ArticlePDF Available

Methamphetamine Alters the Antimicrobial Efficacy of Phagocytic Cells during Methicillin-Resistant Staphylococcus aureus Skin Infection

American Society for Microbiology
mBio®
Authors:

Abstract and Figures

Unlabelled: Methamphetamine (METH) is a major drug of abuse in the United States and worldwide. Furthermore, Staphylococcus aureus infections and METH use are coemerging public health problems. S. aureus is the single most important bacterial pathogen in infections among injection drug users, with skin and soft tissue infections (SSTI) being extremely common. Notably, the incidence of SSTI, especially in drug users, is difficult to estimate because such infections are often self-treated. Although there is substantial information on the behavioral and cognitive defects caused by METH in drug users, there is a dearth of knowledge regarding its impact on bacterial infections and immunity. Therefore, we hypothesized that METH exacerbates S. aureus skin infection. Using a murine model of METH administration and wound infection, we demonstrated that METH reduces wound healing and facilitates host-mediated collagen degradation by increased expression and production of matrix metalloproteinase-2 (MMP-2). Additionally, we found that METH induces S. aureus biofilm formation and leads to detrimental effects on the functions of human and murine phagocytic cells, enhancing susceptibility to S. aureus infection. Our findings provide empirical evidence of the adverse impact of METH use on the antimicrobial efficacy of the cells that comprise innate immunity, the initial host response to combat microbial infection. Importance: METH is an extremely addictive central nervous system stimulant that is frequently administered by injection. SSTI, common problems among injection drug users, result in serious morbidity for patients and costly hospitalizations for treatment of superficial wounds and incision and drainage of abscesses; however, there has been little etiologic or preventive epidemiological research on this problem. In addition, the evasive nature of injection drug users toward medical care complicates our ability to accurately predict the prevalence of these infections. Hence, this study investigated the impact of METH use on S. aureus skin infection. Our findings demonstrate that this drug of abuse promotes biofilm formation and negatively impacts the wound healing process and innate immune function, exacerbating susceptibility to S. aureus infection. The findings may translate into new knowledge and development of therapeutic and public health strategies to deal with the devastating complications of METH abuse.
METH impairs wound healing by enhancing the host's MMP-2-mediated collagen degradation. (A) Histological analysis of untreated, MRSA, METH, or METH MRSA treatment groups of BALB/c mice at day 7 after wounding. The blue stain indicates collagen. Bar, 25 m. (B) Quantitative measurement of collagen intensity in tissues from untreated, MRSA, METH, and METH MRSA mice were measured using ImageJ software. Bars are the averages of the results, and error bars denote standard deviations. *, #, and & indicate significantly lower collagen intensites than in untreated, MRSA-infected, or METH-treated groups, respectively. (C) Gene expression analysis of collagen type I and III in cutaneous lesions. , , and indicate significantly higher fold changes than in untreated, MRSA-infected, or METH-treated groups, respectively. (D) Collagen degradation by MRSA staphopain A. FITC-conjugated type I collagen was incubated with 10 M staphopain A and METH (25 or 50 M) for 3 h at 37°C. The fluorescence of the supernatant was measured. Heat-inactivated staphopain A was utilized as a control. Dashed lines represent the averages for three measurements per condition, and error bars denote standard deviations. *, #, and & indicate significantly lower relative fluorescence than in the untreated or 25 M METH groups after cells were incubated with active staphopain A, respectively. (E) Gene expression analysis of MMP-2 in murine wounds. , , and indicate significantly higher fold changes than in untreated, MRSA-infected, or METH-treated groups, respectively. (F) Collagen degradation by MMP-2. FITC-conjugated type I collagen was incubated with 10 M MMP-2 and METH (25 or 50 M) for 3 h at 37°C. and indicate significantly higher relative fluorescence values than in the untreated or 25 M METH groups, respectively. *, #, and & indicate significantly lower relative fluorescence than in cells from untreated or the 25 or 50 M METH groups incubated with active MMP-2, respectively. (G) Western blot analysis of MMP-2 in wounded tissue (7 days) of untreated, MRSA, METH, or METH MRSA treatment in BALB/c mice. GAPDH was used as a loading control. For panels B to F, P values (with significance defined as P 0.05) were calculated by using an ANOVA. All the experiments in the figure were performed twice, and similar results were obtained. The results shown are representative of an individual experiment.
… 
Content may be subject to copyright.
Methamphetamine Alters the Antimicrobial Efficacy of Phagocytic
Cells during Methicillin-Resistant Staphylococcus aureus Skin Infection
Mircea Radu Mihu,
a
Jessica Roman-Sosa,
b,c
Avanish K. Varshney,
b,c
Eliseo A. Eugenin,
d,e
Bhavikkumar P. Shah,
f
Hiu Ham Lee,
g
Long N. Nguyen,
h
Allan J. Guimaraes,
i
Bettina C. Fries,
j,k
Joshua D. Nosanchuk,
b,c
Luis R. Martinez
b,c,g
Division of Infectious Diseases, Montefiore Medical Center, Bronx, New York, USA
a
; Division of Infectious Diseases, Department of Medicine,
b
and Department of
Microbiology and Immunology,
c
Albert Einstein College of Medicine, Bronx, New York, USA; Public Health Research Institute
d
and Department of Microbiology and
Molecular Genetics,
e
New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA; Department of Biomedical Sciences, Long Island
University—Post, Brookville, New York, USA
f
; College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
g
; Signature Research
Program in Cardiovascular and Metabolic Disorders, Duke—NUS Graduate Medical School, Singapore
h
; Instituto Biomédico, Universidade Federal Fluminense, Rio de
Janeiro, Brazil
i
; Division of Infectious Diseases, Department of Medicine,
j
and Department of Molecular Genetics and Microbiology,
k
Stony Brook University, Stony Brook,
New York, USA
ABSTRACT Methamphetamine (METH) is a major drug of abuse in the United States and worldwide. Furthermore, Staphylococ-
cus aureus infections and METH use are coemerging public health problems. S. aureus is the single most important bacterial
pathogen in infections among injection drug users, with skin and soft tissue infections (SSTI) being extremely common. Nota-
bly, the incidence of SSTI, especially in drug users, is difficult to estimate because such infections are often self-treated. Although
there is substantial information on the behavioral and cognitive defects caused by METH in drug users, there is a dearth of
knowledge regarding its impact on bacterial infections and immunity. Therefore, we hypothesized that METH exacerbates S.
aureus skin infection. Using a murine model of METH administration and wound infection, we demonstrated that METH re-
duces wound healing and facilitates host-mediated collagen degradation by increased expression and production of matrix
metalloproteinase-2 (MMP-2). Additionally, we found that METH induces S. aureus biofilm formation and leads to detrimental
effects on the functions of human and murine phagocytic cells, enhancing susceptibility to S. aureus infection. Our findings pro-
vide empirical evidence of the adverse impact of METH use on the antimicrobial efficacy of the cells that comprise innate immu-
nity, the initial host response to combat microbial infection.
IMPORTANCE METH is an extremely addictive central nervous system stimulant that is frequently administered by injection.
SSTI, common problems among injection drug users, result in serious morbidity for patients and costly hospitalizations for
treatment of superficial wounds and incision and drainage of abscesses; however, there has been little etiologic or preventive
epidemiological research on this problem. In addition, the evasive nature of injection drug users toward medical care compli-
cates our ability to accurately predict the prevalence of these infections. Hence, this study investigated the impact of METH use
on S. aureus skin infection. Our findings demonstrate that this drug of abuse promotes biofilm formation and negatively im-
pacts the wound healing process and innate immune function, exacerbating susceptibility to S. aureus infection. The findings
may translate into new knowledge and development of therapeutic and public health strategies to deal with the devastating com-
plications of METH abuse.
Received 23 September 2015 Accepted 28 September 2015 Published 27 October 2015
Citation Mihu MR, Roman-Sosa J, Varshney AK, Eugenin EA, Shah BP, Ham Lee H, Nguyen LN, Guimaraes AJ, Fries BC, Nosanchuk JD, Martinez LR. 2015. Methamphetamine
alters the antimicrobial efficacy of phagocytic cells during methicillin-resistant Staphylococcus aureus skin infection. mBio 6(6):e01622-15. doi:10.1128/mBio.01622-15.
Editor Larry S. McDaniel, University of Mississippi Medical Center
Copyright © 2015 Mihu et al. This is an open-access article distributed under the terms of the Creative Commons Attribution-Noncommercial-ShareAlike 3.0 Unported license,
which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original author and source are credited.
Address correspondence to Luis R. Martinez, lmarti13@nyit.edu.
Staphylococcus aureus is an immotile Gram-positive coccus that
frequently colonizes human nasal membranes and skin. It is
responsible for the majority of superficial and invasive skin infec-
tions, resulting in over 12,000,000 outpatient/emergency room
(ER) visits (1) and 400,000 hospital admissions annually in the
United States (2). Notably, in a study performed with multiple
ERs across the United States, methicillin-resistant S. aureus
(MRSA) strains were isolated from 61% of abscesses and 53% of
purulent wounds (3). Also, certain S. aureus clinical strains have
recently evolved with resistance to vancomycin, an antibiotic to
which staphylococci had previously been uniformly susceptible.
Although the vancomycin-resistant strains remain rare, MRSA
infections are increasingly common (4), and the incidence of
community-acquired MRSA strains has increased severalfold over
the past several years (5).
Methamphetamine (METH) is an extremely addictive central
nervous system stimulant abused by individuals worldwide, and
the drug is a major threat in many developed countries. The in-
toxicating effects of METH alter judgment and reduce inhibitions,
leading people to engage in unsafe activities that put them at risk
for acquiring transmissible microbes (6, 7). S. aureus, including
community-acquired MRSA, is the most important bacterial
RESEARCH ARTICLE crossmark
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 1
pathogen in skin and soft tissue infections (SSTI) among drug
users (8–10). SSTI incidence is difficult to assess among drug users
because such infections are often self-treated (11). However, crys-
tal METH injection is associated with frequent visits to the ER due
to abscesses, cellulitis, and other skin infections (12). Additionally,
MRSA, particularly USA300 strains, causes most SSTI in METH
users (13).
METH is associated with several socioeconomic and behav-
ioral risk factors that may predispose individuals to MRSA SSTI
(11). Notably, even noninjection use is associated with MRSA
SSTI, as smoking METH is an independent risk factor for MRSA,
including primary skin abscesses and invasive infections (13, 14).
In addition, skin picking is associated with MRSA SSTI. METH
use causes formication, a sensation of something crawling on the
body or under the skin, which can lead to skin-picking behavior
and skin breakdown (10, 15). Furthermore, METH injection is
also associated with the introduction of bacteria into the skin,
especially if the user fails to clean injection sites or shares drug
paraphernalia (16).
The effects of METH on host responses have not been exten-
sively described. However, limited studies about the effects of
METH on immune function show that its abuse has profound
implications for host immunity. The injection of 25 mg/kg of body
weight of METH into rats induces apoptotic death in thymic and
splenic lymphocytes (17). METH also reduces thymic and splenic
cellularity and alters peripheral T lymphocyte populations in
treated mice (18). METH exposure results in mitochondrial oxi-
dative damage and causes dysfunction of primary human T cells
(19). Also, METH is an immunosuppressive agent; it alkalizes
normally acidic organelles within immune cells, inhibits antigen
presentation, and impairs phagocytosis (20). Moreover, METH
negatively alters antibody and cytokine production (21).
Although there is a clear clinical association of METH use and
MRSA disease, there has not been a defined biological link be-
tween the immune response and wound healing capacity of a
METH user with an increased susceptibility to S. aureus. However,
animal studies demonstrate that METH suppresses both innate
and adaptive immunity (22, 23) and alters immune cell gene ex-
pression (24). Here, we investigated whether METH facilitates
MRSA skin infections and provide evidence that the drug has neg-
ative effects on human-derived phagocytic cells.
RESULTS
METH alters wound healing. METH administration decreased
the wound healing rate significantly compared with results in un-
treated animals (Fig. 1A). Moreover, METH administration and
FIG 1 METH decreases wound healing in mice infected with MRSA. (A) Wounds of BALB/c mice (n5 per group) uninfected and untreated (untreated),
infected with MRSA, treated with methamphetamine (METH), or treated with METH and infected with MRSA (METH MRSA), at days 3, 5, 7, 9, 11, 13, 15,
and 17. Bar, 5 mm. (B) Wound size analysis of BALB/c mouse skin lesions. Wounds were uninfected and untreated, infected with MRSA, treated with METH,
or treated with METH and infected with MRSA. Symbols are the averages of the results for six measurements at each time interval, and error bars denote standard
deviations. Pvalues of 0.05 were considered significant, calculated by ANOVA.
,
, and
indicate significantly higher eschar sizes in METH MRSA mice
than sizes for untreated, MRSA-infected, or METH-treated groups, respectively. This experiment was performed twice and similar results were obtained. The
results shown are representative of an individual experiment.
Mihu et al.
2®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
MRSA infection (METH-MRSA) induced visible inflammation
and decreased wound healing compared with all other conditions
(Fig. 1A). In fact, the wounds in infected or METH-treated mice
demonstrated a significant increase in size relative to the initial
wound, which did not occur in the untreated, uninfected animals.
At day 3, eschars in untreated wounds were ~4.4 mm in diameter,
whereas eschars of animals treated with METH alone or untreated
MRSA-infected or METH-MRSA wounds were ~7 mm (P
0.001, compared with untreated). At day 7 (Fig. 1B), eschars in the
untreated group were ~2.4 mm, whereas the eschars of wounds in
the MRSA, METH, or METH-MRSA treatment groups were ~3.9
mm (P0.05), ~6.5 mm (P0.001), and ~6.9 mm (P0.001),
respectively. At day 13 (Fig. 1B), the wounds of untreated mice
showed complete closure, whereas eschars of MRSA, METH, or
METH-MRSA wounds were ~1.7 mm (P0.01), ~2.1 mm (P
0.01), and ~4.9 mm (P0.001), respectively. Complete wound
closure was reached by day 17 for MRSA- or METH-treated mice,
whereas complete wound healing in the METH-MRSA group
took 19 days. Mice treated with METH or MRSA alone had similar
resolution of wounds during days 9 to 17, although METH-
exposed mice had slower healing at day 7.
