ArticlePDF Available

Doxycycline Attenuates Peripheral Inflammation in Rat Experimental Autoimmune Neuritis

Authors:
  • Seventh Affiliated Hospital of Sun Yat-Sen University

Abstract and Figures

Experimental autoimmune neuritis (EAN) is a T cell-mediated autoimmune inflammatory demyelinating disease of the peripheral nervous system and widely-used animal model of human inflammatory demyelinating polyradiculoneuropathies. Doxycycline is a well-known antibiotic and has been reported to have neuroprotective and anti-inflammatory effects. Here we investigated the effects of doxycycline on rat EAN. Therapeutic treatment with doxycycline (40 mg/kg body weight daily from the Day 9 to Day 14 post immunization) significantly attenuated the severity of EAN, decreased inflammatory infiltration of macrophages, B- and T-cells and demyelination in sciatic nerves of EAN rats. Pro-inflammatory molecules including matrixmetalloproteinase-9, inducible nitric oxide synthase and interleukin-17 were greatly decreased in sciatic nerves by administration of doxycycline as well. Taken together, our data showed that doxycycline could effectively suppress the peripheral inflammation to improve outcome of EAN, which suggests that doxycycline may be considered as a potential candidate of pharmacological treatment for neuropathies.
Content may be subject to copyright.
ORIGINAL PAPER
Doxycycline Attenuates Peripheral Inflammation in Rat
Experimental Autoimmune Neuritis
Chenju Yi Zhiyuan Zhang Wei Wang
Caroline Zug Hermann J. Schluesener
Zhiren Zhang
Accepted: 28 May 2011 / Published online: 8 June 2011
ÓSpringer Science+Business Media, LLC 2011
Abstract Experimental autoimmune neuritis (EAN) is a
T cell-mediated autoimmune inflammatory demyelinating
disease of the peripheral nervous system and widely-used
animal model of human inflammatory demyelinating
polyradiculoneuropathies. Doxycycline is a well-known
antibiotic and has been reported to have neuroprotective
and anti-inflammatory effects. Here we investigated the
effects of doxycycline on rat EAN. Therapeutic treatment
with doxycycline (40 mg/kg body weight daily from the
Day 9 to Day 14 post immunization) significantly attenu-
ated the severity of EAN, decreased inflammatory infil-
tration of macrophages, B- and T-cells and demyelination
in sciatic nerves of EAN rats. Pro-inflammatory molecules
including matrixmetalloproteinase-9, inducible nitric oxide
synthase and interleukin-17 were greatly decreased in sciatic
nerves by administration of doxycycline as well. Taken
together, our data showed that doxycycline could effec-
tively suppress the peripheral inflammation to improve
outcome of EAN, which suggests that doxycycline may be
considered as a potential candidate of pharmacological
treatment for neuropathies.
Keywords Doxycycline EAN Inflammation
Sciatic nerves
Introduction
Experimental autoimmune neuritis (EAN) is an autoantigen-
specific T-cell-mediated inflammatory peripheral nervous
system (PNS) demyelinating animal model [1]. EAN can be
induced in susceptible animals by active immunization with
whole PNS myelin or its components, the proteins P2 or P0,
as well as their immunogenic peptides [2]. EAN is patho-
logically characterized by breakdown of the blood-nerve
barrier, infiltration of reactive immune cells, local inflam-
mation and demyelination in the peripheral nervous. EAN
shares many clinical, electrophysiological and immunolog-
ical features of human acute inflammatory demyelinating
polyradiculoneuropathy (AIDP) and has been widely used
as an animal model to study disease mechanism and therapy
of AIDP. Treatments of AIDP include plasma exchange,
intravenous immunoglobulin or supportive management
such as intensive care and respiratory assistance. But all of
these treatments are unsatisfying [1], and new therapeutic
options are therefore needed.
Doxycycline is a tetracycline antibiotic. Besides its
broad-spectrum antibiotic activity, doxycycline has been
shown to display neuroprotective and anti-inflammatory
properties in animal models of global and focal cerebral
ischemia [3,4], multiple sclerosis [5], Parkinson’s disease
[6]; and in clinical trials of Alzheimer’s disease, lym-
phangioleiomyomatosis, osteoarthritis and others [7].
