1ß release, although residual expression of both mediators was still significant (Figure 13, A and B , respectively). The reduction in astrocytic proinflammatory mediator expression following minocycline treatment was not the result of toxic effects at any of the concentrations examined (Figure 13C) . Minocycline was not effective at inhibiting microglial or astrocyte activation when administered at either 1 or 6 hours following bacterial exposure (data not shown), which is not unexpected given the relatively modest ability of the compound to affect glial proinflammatory mediator release in the pretreatment dosing paradigm presented.


Figure 13. Minocycline inhibits S. aureus- and PGN-induced proinflammatory mediator expression in astrocytes. Astrocytes were pretreated with the indicated concentrations of minocycline for 1 hour before S. aureus or PGN stimulation, whereupon NO (A) and IL-1ß (B) release were quantified by nitrite assay and ELISA, respectively. Astrocyte viability was assessed using a standard MTT assay, and the raw OD570 readings are reported (mean ?? SD; C). As a control to determine whether minocycline alone exerts any effects on astrocyte responses, cells were also subjected to treatment with the highest dose of minocycline examined (ie, 50 µg/ml) in the absence of bacterial stimulation. Significant differences between astrocytes exposed to S. aureus or PGN only versus cells pretreated with the various concentrations of minocycline + S. aureus or PGN are denoted with asterisks (*P < 0.01, **P < 0.001). Results are representative of four independent experiments.


Minocycline Attenuates TLR2 Expression in Activated Microglia and Astrocytes


TLRs enable the host to recognize microbial motifs that are highly conserved across various classes of microorganisms.81-83 Among the 13 TLR family members identified to date, TLR2 is capable of recognizing lipid-based structural components from a broad range of microbes, including various gram-positive bacteria, fungi, and protozoa.81,84 We elected to focus on evaluating the effects of minocycline on glial TLR2 expression, since we have previously published that this receptor plays a pivotal role in S. aureus and PGN recognition by both microglia and astrocytes, triggering the production of a wide array of inflammatory mediators by activated glia.69,73 Therefore, evaluation of TLR2 expression represents a read-out to imply the degree of "bacterial responsiveness" of glia to infection. To our knowledge, the ability of minocycline to modulate TLR2 expression in activated glia has not yet been examined. TLR2 expression in both microglia and astrocytes was increased following bacterial exposure and was partially suppressed by minocycline (Figure 14, A and B , respectively).


Figure 14. Minocycline attenuates the S. aureus-dependent increase in glial TLR2 expression. Primary microglia (A) and astrocytes (B) were stimulated with 107 cfu of heat-inactivated S. aureus or PGN (10 µg/ml) in the presence or absence of 50 µg/ml minocycline. Protein extracts from whole cell lysates were prepared at 24 hours following stimulation and 40 µg of total protein from each sample was evaluated for TLR2 levels by Western blotting. Membranes were subsequently stripped and re-probed with ß-actin to verify uniformity in gel loading. Results are representative of six independent experiments.


Discussion


Recent studies by others have demonstrated that minocycline exhibits beneficial effects in several infectious disease models including simian immunodeficiency virus, reovirus, neuroadapted Sindbus virus, and African trypanosomes.61-64 Interestingly, in the human immunodeficiency virus-1 and reovirus studies, minocycline was shown to attenuate viral replication, which was thought to result from the inhibitory effects of the drug on transcription factors known to influence human immunodeficiency virus-1 transcription, including p38 mitogen-activated protein kinase.62,63 This inhibitory effect of minocycline on human immunodeficiency virus-1 replication has also been demonstrated in vitro in macrophages62 and microglia.50 Our present work adds to the growing list of infectious neuropathologies for which minocycline has been shown to play a beneficial role. However, to our knowledge, this work is the first to describe an immune modulatory role for minocycline in the context of a CNS bacterial infection independently of the drug??s antimicrobial activity.