METH enhances MRSA burden. Qualitative histological ex-
aminations revealed that wounds of untreated, uninfected mice
and of METH-treated uninfected mice had less inflammation
along with increased fibrin deposition than MRSA-infected tis-
sues, and no evidence of bacteria (Fig. 2A). MRSA-infected mice
showed localized epidermal inflammation (Fig. 2A). However,
wounds of METH-treated MRSA-infected mice had intense in-
flammatory infiltrates in both the epidermal and dermal layers,
along with extensive cell necrosis (Fig. 2A). Tissue Gram stains of
MRSA-infected and of METH-treated MRSA-infected samples
displayed large numbers of Gram-positive cocci (Fig. 2A, insets),
with larger bacterial clusters in biofilm-like arrangements evident
in the sections from METH-treated animals.
MRSA-infected mice treated with METH had significantly
higher, by 2 logs, microbial burdens than untreated MRSA-
infected mice at day 7 postinfection (P0.05) (Fig. 2B). Less than
30 bacterial CFU were detected in the skin of uninfected mice, and
MRSA was not identified.
METH impairs wound healing by mediating host matrix
metalloproteinase-2 collagen degradation. The mechanisms
through which METH impairs wound healing were explored by
examining whether METH decreased collagen deposition in
wound tissue (Fig. 3). Collagen content, in both the epidermal and
dermal layers, was lowest in METH-treated MRSA-infected
wounds (Fig. 3A). The lack of blue stain indicated lower tissue
collagen formation in METH-treated MRSA-infected wounds,
suggesting that METH administration augmented MRSA dermal
collagen degradation. Figure 3B presents the results of a morpho-
metric analysis of the data shown in Fig. 3A.
To determine whether METH affects collagen deposition dur-
ing wound healing, we assessed mRNA expression of collagen type
I and III in murine wound tissue (Fig. 3C). METH-treated MRSA-
infected samples had significantly increased mRNA collagen type
I and III expression, ~18-fold higher, compared to untreated, un-
infected control tissues (P0.001). Notably, tissues from MRSA-
infected mice had an 8-fold increase in collagen type III expression
compared to tissues from untreated, uninfected animals (P
0.001) (Fig. 3C), but there was no difference in collagen I expres-
sion.
S. aureus staphopains are cysteine proteases involved in tissue
destruction and collagen degradation. Therefore, we examined
whether METH enhances collagen degradation by staphopain A
(Fig. 3D). Surprisingly, increased concentrations of METH signif-
icantly reduced the degradation of collagen by staphopain A (P
0.05). Similarly, untreated and METH-treated heat-inactivated
staphopain A showed lower collagen degradation than active pro-
tease.
FIG 2 METH enhances MRSA burden in superficial skin lesions. (A) Histological analysis of BALB/c mice uninfected and untreated (untreated), infected with
MRSA (MRSA), treated with METH (METH), or treated with METH and infected with MRSA (METH MRSA), at day 7. Mice were infected with 10
7
MRSA
bacterial cells. Representative H&E-stained sections of the skin lesions are shown. Bars, 25
m. The insets depict Gram stain results for the boxed areas, and the
panels reveal MRSA cells (shown in purple). The arrows in these insets indicate bacterial clusters in biofilm-like arrangements. (B) Wound bacterial burdens
(CFU) in METH-treated mice infected with 10
7
MRSA cells were significantly higher than those in MRSA-infected, untreated mice at 7 days after infection (n
10 per group). Each symbol represents one animal; dashed bars are the averages for each group, and error bars denote standard deviations. Asterisks denote
Pvalues of 0.05, calculated by using Student’s ttest. These experiments were performed twice, and similar results were obtained. The results shown are
representative of an individual experiment.
Methamphetamine Exacerbates MRSA Skin Infection
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 3
To define the host’s contribution in collagen degradation after
METH administration and MRSA infection, we analyzed the ex-
pression of matrix metalloproteinase-2 (MMP-2) in mouse
wounded tissue (Fig. 3E). METH-treated or METH-treated,
MRSA-infected tissues had significantly increased mRNA MMP-2
expression levels, by ~4- and 6-fold, respectively, above those in
untreated, uninfected control tissues (P0.001). Then, we inves-
tigated whether METH increased collagen degradation by MMP-2
(Fig. 3F). METH exacerbated collagen degradation by MMP-2
relative to that in untreated tissue (25
M[P0.05]; 50
M[P
0.001]). No differences were observed between untreated tissues
and those in the METH treatment group that received heat-
inactivated MMP-2. Western blot analysis was performed to con-
firm the upregulation of MMP-2 protein levels in METH-treated
tissues (Fig. 3G). We found that METH increased the expression
of MMP-2 in both uninfected and MRSA-infected wounds. To-
FIG 3 METH impairs wound healing by enhancing the host’s MMP-2-mediated collagen degradation. (A) Histological analysis of untreated, MRSA, METH,
or METH MRSA treatment groups of BALB/c mice at day 7 after wounding. The blue stain indicates collagen. Bar, 25
m. (B) Quantitative measurement of
collagen intensity in tissues from untreated, MRSA, METH, and METH MRSA mice were measured using ImageJ software. Bars are the averages of the results,
and error bars denote standard deviations. *, #, and & indicate significantly lower collagen intensites than in untreated, MRSA-infected, or METH-treated groups,
respectively. (C) Gene expression analysis of collagen type I and III in cutaneous lesions.
,
, and
indicate significantly higher fold changes than in untreated,
MRSA-infected, or METH-treated groups, respectively. (D) Collagen degradation by MRSA staphopain A. FITC-conjugated type I collagen was incubated with
10
M staphopain A and METH (25 or 50
M)for3hat37°C. The fluorescence of the supernatant was measured. Heat-inactivated staphopain A was utilized
as a control. Dashed lines represent the averages for three measurements per condition, and error bars denote standard deviations. *, #, and & indicate
significantly lower relative fluorescence than in the untreated or 25
M METH groups after cells were incubated with active staphopain A, respectively. (E) Gene
expression analysis of MMP-2 in murine wounds.
,
, and
indicate significantly higher fold changes than in untreated, MRSA-infected, or METH-treated
groups, respectively. (F) Collagen degradation by MMP-2. FITC-conjugated type I collagen was incubated with 10
M MMP-2 and METH (25 or 50
M) for 3 h
at 37°C.
and
indicate significantly higher relative fluorescence values than in the untreated or 25
M METH groups, respectively. *, #, and & indicate
significantly lower relative fluorescence than in cells from untreated or the 25 or 50
M METH groups incubated with active MMP-2, respectively. (G) Western
blot analysis of MMP-2 in wounded tissue (7 days) of untreated, MRSA, METH, or METH MRSA treatment in BALB/c mice. GAPDH was used as a loading
control. For panels B to F, Pvalues (with significance defined as P0.05) were calculated by using an ANOVA. All the experiments in the figure were performed
twice, and similar results were obtained. The results shown are representative of an individual experiment.
Mihu et al.
4®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
gether, these results suggest that wound healing in METH-treated
animals is reduced, at least in part, by an increase in the host’s
MMP-2 levels.
METH decreases the number of phagocytic cells in the blood
of treated BALB/c mice. We examined whether METH adminis-
tration depleted phagocytes in the blood of BALB/c mice by using
differential leukocyte staining. Light microscopy images showed
reduced numbers of phagocytic cells in the blood of METH-
treated mice compared with numbers in untreated mice (Fig. 4A).
Cell count analysis showed that METH-treated animals had sig-
nificantly lower numbers of circulating phagocytes in blood com-
pared to numbers in controls (P0.05) (Fig. 4B).
METH does not inhibit S. aureus growth. MRSA growth with
and without exposure to METH was determined in real time for
24 h (Fig. 5). METH did not alter bacterial growth during a 24-h
coincubation with 25 or 50
M METH compared with untreated
S. aureus controls.
METH enhances S. aureus biofilm formation. We evaluated
the effect of METH on biofilm formation by six clinical S. aureus
isolates to confirm the results obtained in the murine model. S. aureus
biofilms on 96-well plates were incubated with METH (25 or 50
M)
or phosphate-buffered saline (PBS) for 24 h. Cell viability and meta-
bolic activity were determined by CFU and 2,3-bis(2-methoxy-4-
nitro-5-sulfophenyl)-5-[(phenylamino)carbonyl]-2H-tetrazolium
hydroxide (XTT) reduction assays, respectively (Fig. 6A and B). On
average, METH promoted S. aureus biofilm formation compared to
untreated controls. S. aureus biofilms incubated with METH dis-
played higher CFU counts (P0.0001) (Fig. 6A), and cells within
biofilms were more metabolically active (P0.0001) (Fig. 6B) than
untreated controls.
Confocal microscopic examination was used to correlate the
decreases in CFU and the XTT reduction assay results with the
visual effects on biofilm structure (Fig. 6C). In the figure, regions
of green fluorescence represent viable cells; the red fluorescence
indicates metabolically inactive or nonviable cells. Untreated
MRSA strain 6498 biofilms were ~21
m thick, whereas biofilms
grown in the presence of 25
M METH showed a more robust
architecture with a thickness of ~71
m (Fig. 6C). The METH-
treated biofilm metabolic activity observed corresponded with the
XTT results.
The S. aureus gene ica encodes an intercellular adhesion pro-
tein, and disruption of this gene leads to decreased biofilm forma-
tion (25). All isolates in the present study were found to carry the
ica gene (data not shown). Hence, we investigated the impact of
METH on the expression of the ica gene by biofilm-associated
cells via quantitative reverse transcription-PCR (qRT-PCR)
(Fig. 6D). S. aureus biofilm-associated cells exhibited significantly
increased ica expression after exposure to METH (25
M[P
0.0001] and 50
M[P0.0001]).
METH affects murine neutrophil functions. We investigated
the effect of METH on murine neutrophil effector functions in
MRSA infection. METH significantly inhibited neutrophil migra-
tion (25
M, P0.05; 50
M, P0.01) in a concentration-
dependent manner compared to untreated or chloroquine
(Chlq)-treated cells (Fig. 7A). As expected, cytochalasin D
(CytD)-treated cells (25
M) displayed a significant reduction in
migration (P0.01) compared to untreated cells.
We examined whether METH interfered with neutrophil-
mediated killing (NMK) of MRSA strain 6498. When untreated
neutrophils and MRSA 6498 were coincubated, the majority of
bacteria were killed within 80 min (Fig. 7B). Similarly, Chlq-
treated neutrophils showed an increase in bacterial killing over
time, albeit to a lesser extent than untreated neutrophils. In con-
trast, METH more significantly impaired bacterial killing, in a
concentration-dependent manner (P0.05, 60 and 80 min) by
stationary neutrophils, and likewise, CytD interfered with phago-
cytic killing (Fig. 7B).
Confocal microscopic examination was used to correlate the
killing assay results with the visual effects on the impact of METH
on neutrophil morphology (red fluorescence [actin]) and phago-
cytosis of MRSA (blue fluorescence [Alexa-labeled S. aureus])
(Fig. 7C). METH-treated neutrophils (25 and 50
M; P0.05)
and CytD-treated neutrophils (25
M; P0.05) significantly
FIG 4 METH-treated animals displayed fewer phagocytes in blood. (A) Light microscopy images of blood smears from untreated or METH-treated mice
preinfection. Arrows indicate phagocytic cells (purple). Bar, 20
m. (B) Numbers of phagocytic cells per field in blood of untreated or METH-treated mice. Each
black circle or red square represents the number of neutrophils per individual field. Dashed lines and error bars denote averages and standard deviations of 40
counts, respectively. Asterisks denote significant Pvalues (P0.05), calculated using Student’s ttest. The experiment was performed twice with similar results
obtained. The results shown are a combination of two independent experiments.
FIG 5 METH does not affect S. aureus viability in vitro. The effect of METH
on S. aureus growth kinetics was determined via Bioscreen C analysis. S. aureus
was grown in the absence (untreated) or presence of METH (25 or 50
M).
Each point represents the average of three measurements. The experiment was
performed twice with similar results obtained. The results shown are represen-
tative of an individual experiment.
Methamphetamine Exacerbates MRSA Skin Infection
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 5
reduced phagocytosis of MRSA, compared to that of untreated or
Chlq-treated cells 30 min after coincubation (Fig. 7C). Untreated
neutrophils displayed multiple protrusions and actin polymeriza-
tion, engaged in phagocytic and killing interactions with S. aureus
(Fig. 7D), and contained an increased number of internalized bac-
teria. In contrast, METH- or CytD-treated neutrophils displayed a
large rounded morphology, fewer protrusions, and reduced inter-
actions with bacteria, and the cells were surrounded by multiple
bacterial cells (Fig. 7D).
Neutrophils treated with METH (50
M; P0.001) or CytD
(25
M; P0.001) showed significantly lower levels of NO pro-
duction after MRSA infection than did untreated neutrophils
(Fig. 7E). Both METH (25
M) and Chlq (25
M) showed a trend
toward a reduction in NO production. Similarly, the release of
myeloperoxidase (MPO) by neutrophils was significantly de-
creased by METH (25
M; P0.01; 50
M, P0.001) or CytD
(25
M; P0.001) (Fig. 7F).
METH impairs the effector functions of human neutrophils.
We analyzed the effects of achievable physiological METH doses
on phagocytosis and killing of six clinical S. aureus isolates by
human neutrophils via fluorescence-activated cell sorting (FACS)
analysis. METH significantly reduced phagocytosis of S. aureus by
neutrophils, compared with the untreated or Chlqtreated (25
M
Chlq) cells (25
M METH, P0.01; 50
M METH, P0.01;
compared to untreated and Chlq groups) (Fig. 8A). Our results
showed 52.5% and 46% S. aureus phagocytosis by cells treated
with METH (25 or 50
M) compared to untreated (77%) or 25
M Chlq-treated cells (75.5%). As expected, CytD-treated cells
displayed a significant reduction in phagocytosis compared to
cells in all the other groups, with the exception of 50
M METH
(P0.001).
We again also examined whether METH interfered with
neutrophil-mediated killing of S. aureus cells. CytD and METH
significantly reduced bacterial killing by human neutrophils (P
0.0001) compared to untreated cells (Fig. 8B). These phagocytic
cells mostly kill bacteria via NADPH oxidase-derived reactive ox-
ygen species (ROS). As a result, we evaluated the impact of METH
on the neutrophil oxidative burst by measuring luminol chemilu-
minescence intensity. METH significantly decreased ROS pro-
duction compared to untreated or Chlq-treated, S. aureus-
exposed neutrophils (Fig. 8C) (P0.05; 25 to 60 min). Minimal
production of ROS was observed in untreated and unstimulated
controls or CytD-treated cells (P0.05; 30 to 60 min).