Neuroprotective and anti-inflammatory effects of doxycy-
cline in these disorders are associated with inhibition of
matrix metalloproteases [8,9], depression of oxygen radi-
cal release from polymorphonuclear neutrophils [10],
C. Yi and Z. Zhang contributed equally to this work.
C. Yi Z. Zhang C. Zug H. J. Schluesener Z. Zhang (&)
Institute of Brain Research, University of Tuebingen,
Calwer Street 3, 72076 Tuebingen, Germany
e-mail: zhangzhiren@yahoo.com
C. Yi W. Wang
Department of Neurology, Tongji Hospital, Tongji Medical
College, Huazhong University of Science and Technology,
Wuhan, People’s Republic of China
Z. Zhang
Institute of Immunology, Third Military Medical University
of PLA, Chongqing, People’s Republic of China
123
Neurochem Res (2011) 36:1984–1990
DOI 10.1007/s11064-011-0522-2
inhibition of inducible nitric oxide synthase and inter-
leukin-1b-converting enzyme [3], reduction of cleaved
caspase-3 protein expression and decrease of microglial
activation [11,12].
Doxycycline is commonly used as an antibiotic and its
toxicity has been characterized in detail [13]. Additionally,
the high lipophilicity of doxycycline allows it to diffuse
into the central and peripheral nervous system (CNS and
PNS) at therapeutically effective levels [14]. Therefore, the
proven reliability and safety of doxycycline suggest its
potential prospect as an effective and cheap treatment of
human inflammatory neuropathies. In our present investi-
gation, we determined the potential protective effect of
doxycycline on rat EAN.
Experimental Procedure
Animal Experiments
Male Lewis rats (8–10 wee ks old, 200–250 g, Charles River,
Sulzfeld, Germany) were housed with equal daily periods of
light and dark and free access to food and water. All proce-
dures were performed in accordance with the published
International Health Guidelines under a protocol approved
by the local Administration District Official Committee. All
efforts were made to minimize the number of animals and
their suffering. EAN was induced by subcutaneous injection
at the base of the tails with 100 ll of an inoculum containing
100 lg of synthetic neuritogenic P2
53–78
peptide (Gene
Script Corporation, Scotch Plains, NJ, USA). The peptide
was dissolved in phosphate-buffered saline (PBS) (2 mg/ml)
and then emulsified with an equal volume of complete Fre-
und’s adjuvant (CFA) containing 2 mg/ml Mycobacterium
tuberculosis to get a final concentration of 1 mg/ml. The
severity of EAN was scored daily as follows: 0-normal,
1-reduced tonus of tail, 2-limp tail, impaired righting,
3-absent righting, 4-gait ataxia, 5-mild paresis of the hind
limbs, 6-moderate paraparesis, 7-severe paraparesis or
paraplegia of the hind limbs, 8-tetraparesis, 9-moribund,
10-death. As soon as first neurological signs were observed,
all the immunized rats were randomly divided into two
groups and were treated with doxycycline and control,
respectively. For therapeutic treatment, doxycycline (Sigma,
St. Louis, MO, USA; 40 mg/kg body weight in 1 ml PBS)
was intraperitoneally injected once daily from the Day 9 to
Day 14 post immunization. The PBS control group received
the same volume of PBS.
LFB Staining and Immunohistochemistry
Luxol fast blue (LFB) staining was applied to show
myelin and to evaluate demyelination of sciatic nerves.
For LFB staining, histological changes between doxy-
cycline and PBS treated EAN rats were compared by an
established semi-quantitative method. Briefly, four cross-
sections from sciatic nerves of both sides of EAN rats
were analyzed. All perivascular areas present in cross-
section were evaluated by two observers unaware of
treatment, and the degree of pathological alteration was
graded semi-quantitatively by the following scale:
0=normal perivascular area; 1 =mild cellular infiltra-
tion adjacent to the vessel; 2 =cellular infiltration plus
demyelination in immediate proximity to the vessel;
3=cellular infiltration and demyelination throughout the
section. Results were given as means of histological
scores [15]. Results were calculated as arithmetic means
of positive cells per square millimeter and standard
errors of means (SEM).
Immunohistochemistry (IHC) was performed on 3 lm
paraffin-embedded sections using antibodies serially to
evaluate local inflammation, cellular infiltration and
demyelination in sciatic nerves: ED-1 (1:100; Serotec,
Oxford, UK) to detect activated microglia/macrophages,
OX22 (1:200; Serotec, Oxford, UK) predominantly for B
cells or CD3 (1:50; Serotec, Oxford, UK) predominantly
for T-lymphocytes, Interleukin-17 (IL-17; 1:100; Santa
Cruz Biotechnology, Santa Cruz, CA, USA), iNOS (1:100;
Serotec, Oxford, UK) and MMP9 (matrix metalloprotein-
ase-9; 1:500; Neuromics, Edina, MN, USA). After
dewaxing, sections were boiled (in an 850 W microwave
oven) for 15 min in citrate buffer (2.1 g citric acid
monohydrate/L, pH6) (Carl Roth, Karlsruhe, Germany).