In bacterial meningitis, the CNS inflammatory response that ensues following infection plays a role in disease pathophysiology.19,20 Specifically, numerous cytokines, free radicals, and excitatory amino acids have all been implicated in negatively impacting neuron survival. Although it was possible that if excessive, the immediate antibacterial response to S. aureus in brain abscess might exacerbate disease severity, this possibility remained unclear, since we had previously demonstrated that several inflammatory mediators, including IL-1, TNF-, and CXCL2, were critical during the early phases of infection.7,8 With the generation of a minocycline-resistant S. aureus strain, our results demonstrated that minocycline modulates the acute host response to S. aureus in the CNS parenchyma as evident by the fact that minocycline significantly reduced abscess-associated mortality and improved brain parenchymal survival. Importantly, these effects did not result from differences in bacterial burdens, because both minocycline- and vehicle-treated mice displayed equivalent S. aureus levels. The ability of minocycline to minimize tissue damage may result, in part, from a reduction in cytokine-mediated bystander lysis, because several cytokines/chemokines were attenuated by minocycline in brain abscesses including IL-1ß, TNF-, CXCL2, and CCL2. The ability of minocycline to attenuate IL-1 production may be due to its ability to inhibit caspase-1 activity, as previously reported by others.31,38 These findings indicate that although inflammation is a prerequisite for effective pathogen clearance, this response can become exaggerated and culminate in adverse effects to the host. This dysregulated inflammatory response is thought to be driven, in part, by bacterial cell wall debris and/or DNA associated with the necrotic abscess milieu that serve as further triggers for pattern recognition receptors such as TLRs. In addition, many antibiotics that are used to treat CNS gram-positive infections can enhance the shedding of cell wall antigens, further confounding this issue.85,86 Therefore, we propose that the exaggerated release of proinflammatory mediators such as IL-1ß, TNF-, and CXCL2 may contribute to the nonspecific destruction of surrounding normal brain parenchyma by bystander lysis. In addition, minocycline was capable of reducing the S. aureus-dependent increase in glial TLR2 expression, suggesting that one way minocycline may attenuate inflammation is via interference with bacterial recognition pathways. Collectively, these results indicate that minocycline may serve to balance the ensuing inflammatory response to infection and ensure the development of an optimal antibacterial immune response.


One important question that remains is defining the complete repertoire of neuroprotective mechanisms for minocycline in the experimental brain abscess model, in addition to its ability to modulate proinflammatory mediator expression as demonstrated in this study. However, the accurate identification of affected host responses will require the establishment of an alternative dosing paradigm to overcome obstacles related to the short half-life of minocycline in the mouse (ie, 2 to 3 hours),22,66,67 compounded with the intense inflammation associated with a bacterial infection. For example, in brain abscess, the continued proliferation of bacteria coupled with shedding of cell wall determinants that are potent activators of TLRs83 probably overwhelms the anti-inflammatory effects of minocycline more rapidly compared with a noninfectious inflammatory milieu that is observed in Parkinson??s or Huntington??s disease or following spinal cord injury. Therefore, in brain abscess we envision that a continuous dosing strategy for minocycline would be required to achieve maximal anti-inflammatory effects. We plan to address these issues by delivering a constant supply of minocycline via either slow release pellets or osmotic minipumps. By maintaining therapeutic levels of minocycline in infected animals, we anticipate that potential differences in host responses will be more pronounced, enabling the accurate identification of the pathways affected by minocycline. This was considered to be beyond the scope of this initial report demonstrating the effectiveness of minocycline in the experimental brain abscess model. Notably, we were not able to detect any significant effects of minocycline on astrocyte or microglial/macrophage activation in the peri-abscess area, suggesting that the observed differences in inflammation were not attributable to an overt reduction in glial activation using the dosing paradigms tested in the current study. These results are in agreement with the relatively modest effects of minocycline on microglial and astrocyte activation in vitro, suggesting that mechanisms other than cytokine/chemokine production could play a role in the ability of minocycline to reduce brain abscess size. However, it is important to acknowledge that differences in the degree of astrocyte and/or microglial/macrophage activation could still exist but may not be discernable by merely examining GFAP and F4/80 expression. More detailed studies examining the cytokine expression profiles of these cell populations upon recovery from brain abscesses by fluorescence-activated cell sorting are warranted in the context of a more continuous dosing paradigm as discussed above.