METH alters macrophage function. We identified macrophage-
like infiltration in wound tissue by measuring the expression of
Iba-1, which is mostly expressed and upregulated during the acti-
vation of these cells. Tissue sections from murine wounds in the
METH-treated (uninfected) group showed only scattered
macrophage-like cells, whereas untreated or MRSA-infected mice
displayed massive macrophage-like cell infiltrations throughout
the wound area (Fig. 9A). Additionally, METH-treated MRSA-
FIG 6 METH promotes biofilm formation by S. aureus clinical isolates (n6), based on CFU counts (A) and XTT reduction (B) after 24 h. Dashed lines and
error bars represent the averages and standard deviations of six measurements (each symbol represents a single strain) from three plates per well per strain. (C)
Confocal microscopy of MRSA 6498 biofilms after treatment with METH. Representative three-dimensional images of biofilms show viable (green [SYTO 9])
and dead (red [propidium iodide]) cells. The thickness and morphology of the bacterial biofilms can be observed in the Z-stack reconstruction. The pictures were
taken at a magnification of 63. Bar, 50
m. (D) Expression analysis of the ica gene in S. aureus strains after incubation with METH. For panels A, B, and D,
significance (P0.05) was calculated by ANOVA.
indicates significantly higher fold change than untreated group. These experiments were performed twice
with similar results obtained. The results shown are representative of an individual experiment.
Mihu et al.
6®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
infected wounds demonstrated minimal macrophage-like cell in-
filtration.
METH (25
M,P0.05; 50
M, P0.01) significantly in-
hibited macrophage migration in a concentration-dependent
manner compared to untreated or Chlq-treated cells (Fig. 9B).
CytD-treated cells (25
M; P0.01) displayed a significant re-
duction compared to untreated cells.
METH (25 and 50
M) significantly stimulated phagocytosis
of bacterial cells by murine macrophages, compared with the un-
treated, Chlqtreated (25
M), or CytD-treated (25
M) cells (P
0.01 for 25
M METH; P0.001 for 50
M METH) (Fig. 9C).
Furthermore, METH decreased the eradication of bacteria within
macrophages (Fig. 9D).
After phagocytosis, macrophages produce NO in response to
pathogens within the phagosome. Macrophages treated with
METH (50
M; P0.001) or CytD (25
M; P0.001) showed
significantly lower levels of NO production after infection with
MRSA strain 6498 than did untreated macrophages (Fig. 9E).
Both the METH (25
M) and Chlq (25
M) treatment groups
showed a trend toward a decrease in NO production.
Phagosome acidification was studied to elucidate the mecha-
nism by which METH interferes with MRSA 6498 killing after
phagocytosis. The acidification of phagosomes of primary macro-
phages containing MRSA labeled with pH-sensitive and pH-
insensitive probes was measured using a spectrofluorimeter. Stan-
dard curves were generated with fluorophores. METH induced
the alkalization of macrophage phagosomes that contained MRSA
6498 cells. The pH of phagosomes in macrophages infected with
FIG 7 METH modifies neutrophil effector functions. (A) Percentages of neutrophils (polymorphonuclear lymphocytes [PMNs]) migrating at various time
points (20, 40, and 60 min) after exposure of uninfected cells to the chemoattractant fMLP. Neutrophils were isolated from BALB/c mice and treated with PBS
(untreated), Chlq (25
M), CytD (25
M), or METH (25 or 50
M) and coincubated with MRSA. (B and C) METH interferes with neutrophil-mediated killing
(NMK) (B) and phagocytosis of MRSA cells (C). For panel C, fluorescence intensity (in arbitrary units [A.U.]) refers to blue-labeled bacteria inside neutrophils
after their phagocytosis. (D) Confocal microscopy of untreated or CytD- or METH-treated neutrophils interacting with MRSA. Alexa Fluor 350 (blue) was used
to label bacterial cells. Actin-specific (red) staining was used to label cell bodies of neutrophils. 4=,6-Diamidino-2-phenylindole (blue) was used to label the nuclei.
Bar, 10
m. (E) NO production in infected neutrophils was quantified using the Griess method. (F) MPO released by neutrophils was measured using the
EnzChek MPO activity assay. For panels A to C and E to F, the values presented are averages and standard deviations. All experiments were performed three times
with similar results obtained. Each point or bar represents the average of three measurements per condition. The results shown are representative of an individual
experiment. Significance (P0.05) was calculated by an ANOVA. *, #, and & indicate results significantly lower than in untreated, Chlq, or 25
M METH-
treated groups, respectively.
Methamphetamine Exacerbates MRSA Skin Infection
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 7
the bacteria for 2 h was analyzed. In macrophages treated with
METH (25 or 50
M) or Chlq (25
M), the phagosomal pH was
significantly elevated (pH 5.6, 6.4, and 6.8, respectively) compared
with the pH in untreated control or CytD-treated (25
M) mac-
rophages (pH 4.8 and 4.4; P0.001) (Fig. 9F).
METH-treated MRSA-infected mice displayed decrease an-
giogenesis. We investigated the effect of METH on angiogenesis
in wounded skin infected with MRSA by measuring the expression
of CD34 (Fig. 10). Tissue sections from untreated or METH-
treated uninfected murine wounds displayed dense vasculariza-
tion, whereas wounds of MRSA-infected mice showed localized
formation of blood vessels in dermal tissue. Moreover, METH-
treated MRSA-infected wounds demonstrated minimal vessel for-
mation in the epidermis and dermis. Therefore, the combination
of METH and MRSA infection visually reduces vascularization in
murine cutaneous wounds, and this qualitative observation is as-
sociated with decreased healing.
METH alters cytokine expression. Twenty-four hours after
infection, wound tissue of mice infected with MRSA contained
significantly higher quantities of gamma interferon (IFN-
),
interleukin-1
(IL-1
), IL-12, monocyte chemoattractant pro-
tein 1 (MCP-1), and tumor necrosis factor alpha (TNF-
) than
those under all other conditions (Table 1). Transforming growth
factor
(TGF-
) was significantly elevated in MRSA-infected
mice (MRSA and METH-MRSA) groups. There was a significant
increase in IL-6 and IL-10 production in METH-treated animals
(METH and METH-MRSA) compared to untreated conditions
(uninfected or MRSA groups). There were significant decreases in
vascular endothelial growth factor (VEGF) and TNF-
for the
MRSA and METH-MRSA groups, respectively, compared to all
other conditions. IL-6 and TGF-
levels were significantly re-
duced in untreated animals relative to the other groups. There
were no differences in IL-4 production between any of the exper-
imental groups.
At day 7 after infection, wound tissue of METH-treated mice
infected with MRSA contained significantly higher quantities of
IFN-
, TNF-
, IL-4, and TGF-
than tissues of uninfected or
MRSA-treated mice (Table 2). There was a significant increase in
IL-12 and IL-10 production in MRSA-infected animals compared
to all the other treatment groups. Significant increases in IL-6 and
IL-1
and decreases in VEGF were measured under all the other
conditions, compared to results in uninfected mice. MCP-1 and
VEGF levels were significantly reduced in MRSA-treated animals
relative to the other groups.
DISCUSSION
We used a murine model to investigate the effects of METH on
MRSA superficial skin infection. Our findings showed that
METH-treated mice displayed reduced wound healing in the
presence or absence of MRSA infection compared with untreated,
FIG 8 METH reduces human neutrophil phagocytosis, respiratory burst, and killing of S. aureus. (A) Phagocytosis (percentage) of FITC-labeled S. aureus
clinical strains (n6) by human neutrophils was determined using FACS analysis after a 60-min incubation with METH. (B) Killing of S. aureus by neutrophils
was determined from CFU counts. For panels A and B, dashed lines and error bars represent the averages and standard deviation of six measurements (each
symbol represents a single strain). Significance (P0.05) was calculated by an ANOVA and adjusted by use of the Bonferroni correction. For panel A, *, #, and
& indicate a lower phagocytosis percentage than in untreated, 25
M Chlq, or 25
M METH groups, respectively. For panel B,
,
, @, and $ indicate a higher
survival percentage than in untreated, 25
M Chlq, 25
M METH, and 50
M METH groups, respectively. (C) Oxidative burst was quantified for 60 min based
on luminol chemiluminescence after untreated or Chlq-, CytD-, or METH-treated neutrophils were coincubated with MRSA strain 6498. Untreated and
uninfected neutrophils were also used as controls. Symbols and error bars denote means and standard deviations. Significance (P0.05) was calculated by an
ANOVA and adjusted by use of the Bonferroni correction at each time point. This experiment was performed twice and similar results were obtained. The results
shown are representative of an individual experiment.
Mihu et al.
8®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
uninfected mice, with the longest delay in wound closure occur-
ring in the METH-MRSA group. In this regard, our results from
the measurement of collagen deposition in wound tissue revealed
increased collagen degradation in the METH-MRSA treatment
group compared with the controls. Collagen plays a crucial role in
the proliferative phase of the wound healing process, along with
angiogenesis, granulation tissue formation, and epithelialization
(26). The amount of collagen present in the wound during the
healing process reflects a balance between new collagen produc-
tion by fibroblasts and degradation by collagenases and other fac-
tors. In the early phases of healing of simple, uninfected wounds,
synthesis exceeds degradation, so collagen levels in the wound rise;
later, production and degradation become equal (26). This bal-
ance was disrupted by METH and MRSA. The expression levels of
genes regulating collagen type I and III production were found to
be significantly increased in the METH-MRSA group as well;
however, the accelerated degradation process led to reduced col-
lagen content in the wound and thus impaired healing. Notably,
another contributing factor to the delayed wound closure was the
decreased angiogenesis under the influence of METH-MRSA, as
qualitatively demonstrated by the reduced expression of CD34, a
marker for blood vessel formation. For instance, histological anal-
yses of tissues from METH-MRSA mice showed high infiltration
of inflammatory cells confined to the dermal area of the cutaneous
tissue, but the cells were largely unable to reach the epidermal
layer, where most of the bacteria were located. This observation
FIG 9 METH alters macrophage effector functions. (A) Histological analysis of untreated, MRSA, METH, and METH-MRSA wounded BALB/c mice at day 7.
The brown staining indicates macrophage infiltration. Representative Iba1-immunostained sections of the skin lesions are shown. Bars, 25
m. (B to F) Primary
macrophages were isolated from BALB/c mice and treated with PBS (untreated), Chlq (25
M), CytD (25
M), or METH (25 or 50
M) and coincubated with
MRSA (C to F). (B) Uninfected macrophage migration (as a percentage) after exposure to the chemoattractant fMLP. Significance (P0.05) was calculated by
an ANOVA. *, #, and & indicate a significantly lower percentage of macrophages than in untreated or Chlq- or 25
M METH-treated groups, respectively. (C)
Phagocytosis (percentage of cells) was analyzed by FACS analysis after a 30-min incubation. (D) CFU determinations were performed after the 30-min
incubation. (E) NO production was quantified using the Griess method. Significance (P0.05) was calculated by an ANOVA. Asterisks indicate results
significantly lower than in untreated groups. (F) The pH levels in phagosomes of primary macrophages that contained MRSA labeled with pH-sensitive and
pH-insensitive probes were measured using a spectrofluorimeter. For panels B to F, the values presented are averages and standard deviations. For panels C, D,
and F, Significance (P0.05) was calculated by an ANOVA.
,
,
, and
indicate a significant increase compared to results in untreated or Chlq-, CytD-, or
25
M METH-treated groups, respectively. All the experiments were performed twice with similar results obtained. Each bar represents the average of three
measurements per condition. The results shown are representative of an individual experiment.
Methamphetamine Exacerbates MRSA Skin Infection
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 9
may explain the 2-log-higher bacterial counts in the skin samples
of the METH-treated group relative to the untreated mice. Like-
wise, in vitro and in vivo studies demonstrated that METH impairs
phagocytic cell migration and antimicrobial activity as well as re-
duces phagocytes in the bloodstream. The prolonged presence of
an elevated number of macrophages and neutrophils in the skin
tissue may be associated with increased free radical accumulation
(27), which can inhibit wound healing.
We investigated whether the difference in collagen deposition
was due to METH-induced bacterial or host breakdown of colla-
gen. We originally thought that S. aureus staphopain collageno-
lytic activity might be implicated in cutaneous tissue destruction
in the setting of METH exposure (28). However, in vitro experi-
ments determined that these bacterial cysteine proteases did not
contribute to collagen breakdown. We cannot completely rule out
the possibility that other virulence factors produced by S. aureus
may be detrimental for the healing process. More research is
needed to better identify specific MRSA virulence factors involved
in dysregulating wound healing in the setting of drug abuse.
However, we successfully identified a host factor involved in
collagen degradation after METH administration and MRSA in-
fection. METH increased the expression of MMP-2, a metallopro-
tease that can interfere with wound healing and enhance suscep-
tibility to bacterial infections (29, 30). In HIV-associated
dementia, the HIV-1-encoded protein Tat and METH increase
MMP-2 levels in supernatants of human neuron/astrocyte cul-
tures (31). Previous studies have shown that MMP-2 plays crucial
roles in METH-induced behavioral sensitization and reward by
regulating METH-induced dopamine release and uptake via do-
pamine transporters in the brain (32). Dopamine has been re-
TABLE 1 Cytokine levels in wounds of BALB/c mice 24 h after wounding
Treatment
Cytokine level
a
(pg/ml) (avg SD)
IFN-
IL-1
IL-4 IL-6 IL-10 IL-12 MCP-1 TGF-
TNF-
VEGF
Untreated 1,951.74
16.62
791.80
11.57
81.25
10.74
452.93
8.91
398.26
6.61
367.91
6.45
374.08
11.13
1,996.55
5.02
3,852.01
15.12
4,623.52
15.09
MRSA 2,339.10
10.65
859.25
6.47
91.525.32 614.37
4.63
321.83
7.36*
414.5
35.37
421.54
4.02
2,459.32
4.46
4,157.19
8.12
2,071.90
23.61*
METH 1,398.46
5.68
*#
645.32&#
x00B1;6.04
*#
75.265.97 897.25
7.98
␾␹
497.21
6.54
␾␹
362.83
8.81
#
200.87
9.09*
#
2,085.31
16.25
#
2,736.14
11.95
*#
3,027.11
6.72
*
METH-MRSA 1,765.44
4.21
*#
701.33
9.26
*#
79.69
5.97
811.85
8.23
␾␹
&
505.21
9.22
␾␹
388.24
8.65
#
398.08
7.12
#
2,369.84
6.35
1,977.25
12.81
#&
2,545.15
7.01
*
␹␲
a
,
, and
indicate a significantly higher cytokine level (P0.05) than in the untreated, MRSA, or METH group, respectively. *, #, and & indicate a significantly lower cytokine
level (P0.05) than in the untreated, MRSA, or METH group, respectively.