Endogenous peroxidase was inhibited by 1% H
2
O
2
in pure
methanol (Merck, Darmstadt, Germany) for 15 min. Sec-
tions were incubated with 10% normal pig serum (Bio-
chrom, Berlin, Germany) to block non-specific binding of
immunoglobulins and then with the primary antibodies
overnight at 4°C. Antibodies binding to tissue sections
were visualized with secondary biotinylated antibodies
(rabbit anti-mouse or rabbit anti-goat) (1:400; DAKO,
Hamburg, Germany). Subsequently, sections were incu-
bated with a Streptavidin–Avidin–Biotin complex (DAKO,
Hamburg, Germany), followed by development with
diaminobenzidine (DAB) substrate (Fluka, Neu-Ulm,
Germany). Finally, sections were counterstained with
hemalum. As negative controls, the primary antibodies
were omitted.
After immunostaining, sections from doxycycline and
PBS control groups were examined by light microscopy
and the numbers of ED-1
?
,OX22
?
, CD3
?
, IL-17
?
,
iNOS
?
and MMP-9
?
cells were counted. Positively
stained cell counting based on IHC results has been well
developed to semi-quantify protein expression [16]. The
numbers of positively stained cells were counted by two
investigators independently and only positive cells with
Neurochem Res (2011) 36:1984–1990 1985
123
the nucleus at the focal plane were counted. The sections
were randomly numbered and observers were not aware
of the time points and treatments. To evaluate positive
cell numbers in sciatic nerves, four cross-sections for
each rat were evaluated. Microphotos of the whole sci-
atic nerve cross-sections were taken under 200 9mag-
nification using Nikon Coolscope (Nikon, Du
¨sseldorf,
Germany) and only positive cells with the nucleus at the
focal plane were counted. Areas of sciatic nerve cross-
sections were measured on the same pictures using
software MetaMorph Offline 7.1 (Molecular Devices,
Toronto, Canada).
Statistical Analysis
Statistical analysis was performed by one-way ANOVA
followed by Dunnett’s multiple comparison test (Graph
Pad Prism 4.0 software). For all statistical analyses, sig-
nificance levels were set at P\0.05.
Results
Effects of Doxycycline on Pathological Scores
and Body Weight of EAN Rats
EAN was induced by subcutaneous injection of P2 peptide.
The first neurologic sign (reduced tonus of tail) of EAN
rats was observed at day 9 (mean clinical score ±SEM:
0.20 ±0.20). For therapeutic treatment, doxycycline or
PBS (PBS control group) was injected once daily from the
onset of neurological signs (day 9–14 post immunization).
The neurologic severity of EAN increased fast in the
control group with a maximal score at day 15 (mean
clinical score ±SEM: 6.8 ±0.20). However, doxycycline
therapeutic treatment suppressed severity of EAN (maxi-
mal mean score at day 15: 4.0 ±0.32, P\0.05 compared
to PBS control) from day 12 to 15 (P\0.05, compared to
respective control) (Fig. 1a).
Progressive weight loss during onset of EAN is another
characteristic feature of this disease, which correlates with
Fig. 1 Doxycycline reduced pathological scores and mean histolog-
ical scores. EAN was induced by subcutaneous injection of synthetic
neuritogenic P2 peptide (100 ug) and CFA into the base of the tails.
Doxycycline or PBS (five rats each, 40 mg/kg body weight in 1 ml
PBS) were injected once daily starting from 9th day until 14th day
after immunization. Clinical scores were taken from day 3 after
immunization. aDoxycycline treatment significantly decreased
neurologic severity of EAN. bDoxycycline suppressed histopathol-
ogical alterations in sciatic nerves of EAN rats. 15 days after
immunization, sciatic nerves of doxycyline treated and PBS control
rats were taken and used for LFB staining, followed by hematoxylin
counterstaining. Representative micrographs for PBS or doxycycline
treated EAN rats are shown in cand d, respectively. Means of
histological scores were calculated as described in ‘‘Experimental
Procedures’’ . Bar figure shows that doxycycline treatment signifi-
cantly reduced mean histological scores as compared to PBS controls.
The unpaired ttest was performed (Graph Pad Prism 4.0 for
Windows). * P\0.05 compared to the PBS control
1986 Neurochem Res (2011) 36:1984–1990
123
the severity of EAN. A progressive weight loss was observed
during the onset of EAN from Day 9 to Day 15 in PBS treated
EAN rats. In contrast, the weight of doxycycline-treated rats
increased slightly progressively after day 12. Significant
differences of body weight between PBS- and doxycycline-
treated EAN rats from day 12 to day 15 was proven by one-
way ANOVA followed by Dunnett’s multiple comparison
test (P\0.05)(data not showed). Taken together, these
results indicated a significantly reduced disease severity in
doxycycline-treated EAN.