Alternative pathways that may be affected by minocycline to reduce tissue damage during brain abscess development include the drug??s ability to attenuate matrix metalloproteinase expression and activity, inhibit apoptosis, and/or regulate the influx of peripheral immune cells to minimize nonspecific tissue destruction. The ability of minocycline to modulate leukocyte entry into evolving brain abscesses could be due to a number of mechanisms, namely, a direct action on matrix metalloproteinase enzymatic activity and/or matrix metalloproteinase expression,44,55,66 alterations in the levels of adhesion molecules required for leukocyte extravasation across the blood-brain barrier, and/or chemokine expression. On the other hand, minocycline has been reported to act as a free radical scavenger87 and to inhibit the production of neutrophil-attracting molecules from bacteria.88 Finally, in addition to these effector mechanisms, we cannot discount a potential antiapoptotic role for minocycline in reducing parenchymal damage through its ability to inhibit caspase-3 expression. Although we have not yet examined the extent of apoptosis in the brain abscess model, work by others using this model have demonstrated that apoptosis represents a relatively minor component of cell death.14 Cumulatively, the reversal of these pathways that under certain circumstances can be considered detrimental to cell survival could effectively rescue brain parenchyma from excessive destruction during brain abscess evolution.


In addition to its ability to modulate the host response to infection, it is also important to acknowledge that minocycline may also exert direct effects on S. aureus by modulating its virulence factor expression profile. In susceptible strains, minocycline exerts its bacteriostatic activity by binding to bacterial ribosomes and preventing protein expression.25,26 The tetM gene confers resistance to both minocycline and tetracycline and interferes with antibiotic binding to bacterial ribosomes. However, it is possible that minocycline selection may alter the array of virulence factors produced due to direct actions on genes important for bacterial pathogenesis. Although our in vitro analysis of the parental and S. aureustetM strains did not reveal any dramatic differences in protein expression profiles, variations in the face of antibiotic pressure in vivo are still possible. On the other hand, the ability of minocycline to alter the host immune response to S. aureus may lead to adaptive changes in bacterial virulence factor expression as a result of altered selective pressures in the brain. These possibilities could be addressed by performing gene expression profiling microarrays of bacterial RNA recovered from minocycline- or vehicle-treated mice infected with the S. aureustetM strain, studies we are currently exploring in the laboratory.


From a clinical perspective, any candidate immune prophylactic agent should exhibit beneficial effects when administered following the onset of disease. This criterion seemed to be partially satisfied in our present study as demonstrated by the ability of minocycline to attenuate proinflammatory mediator production and brain abscess size when treatment was initiated 3 days following S. aureus infection. Therefore, our data suggest that minocycline may represent an attractive candidate for the therapeutic management of brain abscess for the following reasons. First, the bacteriostatic properties of minocycline effectively reduce CNS bacterial burdens in susceptible strains. Second, in addition to its antimicrobial effects, minocycline can also attenuate pathological CNS inflammation that probably contributes to the expansion of abscess size through bystander damage to surrounding uninfected brain parenchyma. Third, minocycline exhibits excellent blood-brain barrier permeability because of its small and lipophilic nature and, based on these properties, would be expected to reach high therapeutic levels within the abscess milieu, a requisite for any effective antimicrobial therapy for brain abscess.


Acknowledgements


We thank Dr. Paul Drew and Dr. Joyce DeLeo for critical review of the manuscript, and Anessa Haney, Patrick Mayes, Jennifer Johnson, and Gail Wagoner for excellent technical assistance.


【参考文献】
  Mathisen GE, Johnson JP: Brain abscess. Clin Infect Dis 1997, 25:763-781

Townsend GC, Scheld WM: Infections of the central nervous system. Adv Intern Med 1998, 43:403-447

Kielian T: Immunopathogenesis of brain abscess. J Neuroinflammation 2004, 1:16

Lu CH, Chang WN, Lui CC: Strategies for the management of bacterial brain abscess. J Clin Neurosci 2006, 13:979-985

Cunha BA: Methicillin-resistant Staphylococcus aureus: clinical manifestations and antimicrobial therapy. Clin Microbiol Infect 2005, 11(Suppl 4):33-42

Ruhe JJ, Monson T, Bradsher RW, Menon A: Use of long-acting tetracyclines for methicillin-resistant Staphylococcus aureus infections: case series and review of the literature. Clin Infect Dis 2005, 40:1429-1434

Kielian T, Barry B, Hickey WF: CXC chemokine receptor-2 ligands are required for neutrophil-mediated host defense in experimental brain abscesses. J Immunol 2001, 166:4634-4643