TABLE 2 Cytokine levels in wounds of BALB/c mice 7 days after wounding
Treatment
Cytokine level
a
(pg/ml) (avg SD)
IFN-
IL-1
IL-4 IL-6 IL-10 IL-12 MCP-1 TGF-
TNF-
VEGF
Untreated 2,321.58
15.84
2,598.36
10.89
251.26
10.28
2,245.35
8.65
474.36
13.47
936.32
6.47
4,932.21
10.58
10,365.9
5.47
1,901.26
14.74
1,5369.7
16.56
MRSA 4,531.08
9.45
3,426.65
7.21
384.08
5.05
5,057.15
9.14
4,197.85
7.05
1,682.09
12.75
1,173.07
21.65*
16,658.95
16.71
1,383.62
7.81*
6,312.28
12.43*
METH 5,987.21
6.01
␾␹
3,501.98
5.97
1,223.36
11.71
8,569.61
12.58
␾␹
625.36
6.99
#
1,256.12
8.58
#
4,864.36
9.71
16,723.1
13.89
3,821.36
11.32
␾␹
1,2154.7
10.06
*
METH-MRSA 15,806.1
4.66
␾␹␲
3,568.59
8.17
1,462.59
6.18
␾␹␲
9,841.99
15.04
␾␹␲
592.4
9.54
#
1,145.13
9.16
#
5,215.42
19.14
32,454.7
22.27
␾␹␲
5,121.88
5.81
␾␹␲
1,0172.2
9.28
*
&
a
,
, and
indicate a significantly higher cytokine level (P0.05) than in the untreated, MRSA, or METH group, respectively. *, #, and & indicate a significantly lower cytokine
level (P0.05) than in the untreated, MRSA, or METH group, respectively.
FIG 10 METH-treated MRSA-infected mice displayed decreased angiogenesis in wounded skin. Tissue sections are shown for untreated, MRSA, METH, or
METH-MRSA wounded BALB/c mice at day 7. Brown staining indicates vascularization. Representative CD34-immunostained sections of the skin lesions are
shown. Black arrows denote dense vascularization. Bars, 25
m.
Mihu et al.
10 ®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
ported to decrease mitosis of keratinocytes (33), the most abun-
dant cells in skin tissue, whereas D2-like receptors (D2, D3, and
D4) are involved in epidermal barrier homeostasis (34). For in-
stance, the D2 receptor is present in the basal epidermis, and the
D4 receptor is in the uppermost layer of the epidermis (34). These
receptors have been implicated in the pathogenesis of skin inflam-
matory diseases (35, 36) and regulation of wound healing (37, 38).
Although further studies are warranted, METH may induce do-
pamine production and D2-like receptor expression by keratino-
cytes, which could substantially reduce the wound healing rate.
Additionally, METH promoted the collagenolytic activity of
MMP-2 in a concentration-dependent manner, providing an-
other possible explanation for the negative impact of this drug in
wound healing. The late (day 7 postinjury) augmented produc-
tion of inflammatory cytokines like TNF-
, IFN-
, IL-6, and
TGF-
observed in METH-treated groups promotes the activa-
tion of MMP-2 via NF-
B signaling in dermal fibroblasts embed-
ded in collagen type I (39). Our study is important because self-
management of injection-related wounds is common among
injection drug users (IDUs) (11). Specifically, IDUs are more
likely to engage in potentially harmful self-management behav-
iors, increasing their susceptibility to MRSA infections due to a
combination of aggressive management of their wounds and an
underlying slow healing rate. In a previous study, IDUs who had
ever injected amphetamines were more likely to engage in poten-
tially harmful self-management behaviors (11). Furthermore, the
inability of phagocytic cells to be recruited and/or proactively fight
invading microbes at the site of infection could exacerbate disease
in IDUs.
Another important finding of this study is the increase in in-
flammation and cell necrosis in the wounds of METH-treated and
infected mice compared with control groups. For instance, CFU
analysis showed an increased bacterial burden in the wound tis-
sues in the METH-MRSA group. Chronic wounds on METH us-
ers are an ideal environment for S. aureus biofilm formation. The
necrotic tissue and debris allow bacterial attachment, and wounds
are susceptible to infection due to an impaired host immune re-
sponse. Evidence suggests that biofilms play a significant role in
the inability of chronic wounds to heal. Biofilms are present in
only 6% of acute wounds but over 90% of chronic wounds (40).
We found that METH enhanced biofilm formation by multiple
clinical S. aureus strains. The presence and persistence of biofilms
on chronic skin wounds can affect cellular function (leukocytes,
keratinocytes, endothelial cells, and fibroblasts), the inflamma-
tory cellular response, the cutaneous innate immune response,
and the repair phase of wound healing (angiogenesis and fibro-
genesis). For example, keratinocytes produce more MMPs in
chronic biofilm-challenged wounds (41). These infections de-
velop gradually and may be slow to produce explicit symptoms. In
addition, the vast majority of bacteria reside in the eschar above
the wound bed (42). Once established, however, biofilm infec-
tions often persist. Understanding the impact of METH on S.
aureus pathogenesis in IDUs warrants the pursuit of further stud-
ies addressing the influence of the drug on the multitude of viru-
lence factors expressed by this microbe during infection, includ-
ing its toxins.
Another possible explanation for S. aureus persistence in
wounded tissue of drug users is that METH alters the effector
functions of neutrophils and macrophages. The initial and most
obvious function of inflammatory cells at the site of injury is to
provide specific and nonspecific defenses against pathogens. The
first cells that infiltrate a wound are neutrophils, which remove
foreign particles and bacteria. In the skin, neutrophils appear in
the wound bed within minutes after injury. We demonstrated that
METH inhibits neutrophil chemotaxis and the production of NO
and MPO in a concentration-dependent fashion, thus reducing
the antimicrobial armamentarium of activated murine neutro-
phils. Notably, METH compromised S. aureus phagocytosis, the
respiratory burst, and killing by human neutrophils, suggesting
that the detrimental effects of the drug observed in murine-
derived cells can be similarly achieved in human cells, which val-
idates the mouse as a good model for these types of studies.
METH-mediated phagocytosis dysfunction by neutrophils may
be associated with reduced expression of GTPase-RhoA, a key
regulator of the actin polymerization signaling cascade, given that
CytD-treated cells displayed a similar deficiency in engulfing the
microbe. Also, impaired phagocytosis may be associated with im-
mobilization of the complement receptor 3 (CR-3) on the surface
of neutrophils and deregulation of cytokines (21). Neutrophils are
nevertheless a major source of several inflammatory cytokines,
such as TNF-
and IL-1
, which stimulate attracted monocytes to
differentiate into M1 (classical activation) macrophages (43, 44).
We observed that METH reduces the early (24 h postinjury) pro-
duction of these two cytokines in METH-treated groups. More-
over, elevated levels of IL-10 in homogenates of METH-treated
groups during the early inflammatory response suggest an impor-
tant role of these factors in reducing the recruitment of phagocytic
cells to the site of injury and infection.
METH inhibited the migration of macrophages to the infec-
tion site as observed in vitro and in the histological analysis. Inter-
estingly, we found that primary macrophages exposed to METH
displayed increased MRSA phagocytosis; however, these macro-
phages showed impaired intracellular bacterial killing leading to
increased pathogen survival. As in our prior work, METH dis-
rupted macrophage effector functions (21), including alkalization
of the phagosomal pH and inhibition of NO and superoxide pro-
duction. One study using a mouse model of catheter-associated
biofilm infection showed that S. aureus biofilms caused changes in
macrophage function by suppressing microbicidal activity, alter-
ing gene expression toward M2 phenotype cells, decreasing migra-
tion, and increasing cell death (45). Those investigators showed
that macrophages were able to phagocytose planktonic cells but
not biofilm-associated cells, suggesting that the in vitro experi-
ments do not necessarily reflect in vivo situations. During the pro-
liferative phase, macrophages stimulate proliferation of connec-
tive, endothelial, and epithelial tissues directly and indirectly. In
particular, fibroblasts, keratinocytes, and endothelial cells are
stimulated by macrophages during this phase to induce and com-
plete extracellular matrix formation, reepithelialization, and neo-
vascularization. Once recruited to the wound, macrophages in-
duce angiogenesis. This is predominantly exerted by macrophages
releasing TNF-
and VEGF. TNF-
may in turn induce VEGF
expression by keratinocytes and fibroblasts (46). Depletion of
macrophages in the wound results in reduced vascularization (47)
and also leads to severe hemorrhage and fibrin and serum exu-
dates in macrophage-depleted granulation tissue (48). This has
been reported to be mainly caused by withdrawal of macrophage-
derived TGF-
and VEGF (47, 48), supporting an important role
for macrophages in promoting angiogenesis via their secretory
products.
Methamphetamine Exacerbates MRSA Skin Infection
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 11
METH alters cytokine expression, as indicated by our mea-
surements of cytokine levels in the wounds. The late (day 7 postin-
jury) increased levels of inflammatory cytokines in the METH-
MRSA group correlated with our histological observations of
increased local inflammation in their wounds. Nevertheless, the
disabled immune machinery in the METH-treated mice failed to
contain the infection, and the initial inflammatory cascade led to
detrimental local outcomes with increased cell necrosis and im-
paired healing. METH has been linked to a shift in the immune
response from a dominant Th1 to a mixed Th1-Th2 phenotype
(49). In our infection model, we found high levels of endogenous
TNF-
and IFN-
in METH-treated mice. However, the in-
creased levels of the two Th1 cytokines were not sufficient to res-
cue mice from the lethal effects of MRSA. Thus, METH might play
a role in inducing a dominant nonprotective Th2 response to
MRSA.
In conclusion, this is the first study that has empirically ad-
dressed the effect of METH on immunity during infection by S.
aureus. These findings should raise awareness of the negative im-
pact of METH use on the overall community health status, as
alarming data in this regard have been published on the high in-
cidence of MRSA skin and soft tissue infections among METH
consumers (13). Although wound debridement remains a proven
cornerstone for wound management, perhaps in part because a
microbial biofilm is removed from the scab, it is plausible to think
that wounds in METH users become chronic and difficult to man-
age because of a combination of bacterial persistence, drug-related
immune deficiency, and the IDU behaviors, which include stereo-
typical formication and lack of hygiene during injections, making
these individuals highly susceptible for S. aureus reinfection. A
possible limitation of the study is that the in vitro and in vivo
METH treatments were not equivalent, since in vitro the phago-
cytes were exposed for a few hours to METH, whereas in vivo they
were exposed for 21 days. However, METH doses used in both
animals and tissue cultures were similar or closely related to those
measured in people that chronically abuse the drug. Further re-
search is needed to understand the biological and management
complexity of infected wounds in METH users and to best focus
prevention efforts to reduce morbidity and improve wound care.
MATERIALS AND METHODS
S. aureus.A total of six S. aureus clinical isolates were used in this study.
The characteristics of each strain are described in Table 3. The MRSA 6498
isolate used in the majority of the experiments in this report is a USA300
strain collected from a patient’s wound and has been extensively utilized
in wound healing studies (49, 50). The strains were stored at 80°C in
brain heart infusion (BHI) broth with 40% glycerol until use.Test organ-
isms were grown in tryptic soy broth (TSB) overnight at 37°C on a rotary
shaker set at 150 rpm. Growth was monitored by measuring the optical
density at 600 nm and use of a microtiter plate reader.
METH. Most high-dose METH abusers initially use small amounts of
the drug intermittently before progressively increasing the dose (51). To
simulate this pattern, we used increasing daily doses (2.5, 5, and 10 mg/
kg/day on weeks 1, 2, and 3, respectively) of METH that were intraperi-
toneally (i.p.) administered to female BALB/c mice (6 to 8 weeks old) over
21 days, as described previously (21). As controls, mice received equiva-
lent volumes of PBS.
Wound and infection model. At day 21, METH- and PBS-treated
BALB/c mice were anesthetized (100 mg/kg ketamine, 10 mg/kg xylazine),
their back hair was removed, and skin was disinfected with iodine. Five-
millimeter-diameter full-thickness excision wounds in the center of the
backs were achieved using surgical punches. A suspension containing 10
7
S. aureus strain 6498 in PBS was inoculated onto each wound. Uninfected
METH- or PBS-treated mice were used as additional controls. Photo-
graphs of the wounds were taken on days 3, 5, 7, 9, 11, 13, 15, and 17. The
wound dimensions were measured every other day by two different oper-
ators using dial calipers in a blinded fashion. Although mice experience
formication after METH administration, this behavior did not influence
the results obtained, given that each animal was isolated in their own cage,
eliminating the possibility of mice scratching other mice; in addition, the
location of the wound made it difficult for the animals to scratch their own
wounds. For histology, CFU determinations, and gene expression analy-
sis, animals were euthanized at day 7 and wound tissues were excised.
Animals were otherwise euthanized at day 21, by which time all wounds
had healed.
Ethics statement. Animal experiments were performed according to
the guide published by the Institute of Laboratory Animal Resources of
the National Research Council. Animal care for this study was approved
by the Animal Welfare and Research Ethics Committee at the Albert Ein-
stein College of Medicine (protocol number 20110402).
Histology. Skin tissues were excised and fixed in 4% paraformalde-
hyde (PFA) for 24 h. Tissues were processed and embedded in paraffin,
and 4-
m vertical sections were fixed to glass slides. Tissue sections were
stained with hematoxylin and eosin (H&E), Gram stain, and Gomori’s
trichrome, and for CD34 or Iba-1, to examine tissue morphology, bacte-
ria, collagen deposition, vascularization, and macrophage infiltration, re-
spectively. Slides were visualized using an Axiovert 40CFL inverted mi-
croscope (Carl Zeiss), and images were captured with an AxioCam MrC
digital camera using the Zen 2011 digital imaging software.
Collagen deposition determinations. Collagen deposition was mea-
sured based on intensity, using ImageJ software (National Institutes of
Health, USA) (52). The software possesses threshold filters to discrimi-
nate between the stain colors (white, red, and blue), providing an accurate
measurement of each color’s intensity. Ten 40fields were evaluated per
section.
CFU determinations. Excised wound tissues were homogenized in
PBS and plated on tryptic soy agar (TSA) for CFU determinations. The
results were normalized by tissue weights.
Real-time (RT)-PCR. For RT-PCR analysis of host gene expression,
we analyzed col1,col3, and mmp2 expression. col1 encodes collagen type I,
and col3 encodes collagen type III, whereas mmp2 encodes matrix
metalloproteinase-2. At day 7, excised tissues were homogenized, cells
were collected and washed, and then RNA was isolated using an RNeasy
kit. col1,col3, and mmp2 expression levels were analyzed by RT-PCR as
described elsewhere (52).
In S. aureus,ica encodes the intercellular adhesion locus and is re-
quired for S. aureus biofilm formation. Twenty-four hours after incuba-
tion with METH, bacterial RNA was isolated and analyzed by RT-PCR as
described previously (53).
TABLE 3 Characteristics of S. aureus clinical strains tested in this
study
a
Strain Source mecA SCC ACME MLST Toxin genes
038 Blood IV CC 8 seb,sek,sep,seq,ser,seu
067 Wound MSSA CC 8 sea,seb,sek,sep,seq,ser,seu
085
b
Wound IV CC 8 pvl,sek,sep,ser
112 Wound IV CC 8 sek,ser,seu
132 Wound IV ND
c
seb,sek,ser
6498
b
Wound IV CC 8 pvl,sek,sep,ser
a
Toxin profiles were determined by PCR amplification with primer sets for 19 different
toxins. Abbreviations (corresponding gene): staphylococcal enterotoxin a (sea),B(seb),
K(sek),P(sep),Q(seq),R(ser),U(seu), and Panton-Valentine leukocidin (pvl).
ACME, arginine catabolic mobile element; MLST, multilocus sequence type; CC, clonal
complex; ND, not determined.
b
USA300 strain.
Mihu et al.
12 ®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
For an internal mRNA control, we used primers specific for
glyceraldehyde-3-phosphate dehydrogenase (GAPDH).
Phagocytic cell counts in blood. Twenty-one days after METH ad-
ministration, phagocytic cell counts were performed by differential leu-
kocyte count for samples from all experimental animals by using a Hema
3 Stat Pack kit and light microscopy.
Rationale for METH doses used in in vitro studies. Controlled stud-
ies indicated that a single 260-mg dose peaks at a level of 7.5
M (54).
Thus, a single dose of 260 mg/g would be expected to produce 7.5 to 28.8
M blood METH levels. IDUs tend to self-administer METH in binges,
and as the drug exhibits a half-life of 11.4 to 12 h (55, 56), this can lead to
higher drug levels. Published studies modeling binge patterns of use in
individuals have shown that the fourth administration of 260 mg during a
single day produced blood levels of 17
M and could reach 20
Monthe
second day of such a binge (54). Thus, binge doses of 260 to 1,000 mg
produce 17 to 80
M blood METH levels and levels in the micromolar
range of hundreds in organs, including the brain and the spleen (57).
Therefore, we selected ~25 to 50
M METH to perform our in vitro
experiments.
Bacterial viability assay. To determine the impact of the METH on S.
aureus growth, 1 ml of TS broth (TSB) was inoculated with a 24-h colony
of the bacterium grown on TSA. One hundred microliters of S. aureus
broth suspension was inoculated in each well of a 200-well plate, contain-
ing 100
l of TSB with either 25 or 50
M METH. The inoculated plate
was then incubated for 24 h at 37°C. Controls included wells containing
bacterial suspension alone (untreated). Growth was assessed every 15 min
using a microplate reader set at an optical density (OD) of 600 nm.
Collagenolytic activity assay. Collagenolytic activity was measured
using murine fluorescein isothiocyanate (FITC)-conjugated type I colla-
gen (28). FITC-conjugated type I collagen was incubated with a 10 nM
concentration of either staphopain A or MMP-2 for3hat37°C. Then,
100
l of the solution was mixed with 100
l ethanol (70% ethanol–
0.17 M Tris-HCl buffer, 0.67 M NaCl) and centrifuged for 10 min at 4°C.
To assess collagen degradation, 70
l of the supernatant was withdrawn,
and fluorescence was measured with a multilabel plate reader with emis-
sion at 520 nm and excitation at 495 nm.
Western blot analysis. To further characterize the role of METH in
wound healing, we assessed MMP-2 expression in the wounds of BALB/c
mice via Western blot analysis, as described previously (52).
Biofilm formation. For each strain, 100
l of a suspension with bac-
terial cells in RPMI 1640 medium supplemented with 1% Casamino Acids
was added into individual wells of polystyrene 96-well plates, and the
plates were incubated at 37°C without shaking as previously described
(58). The biofilms grown with PBS or METH (25 and 50
M) were al-
lowed to form for 24 h.
Quantitation of biofilms. Measurement of biofilm formation and vi-
ability was done by CFU determinations and assessment of the metabolic
activity of the attached cells in an XTT reduction assay, as previously
described (59).
Biofilm architecture visualization. The structural integrity of bio-
films formed in the absence or presence of METH was examined by using
the LIVE/DEAD biofilm viability kit and confocal microscopy, as previ-
ously described (60).
Isolation of peripheral blood neutrophils from mice and humans.
Retroorbital puncture was used to obtain blood from BALB/c mice,
whereas whole human blood was purchased from the Interstate Blood
Bank, Inc. (Memphis, TN). Erythrocytes were removed by hypotonic ly-
sis, and neutrophils were separated from the remaining cells by centrifu-
gation over discontinuous Percoll gradients. Neutrophils were 95% vi-
able, as determined by Wright-Giemsa staining. Recovered neutrophils
(~98% as determined by FACS, using Ly-6G as a marker) were briefly
maintained (30 min; 37°C, 10% CO
2
) in RPMI 1640 supplemented with
10 mM HEPES (pH 7.4) and 10% fetal calf serum (FCS) prior to use.
Controls. Chloroquine is a weak base and a well-known inhibitor of
endosomal acidification (61), whereas CytD is a potent inhibitor of actin
polymerization. Both reagents were used as controls in the in vitro studies
of S. aureus-phagocyte interactions.
Murine neutrophil killing assay. For killing assays (62), 3 10
5
neu-
trophils in 200
l of RPMI 1640 with 10% FCS were incubated with
METH (25 or 50
M), Chlq (25
M), CytD (25
M), or PBS in micro-
centrifuge tubes with 8 rpm rotation for2hat37°C, 5% CO
2
.S. aureus
6498 cultures were washed twice in PBS, diluted to a concentration of 4.5
10
6
CFU in 100
l RPMI 1640 plus 10% FCS (feeding medium), and
mixed with washed neutrophils in the same medium (ratio, 15 bacteria:1
neutrophil), centrifuged at 1,200 rpm for 5 min, and then incubated at
37°C in a 5% CO
2
incubator. Gentamicin (final concentration of
400
g/ml for S. aureus) was added after 10 min to kill extracellular bac-
teria. At specified time points (20 to 80 min), the contents of sample wells
were withdrawn, centrifuged to pellet the neutrophils, and washed to
remove the antibiotic medium. Neutrophils were then lysed in 0.02%
Triton X-100, and CFU were calculated after plating on TSA.
Macrophages. Primary macrophages were isolated from untreated
BALB/c mice by lavage of the abdominal cavity with Hanks’ balanced salt
solution with 1 mM EGTA (21).
Phagocytosis. For neutrophils, phagocytosis was determined by using
confocal microscopy. Primary murine neutrophils were incubated with
METH (25 or 50
M), Chlq (25
M), CytD (25
M), or PBS for 90 min
at 37°C and 5% CO
2
, prior to incubation for phagocytosis in the presence
of Alexa Fluor 350 (Alexa)-labeled S. aureus 6498 cells for 30 min. Genta-
micin (400
g/ml) was added to kill extracellular bacteria. Similarly, ex-
tracellular bacteria were quenched with trypan blue to prevent interfer-
ence with the assay. Microscopic examinations of neutrophils were
performed with a Leica TCS SP5 confocal laser scanning microscope.
Z-stack images and measurements were corrected by utilizing Bio-Rad
LaserSharp 2000 software in deconvolution mode. The fluorescent inten-
sity of bacteria inside neutrophils was analyzed using Adobe Photoshop
software (63).
For macrophages, phagocytosis was determined by using FACS anal-
ysis. Primary macrophages were incubated with METH (25 or 50
M),
Chlq (25
M), CytD (25
M),orPBSfor2hat37°C and 5% CO
2
, prior
to incubation for phagocytosis in the presence of Alexa-labeled S. aureus
6498 for 30 min. Gentamicin (400
g/ml) was added to kill extracellular
bacteria. Similarly, extracellular bacteria were quenched with trypan blue
to prevent interference with the assay. Samples were processed (10,000
events per condition) on an LSRII flow cytometer (BD) and were analyzed
using FlowJo software.
Macrophage killing assay. Primary macrophages were first allowed to
phagocytize S. aureus 6498 cells for 30 min. Each well containing interact-
ing cells was gently washed with feeding medium and incubated with
feeding medium supplemented with gentamicin (400
g/ml) and either
PBS, METH (25 or 50
M), Chlq (25
M), or CytD (25
M) for 30 to
120 min. Quantification of viable S. aureus cells was determined by mea-
suring CFU after lysis of macrophages (64).
Phagosomal pH. The pH of S. aureus 6498-containing phagosomes in
primary macrophages was determined as described elsewhere (21).
Nitric oxide and myeloperoxidase production. NO production was
quantified using a Griess method kit (Promega). Similarly, MPO pro-
duced by N-formylmethionyl-leucyl-phenylalanine (fMLP)-stimulated
neutrophils in culture after coincubation with S. aureus was measured
using the myeloperoxidase assay kit.
Chemotaxis. Chemotaxis was measured using a transwell chamber
with 6.5-mm-diameter polycarbonate filters (3-
m pore size). Immedi-
ately after isolation, cells were incubated in the absence or presence of
METH (25 or 50
M) for 2 h. Chlq (25
M) and CytD (25
M) were used
as positive controls. Cells were transferred to RPMI 1640 with heat-
inactivated FCS, cultivated on filters, and allowed to migrate toward the
chemoattractant fMLP or cultured in medium alone at 37°C, 5% CO
2
.
After various time points (20, 40, and 60 min), the filters were removed,
and the cells that migrated through the membrane were fixed, stained, and
Methamphetamine Exacerbates MRSA Skin Infection
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 13
counted by light microscopy (400) as described elsewhere (65). Ten
nonoverlapping and randomized fields were counted by one scorer.
Human neutrophil phagocytic and killing assays. Phagocytosis and
killing assays were performed as previously described (66). For the phago-
cytosis assay, FACS analysis was employed. Human neutrophils (10
6
cells)
were incubated on six-well plates with feeding medium supplemented
with METH (25 or 12.5
M)orPBSfor2hat37°C and 5% CO
2
. FITC-
labeled S. aureus cells were incubated with 25% human serum for 30 min
to allow complement proteins to opsonize S. aureus cells.Bacterial cells
were washed and then 10
7
were added to the 10
6
neutrophils for 60 min.
Similarly, extracellular bacteria were quenched with trypan blue to pre-
vent interference with the assay. Samples were processed on an LSRII flow
cytometer and were analyzed using FlowJo software.
Since METH reduces S. aureus phagocytosis by neutrophils, for the
killing assays leukocytes were first allowed to phagocytize S. aureus cells
for 0.5 h to determine initial uptake. Each well containing interacting cells
was gently washed with feeding medium and incubated with feeding me-
dium supplemented with gentamicin (400
g/ml, to kill extracellular bac-
teria) and either PBS or METH (25 or 50
M) for 4 h. Viable bacteria were
released from neutrophils following 0.5 or4hofhost-cell interaction by
forcibly subjecting cultures to 27-gauge needle passage 5 to 7 times for
efficient lysis. Four microtiter wells per condition were used to ascertain
CFU. For each well, serial dilutions were plated in triplicates onto TSA
plates, which were incubated at 37°C for 24 h prior to CFU tallying.
Luminol chemiluminescence assay. ROS signals were made chemilu-
minescent (CL) by using luminol (1 mM). CL was monitored for 30 min
with an automatic luminescence analyzer (SpectraMax L; Molecular De-
vices) at 37°C.
Cytokine determinations. Cutaneous tissues from mice were excised
at 24 h and 7 days after wounding and homogenized in PBS with protease
inhibitors. Cell debris was removed from homogenates by centrifugation
at 6,000 gfor 10 min. Samples were stored at 80°C until tested.
Supernatants were tested for IFN-
, IL-1
, IL-4, IL-6, IL-10, IL-12p70,
MCP-1, TGF-
, TNF-
, and VEGF in an enzyme-linked immunosorbent
assay (OptEIA kits; Becton, Dickinson Biosciences).
Statistical analysis. All data were analyzed using Prism (GraphPad, La
Jolla, CA). Analyses of CFU (Fig. 2B) and phagocytic cell count (Fig. 4B)
data were performed using Student’s ttest. Analysis of all the other data in
the study was performed using analysis of variance (ANOVA) and ad-
justed by use of the Bonferroni correction. Pvalues of 0.05 were con-
sidered significant.
ACKNOWLEDGMENTS
E.A.E. is supported by the National Institutes of Health (NIH MH096625)
and PHRI internal awards. A.J.G. is supported by Fundação de Apoio a
Pesquisa do Estado do Rio de Janeiro and Conselho Nacional de Pesquisa
e Desenvolvimento (CNPq). B.C.F. is supported by NIH
2R01AI059681-06 and R21AI087564-01. J.D.N. is supported in part by an
Irma T. Hirschl/Monique Weill-Caulier Trust Research award. L.R.M.
was supported by NIH NIAID award 5K22A1087817-02 and NYIT COM
start-up funds.
M.R.M. and J.R.-S. performed CFU, cytokine, and histological analy-
ses. B.P.S. performed the phagocytic cell counts and MMP-2-related ex-
periments. A.K.V. and B.C.F. performed the in vivo wound healing exper-
iments. L.N.N. performed the collagen gene expression analysis. A.J.G.
and J.D.N. performed chemotaxis, phagocytosis, nitric oxide production,
and killing assays. E.A.E. performed the confocal microscopy analysis.