Kielian T, Bearden ED, Baldwin AC, Esen N: IL-1 and TNF- play a pivotal role in the host immune response in a mouse model of Staphylococcus aureus-induced experimental brain abscess. J Neuropathol Exp Neurol 2004, 63:381-396

Kielian T, Haney A, Mayes PM, Garg S, Esen N: Toll-like receptor 2 modulates the proinflammatory milieu in staphylococcus aureus-induced brain abscess. Infect Immun 2005, 73:7428-7435

Kielian T, Cheung A, Hickey WF: Diminished virulence of an -toxin mutant of Staphylococcus aureus in experimental brain abscesses. Infect Immun 2001, 69:6902-6911

Baldwin AC, Kielian T: Persistent immune activation associated with a mouse model of Staphylococcus aureus-induced experimental brain abscess. J Neuroimmunol 2004, 151:24-32

Bloch O, Papadopoulos MC, Manley GT, Verkman AS: Aquaporin-4 gene deletion in mice increases focal edema associated with staphylococcal brain abscess. J Neurochem 2005, 95:254-262

Lo WD, Wolny A, Boesel C: Blood-brain barrier permeability in staphylococcal cerebritis and early brain abscess. J Neurosurg 1994, 80:897-905

Stenzel W, Soltek S, Miletic H, Hermann MM, Korner H, Sedgwick JD, Schluter D, Deckert M: An essential role for tumor necrosis factor in the formation of experimental murine Staphylococcus aureus-induced brain abscess and clearance. J Neuropathol Exp Neurol 2005, 64:27-36

Kielian T, Phulwani NK, Esen N, Syed MM, Haney AC, McCastlain K, Johnson J: MyD88-dependent signals are essential for the host immune response in experimental brain abscess. J Immunol 2007, 178:4528-4537

Kielian T, Hickey WF: Proinflammatory cytokine, chemokine, and cellular adhesion molecule expression during the acute phase of experimental brain abscess development. Am J Pathol 2000, 157:647-658

Flaris NA, Hickey WF: Development and characterization of an experimental model of brain abscess in the rat. Am J Pathol 1992, 141:1299-1307

Stenzel W, Soltek S, Schluter D, Deckert M: The intermediate filament GFAP is important for the control of experimental murine Staphylococcus aureus-induced brain abscess and Toxoplasma encephalitis. J Neuropathol Exp Neurol 2004, 63:631-640

Nau R, Bruck W: Neuronal injury in bacterial meningitis: mechanisms and implications for therapy. Trends Neurosci 2002, 25:38-45

Koedel U, Scheld WM, Pfister HW: Pathogenesis and pathophysiology of pneumococcal meningitis. Lancet Infect Dis 2002, 2:721-736

Jonas M, Cunha BA: Minocycline. Ther Drug Monit 1982, 4:137-145

Colovic M, Caccia S: Liquid chromatographic determination of minocycline in brain-to-plasma distribution studies in the rat. J Chromatogr B Analyt Technol Biomed Life Sci 2003, 791:337-343

Smith DL, Woodman B, Mahal A, Sathasivam K, Ghazi-Noori S, Lowden PA, Bates GP, Hockly E: Minocycline and doxycycline are not beneficial in a model of Huntington??s disease. Ann Neurol 2003, 54:186-196

Agwuh KN, MacGowan A: Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines. J Antimicrob Chemother 2006, 58:256-265

Speer BS, Shoemaker NB, Salyers AA: Bacterial resistance to tetracycline: mechanisms, transfer, and clinical significance. Clin Microbiol Rev 1992, 5:387-399

Roberts MC: Update on acquired tetracycline resistance genes. FEMS Microbiol Lett 2005, 245:195-203

Yrjänheikki J, Keinanen R, Pellikka M, Hokfelt T, Koistinaho J: Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc Natl Acad Sci USA 1998, 95:15769-15774

Yrjänheikki J, Tikka T, Keinanen R, Goldsteins G, Chan PH, Koistinaho J: A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci USA 1999, 96:13496-13500

Fan LW, Lin S, Pang Y, Rhodes PG, Cai Z: Minocycline attenuates hypoxia-ischemia-induced neurological dysfunction and brain injury in the juvenile rat. Eur J Neurosci 2006, 24:341-350