J.D.N. assisted in data analysis and editing of the manuscript. H.H.L.
performed the experiments involving human neutrophils. L.R.M. di-
rected the overall design of the experiments, analysis of the data, and
writing of the manuscript.
REFERENCES
1. McCaig L, McDonald L, Mandal S, Jernigan D. 2006. Staphylococcus
aureus-associated skin and soft tissue infections in ambulatory care.
Emerg Infect Dis 12:1715–1723. http://dx.doi.org/10.3201/
eid1211.060190.
2. Noskin GA, Rubin RJ, Schentag JJ, Kluytmans J, Hedblom EC, Jacob-
son C, Smulders M, Gemmen E, Bharmal M. 2007. National trends in
Staphylococcus aureus infection rates: impact on economic burden and
mortality over a 6-year period (1998 –2003). Clin Infect Dis 45:
1132–1140. http://dx.doi.org/10.1086/522186.
3. Moran GJ, Krishnadasan A, Gorwitz RJ, Fosheim GE, McDougal LK,
Carey RB, Talan DA. 2006. Methicillin-resistant S. aureus infections
among patients in the emergency department. N Engl J Med 355:
666 674. http://dx.doi.org/10.1056/NEJMoa055356.
4. Hiramatsu K, Hanaki H, Ino T, Yabuta K, Oguri T, Tenover FC. 1997.
Methicillin-resistant Staphylococcus aureus clinical strain with reduced
vancomycin susceptibility. J Antimicrob Chemother 40:135–136.
5. Klevens RM, Morrison MA, Nadle J, Petit S, Gershman K, Ray S,
Harrison LH, Lynfield R, Dumyati G, Townes JM, Craig AS, Zell ER,
Fosheim GE, McDougal LK, Carey RB, Fridkin SK. 2007. Invasive
methicillin-resistant Staphylococcus aureus infections in the United States.
JAMA 298:1763–1771. http://dx.doi.org/10.1001/jama.298.15.1763.
6. Ellis R, Childers M, Cherner M, Lazzaretto D, Letendre S, Grant I. 2003.
Increased human immunodeficiency virus loads in active methamphet-
amine users are explained by reduced effectiveness of antiretroviral ther-
apy. J Infect Dis 188:1820 –1826. http://dx.doi.org/10.1086/379894.
7. Gonzales R, Marinelli-Casey P, Shoptaw S, Ang A, Rawson RA. 2006.
Hepatitis C virus infection among methamphetamine-dependent individ-
uals in outpatient treatment. J Subst Abus Treat 31:195–202. http://
dx.doi.org/10.1016/j.jsat.2006.04.006.
8. Salgado CD, Farr BM, Calfee DP. 2003. Community-acquired
methicillin-resistant Staphylococcus aureus: a meta-analysis of prevalence
and risk factors. Clin Infect Dis 36:131–139. http://dx.doi.org/10.1086/
345436.
9. Ebright JR, Pieper B. 2002. Skin and soft tissue infections in injection
drug users. Infect Dis Clin North Am 16:697–712. http://dx.doi.org/
10.1016/S0891-5520(02)00017-X.
10. Gordon RJ, Lowy FD. 2005. Bacterial infections in drug users. N Engl J
Med 353:1945–1954. http://dx.doi.org/10.1056/NEJMra042823.
11. Roose RJ, Hayashi AS, Cunningham CO. 2009. Self-management of
injection-related wounds among injecting drug users. J Addict Dis 28:
74 80. http://dx.doi.org/10.1080/10550880802545200.
12. Kerr T, Wood E, Grafstein E, Ishida T, Shannon K, Lai C, Montaner J,
Tyndall MW. 2005. High rates of primary care and emergency depart-
ment use among injection drug users in Vancouver. J Public Health 27:
62– 66. http://dx.doi.org/10.1093/pubmed/fdh189.
13. Cohen AL, Shuler C, McAllister S, Fosheim GE, Brown MG, Abercrom-
bie D, Anderson K, McDougal LK, Drenzek C, Arnold K, Jernigan D,
Gorwitz R. 2007. Methamphetamine use and methicillin-resistant Staph-
ylococcus aureus skin infections. Emerg Infect Dis 13:1707–1713. http://
dx.doi.org/10.3201/eid1311.070148.
14. Pollini RA, Gallardo M, Hasan S, Minuto J, Lozada R, Vera A, Zúñiga
ML, Strathdee SA. 2010. High prevalence of abscesses and self-treatment
among injection drug users in Tijuana, Mexico. Int J Infect Dis 14(Suppl
3):e117– e122. http://dx.doi.org/10.1016/j.ijid.2010.02.2238.
15. Cespedes C, Saïd-Salim B, Miller M, Lo S, Kreiswirth B, Gordon R,
Vavagiakis P, Klein R, Lowy F. 2005. The clonality of Staphylococcus
aureus nasal carriage. J Infect Dis 191:444 452. http://dx.doi.org/
10.1086/427240.
16. Vlahov D, Sullivan M, Astemborski J, Nelson KE. 1992. Bacterial infec-
tions and skin cleaning prior to injection among intravenous drug users.
Public Health Rep 107:595–598.
17. Iwasa M, Maeno Y, Inoue H, Koyama H, Matoba R. 1996. Induction of
apoptotic cell death in rat thymus and spleen after a bolus injection of
methamphetamine. Int J Leg Med 109:23–28. http://dx.doi.org/10.1007/
BF01369597.
18. Freire-Garabal M, Balboa J, Núñez MJ, Castaño MT, Llovo J,
Fernández-Rial J, Belmonte A. 1991. Effects of amphetamine on T-cell
immune response in mice. Life Sci 49:PL107–PL112. http://dx.doi.org/
10.1016/0024-3205(91)90570-2.
19. Potula R, Hawkins BJ, Cenna JM, Fan S, Dykstra H, Ramirez SH,
Morsey B, Brodie MR, Persidsky Y. 2010. Methamphetamine causes
mitrochondrial oxidative damage in human T lymphocytes leading to
functional impairment. J Immunol 185:2867–2876. http://dx.doi.org/
10.4049/jimmunol.0903691.
20. Tallóczy Z, Martinez J, Joset D, Ray Y, Gácser A, Toussi S, Mizushima
Mihu et al.
14 ®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
N, Nosanchuk J, Goldstein H, Loike J, Sulzer D, Santambrogio L. 2008.
Methamphetamine inhibits antigen processing, presentation, and phago-
cytosis. PLoS Pathog 4:e28. http://dx.doi.org/10.1371/
journal.ppat.0040028.
21. Martinez L, Mihu M, Gácser A, Santambrogio L, Nosanchuk J. 2009.
Methamphetamine enhances histoplasmosis by immunosuppression of
the host. J Infect Dis 200:131–141. http://dx.doi.org/10.1086/599328.
22. Yu Q, Zhang D, Walston M, Zhang J, Liu Y, Watson RR. 2002. Chronic
methamphetamine exposure alters immune function in normal and
retrovirus-infected mice. Int Immunopharmacol 2:951–962. http://
dx.doi.org/10.1016/S1567-5769(02)00047-4.
23. In SW, Son EW, Rhee DK, Pyo S. 2005. Methamphetamine administra-
tion produces immunomodulation in mice. J Toxicol Environ Health A
68:2133–2145. http://dx.doi.org/10.1080/15287390500177156.
24. Mahajan SD, Hu Z, Reynolds JL, Aalinkeel R, Schwartz SA, Nair MPN.
2006. Methamphetamine modulates gene expression patterns in mono-
cyte derived mature dendritic cells: implications for HIV-1 pathogenesis.
Mol Diagn Ther 10:257–269. http://dx.doi.org/10.1007/BF03256465.
25. Magnet S, Courvalin P, Lambert T. 2001. Resistance-nodulation-cell
division-type efflux pump involved in aminoglycoside resistance in Acin-
etobacter baumannii strain BM4454. Antimicrob Agents Chemother 45:
3375–3380. http://dx.doi.org/10.1128/AAC.45.12.3375-3380.2001.
26. Rangaraj A, Harding K, Leaper D. 2011. Role of collagen in wound
management. Wounds UK 7:54 63. http://www.wounds-uk.com/pdf/
content_10039.pdf.
27. Goularte TA, Craven DE. 1986. Results of a survey of infection control
practices for respiratory therapy equipment. Infect Control 7:327–330.
28. Ohbayashi T, Irie A, Murakami Y, Nowak M, Potempa J, Nishimura Y,
Shinohara M, Imamura T. 2011. Degradation of fibrinogen and collagen
by staphopains, cysteine proteases released from Staphylococcus aureus.
Microbiology 157:786 –792. http://dx.doi.org/10.1099/mic.0.044503-0.
29. Kanangat S, Postlethwaite A, Hasty K, Kang A, Smeltzer M, Appling W,
Schaberg D. 2006. Induction of multiple matrix metalloproteinases in
human dermal and synovial fibroblasts by Staphylococcus aureus: implica-
tions in the pathogenesis of septic arthritis and other soft tissue infections.
Arthritis Res Ther 8:R176. http://dx.doi.org/10.1186/ar2086.
30. Wang JH, Kwon HJ, Jang YJ. 2010. Staphylococcus aureus increases cy-
tokine and matrix metalloproteinase expression in nasal mucosae of pa-
tients with chronic rhinosinusitis and nasal polyps. Am J Rhinol Allergy
24:422– 427. http://dx.doi.org/10.2500/ajra.2010.24.3509.
31. Conant K, St Hillaire CS, Anderson C, Galey D, Wang J, Nath A. 2004.
Human immunodeficiency virus type 1 Tat and methamphetamine affect
the release and activation of matrix-degrading proteinases. J Neurovirol
10:21–28. http://dx.doi.org/10.1080/13550280490261699.
32. Mizoguchi H, Yamada K, Nabeshima T. 2011. Matrix metalloproteinases
contribute to neuronal dysfunction in animal models of drug dependence,
Alzheimer’s disease, and epilepsy. Biochem Res Int 2011:681385. http://
dx.doi.org/10.1155/2011/681385.
33. Harper RA, Flaxman BA. 1975. Effect of pharmacological agents on hu-
man keratinocyte mitosis in vitro. II. Inhibition by catecholamines. J Cell
Physiol 86:293–299. http://dx.doi.org/10.1002/jcp.1040860213.
34. Fuziwara S, Suzuki A, Inoue K, Denda M. 2005. Dopamine D2-like
receptor agonists accelerate barrier repair and inhibit the epidermal hy-
perplasia induced by barrier disruption. J Invest Dermatol 125:783–789.
http://dx.doi.org/10.1111/j.0022-202X.2005.23873.x.
35. Grando SA, Pittelkow MR, Schallreuter KU. 2006. Adrenergic and cho-
linergic control in the biology of epidermis: physiological and clinical
significance. J Invest Dermatol 126:1948 –1965. http://dx.doi.org/
10.1038/sj.jid.5700151.
36. Sivamani RK, Lam ST, Isseroff RR. 2007. Beta adrenergic receptors in
keratinocytes. Dermatol Clin 25:643– 653. http://dx.doi.org/10.1016/
j.det.2007.06.012.
37. Shome S, Rana T, Ganguly S, Basu B, Chaki Choudhury S, Sarkar C,
Chakroborty D, Dasgupta PS, Basu S. 2011. Dopamine regulates angio-
genesis in normal dermal wound tissues. PLoS One 6:e25215. http://
dx.doi.org/10.1371/journal.pone.0025215.
38. Sivamani RK, Pullar CE, Manabat-Hidalgo CG, Rocke DM, Carlsen
RC, Greenhalgh DG, Isseroff RR. 2009. Stress-mediated increases in
systemic and local epinephrine impair skin wound healing: potential new
indication for beta blockers. PLoS Med 6:e12. http://dx.doi.org/10.1371/
journal.pmed.1000012.
39. Han YP, Tuan TL, Wu H, Hughes M, Garner WL. 2001. TNF-alpha
stimulates activation of pro-MMP2 in human skin through NF-
B medi-
ated induction of MT1-MMP. J Cell Sci 114:131–139. http://
jcs.biologists.org/content/114/1/131.long.
40. James GA, Swogger E, Wolcott R, Pulcini E, Secor P, Sestrich J,
Costerton JW, Stewart PS. 2008. Biofilms in chronic wounds. Wound
Repair Regen 16:37– 44. http://dx.doi.org/10.1111/j.1524
-475X.2007.00321.x.
41. Zhao G, Usui ML, Underwood RA, Singh PK, James GA, Stewart PS,
Fleckman P, Olerud JE. 2012. Time course study of delayed wound heal-
ing in a biofilm-challenged diabetic mouse model. Wound Repair Regen
20:342–352. http://dx.doi.org/10.1111/j.1524-475X.2012.00793.x.
42. Zhao G, Hochwalt PC, Usui ML, Underwood RA, Singh PK, James GA,
Stewart PS, Fleckman P, Olerud JE. 2010. Delayed wound healing in
diabetic (dB/dB) mice with Pseudomonas aeruginosa biofilm challenge: a
model for the study of chronic wounds. Wound Repair Regen 18:
467– 477. http://dx.doi.org/10.1111/j.1524-475X.2010.00608.x.
43. Hübner G, Werner S. 1996. Serum growth factors and proinflammatory
cytokines are potent inducers of activin expression in cultured fibroblasts
and keratinocytes. Exp Cell Res 228:106 –113. http://dx.doi.org/10.1006/
excr.1996.0305.
44. Werner S, Grose R. 2003. Regulation of wound healing by growth factors
and cytokines. Physiol Rev 83:835– 870. http://dx.doi.org/10.1152/
physrev.00032.2002.
45. Thurlow LR, Hanke ML, Fritz T, Angle A, Aldrich A, Williams SH,
Engebretsen IL, Bayles KW, Horswill AR, Kielian T. 2011. Staphylococ-
cus aureus biofilms prevent macrophage phagocytosis and attenuate in-
flammation in vivo. J Immunol 186:6585– 6596. http://dx.doi.org/
10.4049/jimmunol.1002794.
46. Frank S, H bner G, Breier G, Longaker MT, Greenhalgh DG, Werner S.
1995. Regulation of vascular endothelial growth factor expression in cul-
tured keratinocytes. Implications for normal and impaired wound heal-
ing. J Biol Chem 270:12607–12613. http://dx.doi.org/10.1074/
jbc.270.21.12607.
47. Mirza R, DiPietro LA, Koh TJ. 2009. Selective and specific macrophage
ablation is detrimental to wound healing in mice. Am J Pathol 175:
2454 –2462. http://dx.doi.org/10.2353/ajpath.2009.090248.
48. Lucas T, Waisman A, Ranjan R, Roes J, Krieg T, Muller W, Roers A,
Eming SA. 2010. Differential roles of macrophages in diverse phases of
skin repair. J Immunol 184:3964 –3977. http://dx.doi.org/10.4049/
jimmunol.0903356.
49. Martinez LR, Han G, Chacko M, Mihu MR, Jacobson M, Gialanella P,
Friedman AJ, Nosanchuk JD, Friedman JM. 2009. Antimicrobial and
healing efficacy of sustained release nitric oxide nanoparticles against
Staphylococcus aureus skin infection. J Invest Dermatol 129:2463–2469.
http://dx.doi.org/10.1038/jid.2009.95.
50. Han G, Martinez LR, Mihu MR, Friedman AJ, Friedman JM,
Nosanchuk JD. 2009. Nitric oxide releasing nanoparticles are therapeutic
for Staphylococcus aureus abscesses in a murine model of infection. PLoS
One 4:e7804. http://dx.doi.org/10.1371/journal.pone.0007804.
51. Simon SL, Richardson K, Dacey J, Glynn S, Domier CP, Rawson RA,
Ling W. 2001. A comparison of patterns of methamphetamine and co-
caine use. J Addict Dis 21:35– 44. http://dx.doi.org/10.1300/J069v21n01
_04.
52. Han G, Nguyen LN, Macherla C, Chi Y, Friedman JM, Nosanchuk JD,
Martinez LR. 2012. Nitric oxide-releasing nanoparticles accelerate wound
healing by promoting fibroblast migration and collagen deposition. Am J
Pathol 180:1465–1473. http://dx.doi.org/10.1016/j.ajpath.2011.12.013.
53. Conlon KM, Humphreys H, O’Gara JP. 2002. Regulation of icaR gene
expression in Staphylococcus epidermidis. FEMS Microbiol Lett 216:
171–177. http://dx.doi.org/10.1111/j.1574-6968.2002.tb11432.x.
54. Melega WP, Cho AK, Harvey D, Lac´an G. 2007. Methamphetamine
blood concentrations in human abusers: application to pharmacokinetic
modeling. Synapse 61:216 –220. http://dx.doi.org/10.1002/syn.20365.
55. Cho AK, Melega WP, Kuczenski R, Segal DS. 2001. Relevance of phar-
macokinetic parameters in animal models of methamphetamine abuse.
Synapse 39:161–166. http://dx.doi.org/10.1002/1098
-2396(200102)39:2161::AID-SYN73.0.CO;2-E.
56. Harris D, Boxenbaum H, Everhart ET, Sequeira G, Mendelson JE, Jones
RT. 2003. The bioavailability of intranasal and smoked methamphet-
amine. Clin Pharmacol Ther 74:475– 486. http://dx.doi.org/10.1016/
j.clpt.2003.08.002.
57. Riviere GJ, Gentry WB, Owens SM. 2000. Disposition of methamphet-
amine and its metabolite amphetamine in brain and other tissues in rats
Methamphetamine Exacerbates MRSA Skin Infection
November/December 2015 Volume 6 Issue 6 e01622-15 ®mbio.asm.org 15
after intravenous administration. J Pharmacol Exp Ther 292:1042–1047.
http://jpet.aspetjournals.org/content/292/3/1042.long.
58. Scherr TD, Hanke ML, Huang O, James DBA, Horswill AR, Bayles KW,
Fey PD, Torres VJ, Kielian T. 2015. Staphylococcus aureus biofilms induce
macrophage dysfunction through leukocidin AB and alpha-toxin. mBio
6:e0102-15. http://dx.doi.org/10.1128/mBio.01021-15.
59. Chandra J, Mukherjee PK, Ghannoum MA. 2008. In vitro growth and
analysis of Candida biofilms. Nat Protoc 3:1909 –1924. http://dx.doi.org/
10.1038/nprot.2008.192.
60. Anderson MJ, Lin Y, Gillman AN, Parks PJ, Schlievert PM, Peterson
ML. 2012. Alpha-toxin promotes Staphylococcus aureus mucosal biofilm
formation. Front Cell Infect Microbiol 2:64. http://dx.doi.org/10.3389/
fcimb.2012.00064.
61. Zwart W, Griekspoor A, Kuijl C, Marsman M, van Rheenen J, Janssen
H, Calafat J, van Ham M, Janssen L, van Lith M, Jalink K, Neefjes J.
2005. Spatial separation of HLA-DM/HLA-DR interactions within MIIC
and phagosome-induced immune escape. Immunity 22:221–233. http://
dx.doi.org/10.1016/j.immuni.2005.01.006.
62. Liu GY, Essex A, Buchanan JT, Datta V, Hoffman HM, Bastian JF,
Fierer J, Nizet V. 2005. Staphylococcus aureus golden pigment impairs
neutrophil killing and promotes virulence through its antioxidant activity.
J Exp Med 202:209 –215. http://dx.doi.org/10.1084/jem.20050846.
63. Silveira CP, Piffer AC, Kmetzsch L, Fonseca FL, Soares DA, Staats CC,
Rodrigues ML, Schrank A, Vainstein MH. 2013. The heat shock protein
(Hsp) 70 of Cryptococcus neoformans is associated with the fungal cell
surface and influences the interaction between yeast and host cells. Fungal
Genet Biol 60:53– 63. http://dx.doi.org/10.1016/j.fgb.2013.08.005.
64. Steenbergen JN, Shuman HA, Casadevall A. 2001. Cryptococcus neofor-
mans interactions with amoebae suggest an explanation for its virulence
and intracellular pathogenic strategy in macrophages. Proc Natl Acad Sci
USA98:15245–15250. http://dx.doi.org/10.1073/pnas.261418798.
65. McHugh D, Tanner C, Mechoulam R, Pertwee RG, Ross RA. 2008.
Inhibition of human neutrophil chemotaxis by endogenous cannabinoids
and phytocannabinoids: evidence for a site distinct from CB1 and CB2.
Mol Pharmacol 73:441– 450. http://dx.doi.org/10.1124/mol.107.041863.
66. Gandhi JA, Ekhar VV, Asplund MB, Abdulkareem AF, Ahmadi M,
Coelho C, Martinez LR. 2014. Alcohol enhances Acinetobacter
baumannii-associated pneumonia and systemic dissemination by impair-
ing neutrophil antimicrobial activity in a murine model of infection. PLoS
One 9:e95707. http://dx.doi.org/10.1371/journal.pone.0095707.
Mihu et al.
16 ®mbio.asm.org November/December 2015 Volume 6 Issue 6 e01622-15
... Chemsex is described as the intentional use of recreational drugs, before or during sex, among gay, bisexual and other men who have sex with men (gbMSM) with the aim of prolonging, improving and facilitating sexual intercourse; it has been associated with the transmission of HIV and other sexually transmitted diseases (STDs) through sexual risk behaviors [1]. Previously, CA-MRSA infections have been reported separately in gbMSM [2], PLWH [3] and people who use injected drugs [4] and/or smoke methamphetamine [5,6] or use inhale nitrites and oral erectile dysfunction agents [3,7]. All these situations described in the literature separately occur simultaneously in people who engage in chemsex. ...
... In a murine model, Mihu et al. demonstrated that methamphetamine delays wound healing, accelerates collagen degradation and alters host phagocytic functions. Moreover, methamphetamine induces S. aureus biofilm formation [6]. These findings strongly suggest that methamphetamine use may alter immunity and host response to S. aureus infections and may increase virulence of S. aureus by biofilm formation. ...
Article
Full-text available
Introduction: There are no data on community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections in the context of the chemsex phenomenon. This study aimed to characterize CA-MRSA-related infections in a cohort of people living with HIV (PLWH) who engage in chemsex. Methods: At the Hospital Clinic of Barcelona, from February 2018 to January 2022, we analyzed CA-MRSA infections diagnosed in a cohort of PLWH who engage in chemsex. Epidemiological, behavioral and clinical variables were assessed. Mass spectrometry identification and antimicrobial susceptibility testing were performed on MRSA isolates. Pulse field electrophoresis was used to assess the clonality of the MRSA strains. The presence of Panton-Valentine leukocidin was also investigated. Results: Among the cohort of 299 participants who engage in chemsex, 25 (8%) with CA-MRSA infections were identified, 9 at baseline and 16 with incident cases; the cumulative incidence was 5.5% (95% CI: 3.2%, 8.8%). The most common drugs were methamphetamine (96%) and GHB/GBL (92%). Poly-consumption and slamming were reported by 32% and 46%, respectively. CA-MRSA was isolated from the infection sites of 20 participants, and CA-MRSA colonization was confirmed in the remaining 5 persons. Seventy-one percent had used antibiotics in the previous year. All participants presented with skin and soft tissue infections, 28% required hospitalization, and 48% had recurrence. Of the 23 MRSA isolates further studied, 19 (82,6%) belonged to the same clone. Panton-Valentine leukocidin was detected in all isolates. Conclusion: PLWH who engage in chemsex may present with CA-MRSA infections. Clinical suspicion and microbiological diagnosis are required to provide adequate therapy, and CA-MRSA prevention interventions should be designed.
... 32 In brief, MA itself is acidic and when smoked or inhaled, has been linked to direct corrosive effects on oral tissues. 39,40 It has been found that MA is linked to increased expression of proinflammatory factors, 41 cellular senescence, 41 reduced oral tissue wound healing, 42 and reduced saliva pH and buffering capacity. 43 Additionally, in murine models, MA has been found to facilitate hostmediated collagen degradation by increased expression and production of matrix metalloproteinases (MMPs). ...
... 43 Additionally, in murine models, MA has been found to facilitate hostmediated collagen degradation by increased expression and production of matrix metalloproteinases (MMPs). 42 Collectively, the effects of MA on oral cavity tissues demonstrates conserved properties to many cancer hallmarks, 30 therefore motivating this patient's exclusion from analysis due to confounding effects on the data. It is conceivable that with a large enough training dataset of MA patients, FLIm can differentiate benign MA-affected tissue from cancer, however this has yet to be tested. ...
Article
Background: This study evaluated whether fluorescence lifetime imaging (FLIm), coupled with standard diagnostic workups, could enhance primary lesion detection in patients with p16+ head and neck squamous cell carcinoma of the unknown primary (HNSCCUP). Methods: FLIm was integrated into transoral robotic surgery to acquire optical data on six HNSCCUP patients' oropharyngeal tissues. An additional 55-patient FLIm dataset, comprising conventional primary tumors, trained a machine learning classifier; the output predicted the presence and location of HNSCCUP for the six patients. Validation was performed using histopathology. Results: Among the six HNSCCUP patients, p16+ occult primary was surgically identified in three patients, whereas three patients ultimately had no identifiable primary site in the oropharynx. FLIm correctly detected HNSCCUP in all three patients (ROC-AUC: 0.90 ± 0.06), and correctly predicted benign oropharyngeal tissue for the remaining three patients. The mean sensitivity was 95% ± 3.5%, and specificity 89% ± 12.7%. Conclusions: FLIm may be a useful diagnostic adjunct for detecting HNSCCUP.
... The epidermis is the principal biological barrier that prevents diseases from entering the body and serves as the first line of defense 18 . Sweat glands in the skin produce a variety of bactericidal and regulatory peptides that inhibit the growth of pathogenic bacteria. ...
Article
Full-text available
In Iran, methamphetamine (MA) was acknowledged as "shisheh"1. It's a potent central nervous system stimulant2. In cases with ADHD and obesity, it is the second line of treatment. MA is also referred to as a "recreational drug". MA can be illegally created in meth labs all over the world due to its simple chemical procedure for synthesizing or manufacturing. Higher dosages of MA have numerous negative consequences. Because of the risk of neurotoxicity and recreational usage as a euphoriant, prescribing of this drug is restricted. This article reviews about the history, pharmacology, rapid and reversible change in dopamine transporters, route of administration, indication, contraindication, symptoms of abuse,adverse effects, withdrawal effects,impacts on systemic health, oral manifestations, dental considerations.
... METH can decrease human neutrophil function 59 . In rodents, METH facilitates neutrophil infiltration in various organs, leading to necrosis, atrophy, and other damages 60 . ...
Article
Full-text available
This review focuses on the effects and mechanisms of action of amphetamine-type stimulants (ATS) and their adverse effects on the cardiovascular, nervous, and immune systems. ATS include amphetamine (AMPH), methamphetamine (METH, “crystalmeth,” or “ice”), methylenedioxymethamphetamine (MDMA, “ecstasy,” or “Molly”), MDMA derivatives (e.g., methylenedioxyamphetamine [MDA] and methylenedioxy-N-ethylamphetamine [MDEA]), khat, and synthetic cathinones. The first section of this paper presents an overview of the historical aspects of ATS use, their initial clinical use, and regulations. The second part reviews the acute and chronic impact and the most salient clinical effects of ATS on the central nervous and cardiovascular systems, skin, and mouth. The chemical structure, pharmacokinetics, and classic and non-canonical pharmacological actions are covered in the third section, briefly explaining the mechanisms involved. In addition, the interactions of ATS with the central and peripheral immune systems are reviewed. The last section presents data about the syndemic of ATS and opioid use in the North American region, focusing on the increasing adulteration of METH with fentanyl.
... The epidermis is the principal biological barrier that prevents diseases from entering the body and serves as the first line of defense 18 . Sweat glands in the skin produce a variety of bactericidal and regulatory peptides that inhibit the growth of pathogenic bacteria. ...