Liu Z, Fan Y, Won SJ, Neumann M, Hu D, Zhou L, Weinstein PR, Liu J: Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke 2007, 38:146-152

Chen M, Ona VO, Li M, Ferrante RJ, Fink KB, Zhu S, Bian J, Guo L, Farrell LA, Hersch SM, Hobbs W, Vonsattel JP, Cha JH, Friedlander RM: Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease. Nat Med 2000, 6:797-801

Du Y, Ma Z, Lin S, Dodel RC, Gao F, Bales KR, Triarhou LC, Chernet E, Perry KW, Nelson DL, Luecke S, Phebus LA, Bymaster FP, Paul SM: Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson??s disease. Proc Natl Acad Sci USA 2001, 98:14669-14674

Wu DC, Jackson-Lewis V, Vila M, Tieu K, Teismann P, Vadseth C, Choi DK, Ischiropoulos H, Przedborski S: Blockade of microglial activation is neuroprotective in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson disease. J Neurosci 2002, 22:1763-1771

Peng J, Xie L, Stevenson FF, Melov S, Di Monte DA, Andersen JK: Nigrostriatal dopaminergic neurodegeneration in the weaver mouse is mediated via neuroinflammation and alleviated by minocycline administration. J Neurosci 2006, 26:11644-11651

Seabrook TJ, Jiang L, Maier M, Lemere CA: Minocycline affects microglia activation, Aß deposition, and behavior in APP-tg mice. Glia 2006, 53:776-782

Ryu JK, Franciosi S, Sattayaprasert P, Kim SU, McLarnon JG: Minocycline inhibits neuronal death and glial activation induced by ß-amyloid peptide in rat hippocampus. Glia 2004, 48:85-90

Hunter CL, Bachman D, Granholm AC: Minocycline prevents cholinergic loss in a mouse model of Down??s syndrome. Ann Neurol 2004, 56:675-688

Zhu S, Stavrovskaya IG, Drozda M, Kim BY, Ona V, Li M, Sarang S, Liu AS, Hartley DM, du Wu C, Gullans S, Ferrante RJ, Przedborski S, Kristal BS, Friedlander RM: Minocycline inhibits cytochrome c release and delays progression of amyotrophic lateral sclerosis in mice. Nature 2002, 417:74-78

Kriz J, Nguyen MD, Julien JP: Minocycline slows disease progression in a mouse model of amyotrophic lateral sclerosis. Neurobiol Dis 2002, 10:268-278

Wells JE, Hurlbert RJ, Fehlings MG, Yong VW: Neuroprotection by minocycline facilitates significant recovery from spinal cord injury in mice. Brain 2003, 126:1628-1637

Lee SM, Yune TY, Kim SJ, Park do W, Lee YK, Kim YC, Oh YJ, Markelonis GJ, Oh TH: Minocycline reduces cell death and improves functional recovery after traumatic spinal cord injury in the rat. J Neurotrauma 2003, 20:1017-1027

Stirling DP, Khodarahmi K, Liu J, McPhail LT, McBride CB, Steeves JD, Ramer MS, Tetzlaff W: Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves functional outcome after spinal cord injury. J Neurosci 2004, 24:2182-2190

Popovic N, Schubart A, Goetz BD, Zhang SC, Linington C, Duncan ID: Inhibition of autoimmune encephalomyelitis by a tetracycline. Ann Neurol 2002, 51:215-223

Brundula V, Rewcastle NB, Metz LM, Bernard CC, Yong VW: Targeting leukocyte MMPs and transmigration: minocycline as a potential therapy for multiple sclerosis. Brain 2002, 125:1297-1308

Nessler S, Dodel R, Bittner A, Reuss S, Du Y, Hemmer B, Sommer N: Effect of minocycline in experimental autoimmune encephalomyelitis. Ann Neurol 2002, 52:689-690

Blum D, Chtarto A, Tenenbaum L, Brotchi J, Levivier M: Clinical potential of minocycline for neurodegenerative disorders. Neurobiol Dis 2004, 17:359-366

Tikka TM, Koistinaho JE: Minocycline provides neuroprotection against N-methyl-D-aspartate neurotoxicity by inhibiting microglia. J Immunol 2001, 166:7527-7533

Tikka T, Fiebich BL, Goldsteins G, Keinanen R, Koistinaho J: Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci 2001, 21:2580-2588

Familian A, Boshuizen RS, Eikelenboom P, Veerhuis R: Inhibitory effect of minocycline on amyloid beta fibril formation and human microglial activation. Glia 2006, 53:233-240

Si Q, Cosenza M, Kim MO, Zhao ML, Brownlee M, Goldstein H, Lee S: A novel action of minocycline: inhibition of human immunodeficiency virus type 1 infection in microglia. J Neurovirol 2004, 10:284-292

Nikodemova M, Duncan ID, Watters JJ: Minocycline exerts inhibitory effects on multiple mitogen-activated protein kinases and IkappaBalpha degradation in a stimulus-specific manner in microglia. J Neurochem 2006, 96:314-323

Kloppenburg M, Brinkman BM, de Rooij-Dijk HH, Miltenburg AM, Daha MR, Breedveld FC, Dijkmans BA, Verweij C: The tetracycline derivative minocycline differentially affects cytokine production by monocytes and T lymphocytes. Antimicrob Agents Chemother 1996, 40:934-940

Shapira L, Barak V, Soskolne WA, Halabi A, Stabholz A: Effects of tetracyclines on the pathologic activity of endotoxin: in vitro and in vivo studies. Adv Dent Res 1998, 12:119-122

D??Agostino P, Ferlazzo V, Milano S, La Rosa M, Di Bella G, Caruso R, Barbera C, Grimaudo S, Tolomeo M, Feo S, Cillari E: Anti-inflammatory effects of chemically modified tetracyclines by the inhibition of nitric oxide and interleukin-12 synthesis in J774 cell line. Int Immunopharmacol 2001, 1:1765-1776

Power C, Henry S, Del Bigio MR, Larsen PH, Corbett D, Imai Y, Yong VW, Peeling J: Intracerebral hemorrhage induces macrophage activation and matrix metalloproteinases. Ann Neurol 2003, 53:731-742

Mievis S, Levivier M, Communi D, Vassart G, Brotchi J, Ledent C, Blum D: Lack of Minocycline Efficiency in Genetic Models of Huntington??s Disease. Neuromolecular Med 2007, 9:47-54

Tsuji M, Wilson MA, Lange MS, Johnston MV: Minocycline worsens hypoxic-ischemic brain injury in a neonatal mouse model. Exp Neurol 2004, 189:58-65

Fendrick SE, Miller KR, Streit WJ: Minocycline does not inhibit microglia proliferation or neuronal regeneration in the facial nucleus following crush injury. Neurosci Lett 2005, 385:220-223

Diguet E, Fernagut PO, Wei X, Du Y, Rouland R, Gross C, Bezard E, Tison F: Deleterious effects of minocycline in animal models of Parkinson??s disease and Huntington??s disease. Eur J Neurosci 2004, 19:3266-3276

Diguet E, Gross CE, Tison F, Bezard E: Rise and fall of minocycline in neuroprotection: need to promote publication of negative results. Exp Neurol 2004, 189:1-4

Masocha W, Rottenberg ME, Kristensson K: Minocycline impedes African trypanosome invasion of the brain in a murine model. Antimicrob Agents Chemother 2006, 50:1798-1804

Zink MC, Uhrlaub J, DeWitt J, Voelker T, Bullock B, Mankowski J, Tarwater P, Clements J, Barber S: Neuroprotective and anti-human immunodeficiency virus activity of minocycline. JAMA 2005, 293:2003-2011

Richardson-Burns SM, Tyler KL: Minocycline delays disease onset and mortality in reovirus encephalitis. Exp Neurol 2005, 192:331-339

Darman J, Backovic S, Dike S, Maragakis NJ, Krishnan C, Rothstein JD, Irani DN, Kerr DA: Viral-induced spinal motor neuron death is non-cell-autonomous and involves glutamate excitotoxicity. J Neurosci 2004, 24:7566-7575

Lee CY, Buranen SL, Ye ZH: Construction of single-copy integration vectors for Staphylococcus aureus. Gene 1991, 103:101-105

Yong VW, Wells J, Giuliani F, Casha S, Power C, Metz LM: The promise of minocycline in neurology. Lancet Neurol 2004, 3:744-751