Article
Full-text available
Background : Interleukin-6 (IL-6) is a proinflammatory cytokine expressed in numerous chronic inflammatory diseases whose ability to alter the pathophysiology and progression of periodontitis is well documented. Further its role in diabetes mellitus by creating an insulin resistance responsible for poor glycemic control is also being evaluated. The aim was to compare the levels of IL-6 in gingival crevicular fluid in periodontitis patients with and without diabetes and to analyze these levels in patients with poor glycemic control (HBA1c), in order to assess its role in the progression of periodontal destruction. Methods : 60 chronic periodontitis patients confirmed with CPITN index of age group 30-70 years were enrolled for the study. GCF samples from 30 patients with diabetes confirmed using HBA1c reports and 30 without diabetes using Cimasoni method were collected and stored at -70degreescelsiusand subjected to ELISA for IL-6 using krishgen human IL-6 ELISA kit as per manufacturer's instruction. Descriptive and inferential statistics were used using SPSS software. Results : While the diabetic group readings ranged from 4.4 Pg/µl to 7.0 Pg/µl with a mean of 5.8pg/µl, the non- diabetic group ranged from 1.5 Pg/µl to 4.8 Pg/µl with a mean value of3.24 pg/µl. There was a prominent increase in the IL-6 levels in diabetic when compared to non- diabetic which was statistically significant with p value < 0.001. Further, among the diabetic groups, patients with poor HBA1c with reading more than 7.7% showed a significant increase in IL-6 levels when compared to below 6.8%. Conclusions : The IL-6 levels in GCF were increased in chronic periodontitis patients with diabetes and more so in patients with poor glycemic control when compared to non-diabetic group. Therefore, periodontitis along with diabetes can play a major role in the inflammatory response within the periodontium.
Article
Amphetamines are the second most commonly used illicit drug worldwide. Amphetamine use can result in significant cutaneous morbidity. This review highlights the dermatological manifestations of amphetamine abuse.
Article
Purpose of review: The aim of this review is to discuss the latest evidence of the epidemiology, microbiology, risk factors, diagnosis and management of community-acquired skin and soft tissue infections (SSTIs) in people who inject drug (PWID). Recent findings: SSTIs are common complications in PWID and a major cause of morbidity and mortality. Infections can range from uncomplicated cellulitis, to abscesses, deep tissue necrosis and necrotizing fasciitis. They are predominantly caused by Gram-positive pathogens in particular Staphylococcus aureus and Streptococcus species; however, toxin-producing organisms such as Clostridium botulism or Clostridium tetani should be considered. The pathogenesis of SSTI in the setting of intravenous drug use (IDU) is different from non-IDU related SSTI, and management often requires surgical interventions in addition to adjunctive antibiotics. Harm reduction strategies and education about safe practices should be implemented to prevent morbidity and mortality as well as healthcare burden of SSTI in PWID. Summary: Prompt diagnosis and proper medical and surgical management of SSTI will improve outcomes in PWID.
Article
Full-text available
Methamphetamine is a widely abused psychostimulant and one of the main targets of dopamine transporter (DAT). Methamphetamine reduces DAT‐mediated dopamine uptake and stimulates dopamine efflux leading to increased synaptic dopamine levels many folds above baseline. Methamphetamine also targets DAT‐expressing peripheral immune cells, reduces wound healing, and increases infection susceptibility. Peripheral immune cells such as myeloid cells, B‐cells and T‐cells express DAT. DAT activity on monocytes and macrophages exhibits immune suppressive properties via an autocrine paracrine mechanism, where deletion or inhibition of DAT activity increases inflammatory responses. In this study utilizing a mouse model of daily single dose of methamphetamine administration, we investigated the impact of the drug on DAT expression in peripheral immune cells. We found in methamphetamine‐treated mice, DAT expression was downregulated in most of the innate and adaptive immune cells. Methamphetamine did not increase or decrease total number of innate and adaptive immune cells but changed their immunophenotype to low‐DAT expressing phenotype. Moreover, serum cytokine distributions were altered in methamphetamine‐treated mice. Therefore, resembling its effect in the CNS, in the periphery, methamphetamine regulates DAT expression on peripheral immune cell subsets, potentially describing methamphetamine regulation of peripheral immunity.
Article
Full-text available
Unlabelled: The macrophage response to planktonic Staphylococcus aureus involves the induction of proinflammatory microbicidal activity. However, S. aureus biofilms can interfere with these responses in part by polarizing macrophages toward an anti-inflammatory profibrotic phenotype. Here we demonstrate that conditioned medium from mature S. aureus biofilms inhibited macrophage phagocytosis and induced cytotoxicity, suggesting the involvement of a secreted factor(s). Iterative testing found the active factor(s) to be proteinaceous and partially agr-dependent. Quantitative mass spectrometry identified alpha-toxin (Hla) and leukocidin AB (LukAB) as critical molecules secreted by S. aureus biofilms that inhibit murine macrophage phagocytosis and promote cytotoxicity. A role for Hla and LukAB was confirmed by using hla and lukAB mutants, and synergy between the two toxins was demonstrated with a lukAB hla double mutant and verified by complementation. Independent confirmation of the effects of Hla and LukAB on macrophage dysfunction was demonstrated by using an isogenic strain in which Hla was constitutively expressed, an Hla antibody to block toxin activity, and purified LukAB peptide. The importance of Hla and LukAB during S. aureus biofilm formation in vivo was assessed by using a murine orthopedic implant biofilm infection model in which the lukAB hla double mutant displayed significantly lower bacterial burdens and more macrophage infiltrates than each single mutant. Collectively, these findings reveal a critical synergistic role for Hla and LukAB in promoting macrophage dysfunction and facilitating S. aureus biofilm development in vivo. Importance: Staphylococcus aureus has a propensity to form multicellular communities known as biofilms. While growing in a biofilm, S. aureus displays increased tolerance to nutrient deprivation, antibiotic insult, and even host immune challenge. Previous studies have shown that S. aureus biofilms thwart host immunity in part by preventing macrophage phagocytosis. It remained unclear whether this was influenced solely by the considerable size of biofilms or whether molecules were also actively secreted to circumvent macrophage-mediated phagocytosis. This is the first report to demonstrate that S. aureus biofilms inhibit macrophage phagocytosis and induce macrophage death through the combined action of leukocidin AB and alpha-toxin. Loss of leukocidin AB and alpha-toxin expression resulted in enhanced S. aureus biofilm clearance in a mouse model of orthopedic implant infection, suggesting that these toxins could be targeted therapeutically to facilitate biofilm clearance in humans.
Article
Full-text available
Collagen is structurally and functionally a key protein of the extracellular matrix which is also involved in scar formation during the healing of connective tissues. Many collagen dressings have been developed to enhance wound repair, particularly of non-infected, chronic, indolent skin ulcers. The use of collagen dressings is supported by relatively sparse and insufficient scientific data. This review identifies the supporting evidence for the use of the dressings which are available, often with widely different claimed advantages and modes of action, and considers future developments and assessment of collagen dressings. C ollagen is the unique, triple helix protein molecule, which forms the major part of the extracellular dermal matrix (ECM), together with the glycosaminoglycans, proteoglycans, laminin, fibronectin, elastin and cellular components (Hopkinson, 1992a, b; Berry et al, 1998; Enoch and Leaper, 2008). Collagen is the most abundant protein in animal tissues and accounts for 70–80% of the dry weight of the dermis (Hopkinson, 1992a). Mainly produced by fibroblasts, at least 21 genetically distinct collagens have currently been identified, with six of these being present in the skin. Collagen type I comprises approximately 70% of collagen in the skin, with type III being 10%, and trace amounts of collagen type IV, V, VI and VII (Uitto et al, 1989; Hay, 1991). 8 Protein synthesis in the extracellular matrix (ECM) 8 Synthesis and release of inflammatory cytokines and growth factors 8 Interactions between enzymes which remodel the ECM, including matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs), which are summarised in Table 1.
Article
Full-text available
Acinetobacter baumannii (Ab) is a common cause of community-acquired pneumonia (CAP) in chronic alcoholics in tropical and sub-tropical climates and associated with a >50% mortality rate. Using a murine model of alcohol (EtOH) administration, we demonstrated that EtOH enhances Ab-mediated pneumonia leading to systemic infection. Although EtOH did not affect neutrophil recruitment to the lungs of treated mice, it decreased phagocytosis and killing of bacteria by these leukocytes leading to increased microbial burden and severity of disease. Moreover, we determined that mice that received EtOH prior to Ab infection were immunologically impaired, which was reflected in increased pulmonary inflammation, sequential dissemination to the liver and kidneys, and decreased survival. Furthermore, immunosuppression by EtOH was associated with deregulation of cytokine production in the organs of infected mice. This study establishes that EtOH impairs immunity in vivo exacerbating Ab infection and disease progression. The ability of Ab to cause disease in alcoholics warrants the study of its virulence mechanisms and host interactions.
Article
Full-text available
Staphylococcus aureus causes many diseases in humans, ranging from mild skin infections to serious, life-threatening, superantigen-mediated Toxic Shock Syndrome (TSS). S. aureus may be asymptomatically carried in the anterior nares or vagina or on the skin, serving as a reservoir for infection. Pulsed-field gel electrophoresis clonal type USA200 is the most widely disseminated colonizer and the leading cause of TSS. The cytolysin α-toxin (also known as α-hemolysin or Hla) is the major epithelial proinflammatory exotoxin produced by TSS S. aureus USA200 isolates. The current study aims to characterize the differences between TSS USA200 strains [high (hla⁺) and low (hla⁻) α-toxin producers] in their ability to disrupt vaginal mucosal tissue and to characterize the subsequent infection. Tissue viability post-infection and biofilm formation of TSS USA200 isolates CDC587 and MN8, which contain the α-toxin pseudogene (hla⁻), MNPE (hla⁺), and MNPE isogenic hla knockout (hlaKO), were observed via LIVE/DEAD® staining and confocal microscopy. All TSS strains grew to similar bacterial densities (1–5 × 10⁸ CFU) on the mucosa and were proinflammatory over 3 days. However, MNPE formed biofilms with significant reductions in the mucosal viability whereas neither CDC587 (hla⁻), MN8 (hla⁻), nor MNPE hlaKO formed biofilms. The latter strains were also less cytotoxic than wild-type MNPE. The addition of exogenous, purified α-toxin to MNPE hlaKO restored the biofilm phenotype. We speculate that α-toxin affects S. aureus phenotypic growth on vaginal mucosa by promoting tissue disruption and biofilm formation. Further, α-toxin mutants (hla⁻) are not benign colonizers, but rather form a different type of infection, which we have termed high density pathogenic variants (HDPV).
Article
Werner, Sabine, and Richard Grose. Regulation of Wound Healing by Growth Factors and Cytokines. Physiol Rev 83: 835–870, 2003; 10.1152/physrev.00032.2002.—Cutaneous wound healing is a complex process involving blood clotting, inflammation, new tissue formation, and finally tissue remodeling. It is well described at the histological level, but the genes that regulate skin repair have only partially been identified. Many experimental and clinical studies have demonstrated varied, but in most cases beneficial, effects of exogenous growth factors on the healing process. However, the roles played by endogenous growth factors have remained largely unclear. Initial approaches at addressing this question focused on the expression analysis of various growth factors, cytokines, and their receptors in different wound models, with first functional data being obtained by applying neutralizing antibodies to wounds. During the past few years, the availability of genetically modified mice has allowed elucidation of the function of various genes in the healing process, and these studies have shed light onto the role of growth factors, cytokines, and their downstream effectors in wound repair. This review summarizes the results of expression studies that have been performed in rodents, pigs, and humans to localize growth factors and their receptors in skin wounds. Most importantly, we also report on genetic studies addressing the functions of endogenous growth factors in the wound repair process.
Article
The responses to a comprehensive survey of infection control practices for respiratory therapy equipment, previously published in the Readers' Forum section of Infection Control , are summarized in Tables 1 through 3. Responses were obtained from 205 United States hospitals; 65% were community hospitals and 38% had <500 ventilator days/year. In some cases, the total number of responses exceeded 100% because of practices that required multiple responses.
Article
The pathogenic yeast Cryptococcus neoformans secretes numerous proteins, such as heat shock proteins, by unconventional mechanisms during its interaction with host cells. Hsp70 is a conserved chaperone that plays important roles in various cellular processes, including the interaction of fungi with host immune cells. Here, we report that sera from individuals with cryptococcosis infection recognize a recombinant C. neoformans Hsp70 (Cn_rHsp70). Moreover, immunofluorescence assays using antibodies against Cn_rHsp70 revealed the localization of this protein at the cell surface mainly in association with the capsular network. We found that the addition of Cn_rHsp70 positively modulated the interaction of C. neoformans with human alveolar epithelial cells and decreased fungal killing by mouse macrophages, without affecting phagocytosis rates. Immunofluorescence analysis showed that there was a competitive association among the receptor, GXM and Cn_rHsp70, indicating that the Hsp70-binding sites in host cells appear to be shared by glucuronoxylomannan (GXM), the major capsular antigen in C. neoformans. Our observations suggest additional mechanisms by which Hsp70 influences the interaction of C. neoformans with host cells.
Article
Bacterial biofilm has been shown to play a role in delaying wound healing of chronic wounds, a major medical problem that results in significant health care burden. A reproducible animal model could be very valuable for studying the mechanism and management of chronic wounds. Our previous work showed that Pseudomonas aeruginosa (PAO1) biofilm challenge on wounds in diabetic (db/db) mice significantly delayed wound healing. In this wound time course study, we further characterize the bacterial burden, delayed wound healing, and certain aspects of the host inflammatory response in the PAO1 biofilm-challenged db/db mouse model. PAO1 biofilms were transferred onto 2-day-old wounds created on the dorsal surface of db/db mice. Control wounds without biofilm challenge healed by 4 weeks, consistent with previous studies; none of the biofilm-challenged wounds healed by 4 weeks. Of the biofilm-challenged wounds, 64% healed by 6 weeks, and all of the biofilm-challenged wounds healed by 8 weeks. During the wound-healing process, P. aeruginosa was gradually cleared from the wounds while the presence of Staphylococcus aureus (part of the normal mouse skin flora) increased. Scabs from all unhealed wounds contained 10(7) P. aeruginosa, which was 100-fold higher than the counts isolated from wound beds (i.e., 99% of the P. aeruginosa was in the scab). Histology and genetic analysis showed proliferative epidermis, deficient vascularization, and increased inflammatory cytokines. Hypoxia inducible factor expression increased threefold in 4-week wounds. In summary, our study shows that biofilm-challenged wounds typically heal in approximately 6 weeks, at least 2 weeks longer than nonbiofilm-challenged normal wounds. These data suggest that this delayed wound healing model enables the in vivo study of bacterial biofilm responses to host defenses and the effects of biofilms on host wound healing pathways. It may also be used to test antibiofilm strategies for treating chronic wounds.