Andes D, Craig WA: Animal model pharmacokinetics and pharmacodynamics: a critical review. Int J Antimicrob Agents 2002, 19:261-268

Hickey MJ, Hurley JV, Morrison WA: Temporal and spatial relationship between no-reflow phenomenon and postischemic necrosis in skeletal muscle. Am J Physiol 1996, 271:H1277-H1286

Esen N, Tanga FY, DeLeo JA, Kielian T: Toll-like receptor 2 (TLR2) mediates astrocyte activation in response to the gram-positive bacterium Staphylococcus aureus. J Neurochem 2004, 88:746-758

Esen N, Kielian T: Central role for MyD88 in the responses of microglia to pathogen-associated molecular patterns. J Immunol 2006, 176:6802-6811

Phulwani NK, Feinstein DL, Gavrilyuk V, Akar C, Kielian T: 15-Deoxy-12,14-prostaglandin J2 (15d-PGJ2) and ciglitazone modulate Staphylococcus aureus-dependent astrocyte activation primarily through a PPAR-gamma-independent pathway. J Neurochem 2006, 99:1389-1402

Kielian T, McMahon M, Bearden ED, Baldwin AC, Drew PD, Esen N: S. aureus-dependent microglial activation is selectively attenuated by the cyclopentenone prostaglandin 15-deoxy-12,14-prostaglandin J2 (15d-PGJ2). J Neurochem 2004, 90:1163-1172

Kielian T, Esen N, Bearden ED: Toll-like receptor 2 (TLR2) is pivotal for recognition of S. aureus peptidoglycan but not intact bacteria by microglia. Glia 2005, 49:567-576

Tanga FY, Nutile-McMenemy N, DeLeo JA: The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci USA 2005, 102:5856-5861

Trzcinski K, Cooper BS, Hryniewicz W, Dowson CG: Expression of resistance to tetracyclines in strains of methicillin-resistant Staphylococcus aureus. J Antimicrob Chemother 2000, 45:763-770

Aloisi F: Immune function of microglia. Glia 2001, 36:165-179

Hanisch UK: Microglia as a source and target of cytokines. Glia 2002, 40:140-155

Kielian T: Microglia and chemokines in infectious diseases of the nervous system: views and reviews. Front Biosci 2004, 9:732-750

Dong Y, Benveniste EN: Immune function of astrocytes. Glia 2001, 36:180-190

Farina C, Aloisi F, Meinl E: Astrocytes are active players in cerebral innate immunity. Trends Immunol 2007, 28:138-145

Akira S, Uematsu S, Takeuchi O: Pathogen recognition and innate immunity. Cell 2006, 124:783-801

Konat GW, Kielian T, Marriott I: The role of Toll-like receptors in CNS response to microbial challenge. J Neurochem 2006, 99:1-12

Kielian T: Toll-like receptors in central nervous system glial inflammation and homeostasis. J Neurosci Res 2006, 83:711-730

Kopp E, Medzhitov R: Recognition of microbial infection by Toll-like receptors. Curr Opin Immunol 2003, 15:396-401

Weber JR, Moreillon P, Tuomanen EI: Innate sensors for Gram-positive bacteria. Curr Opin Immunol 2003, 15:408-415

van der Flier M, Geelen SP, Kimpen JL, Hoepelman IM, Tuomanen EI: Reprogramming the host response in bacterial meningitis: how best to improve outcome? Clin Microbiol Rev 2003, 16:415-429

Kraus RL, Pasieczny R, Lariosa-Willingham K, Turner MS, Jiang A, Trauger JW: Antioxidant properties of minocycline: neuroprotection in an oxidative stress assay and direct radical-scavenging activity. J Neurochem 2005, 94:819-827

Akamatsu H, Niwa Y, Kurokawa I, Masuda R, Nishijima S, Asada Y: Effects of subminimal inhibitory concentrations of minocycline on neutrophil chemotactic factor production in comedonal bacteria, neutrophil phagocytosis and oxygen metabolism. Arch Dermatol Res 1991, 283:524-528


作者单位:From the Departments of Neurobiology and Developmental Sciences* and Infectious Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and the Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire

作者: Tammy Kielian*, Nilufer Esen*, Shuliang Liu*, Nirm 2008-5-29
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具