Effects of Doxycycline on Demyelination and Cell
Infiltration in EAN Sciatic Nerves
Sciatic nerves were taken from doxycycline-treated and
PBS control EAN rats (n=5) at Day 15 and were
analyzed histologically. LFB staining demonstrated a
significant decreased degree of perivascular demyelina-
tion and inflammatory cell infiltration in EAN rats
treated with doxycycline (mean histological score 1.45 ±
0.09) (Fig. 1b, d), as compared to the PBS control group
(mean histological score 2.13 ±0.11) (Fig. 1b, c),
P\0.05.
Distinct infiltration of different types of inflammatory
cells into sciatic nerves was further analyzed by IHC.
Dense inflammatory accumulation of macrophages
(ED-1
?
) (Fig. 2a), T cells (CD3
?
) (Fig. 2b), B cells
(OX22
?
) (Fig. 2c) were seen in sciatic nerves of rats from
the PBS control group at Day 15 and the most dominant
cell population were macrophages, with a mean density of
652.2 ±77.26 cells/mm
2
(Fig. 2g). Doxycycline treatment
significantly decreased inflammatory cell accumulation
Fig. 2 Doxycycline suppressed macrophage, T cell and B cell
infiltration into sciatic nerves of EAN rats. 15 days after immuniza-
tion, sciatic nerveswere analyzed by immunohistochemstry. ED-1
(aand d) immunostaining was used for macrophages, CD3 (band
e) for T cells and OX22 (cand f) for B cells. Representative
micrographs from doxycycline treated and PBS control EAN rats are
shown in (ac) and (bd), respectively. Infiltration of macrophages,
T- and B-cells into sciatic nerves was further semi-quantified as
indicated in Experimental Procedures and the figures show the
quantified results (gi). Doxycycline treatment significantly reduced
infiltration of macrophages, T cell and B cells into EAN sciatic nerves
(doxy: doxycycline treatment). The unpaired ttest was performed to
compare the differences (Graph Pad Prism 4.0 for Windows).
*P\0.05 compared to their respective control
Neurochem Res (2011) 36:1984–1990 1987
123
(P\0.05, compared to PBS control, respectively)
(Fig. 2d–i).
Effects of Doxycycline on Levels of IL-17, iNOS
and MMP-9 in EAN Sciatic Nerves
We further studied cellular expression of several important
inflammatory cytokines/molecule. In sciatic nerves from
control EAN rats receiving injections of PBS only, IL-17,
iNOS and MMP9 were observed in aggregated immune
cells. Doxycycline treatment strikingly reduced locally
accumulated IL-17
?
(Fig. 3a, d, g), iNOS
?
(Fig. 3b, e, h)
and MMP9
?
(Fig. 3c, f, i) cells in EAN sciatic nerves
(P\0.05, compared to PBS controls separately)
Discussion
Here we have studied the therapeutic effects of doxycy-
cline on EAN, which is the prime animal model for AIDP
and useful for investigating therapeutic approaches. Ther-
apeutic treatment with doxycycline significantly reduced
neurologic severity of EAN through reducing local demy-
elination, suppression of local inflammatory cell infiltration
and decreasing expression of pro-inflammatory molecules
MMP-9, iNOS and IL-17 in sciatic nerves.
As a member of the tetracycline class of antibiotics,
doxycycline has been shown interesting pleiotropic prop-
erties, such as neuroprotective and anti-inflammatory
effects. In vitro and in vivo data have suggested that
doxycycline inhibited inflammation by modulating cellular
activation and subsequent release of cytokines, chemo-
kines, lipid mediators of inflammation, MMPs and nitric
oxide [14]. In our EAN models, doxycycline greatly sup-
pressed infiltration of T cells, B cells and macrophages into
peripheral nerves and thus greatly reduced the patholog-
icalinfiltration of reactive leucocyte into the PNS [17].
Doxycycline inhibited MMP expression in sciatic nerves
to reduce immune cell infiltration in sciatic nerves of EAN
rats. MMPs, particularly MMP-9, could participate in the
disruption of the BNB, breakdown of the myelin sheath,
the release of TNF-a, and finally facilitate leukocyte
invasion into the PNS [18]. Previous studies have shown an
increased expression of MMP-9 in inflamed peripheral
nerves in EAN [19]. In the nervous system, aberrant
expression of MMPs may support disease activity by
converting pro-forms of several inflammatory molecules,
such as TNF-a, into their active forms, resulting in the
Fig. 3 Doxycycline suppressed
accumulation of IL-17, iNOS
and MMP-9 in sciatic nerves of
EAN rats. 15 days after
immunization, sciatic nerves of
both groups were taken and
used for immunohistochemical
staining of IL-17, iNOS and
MMP-9 (PBS control: a,band
cseparately; Doxycyline group:
d,eand fseparately).
Representative
immunohistochemical
micrographs showed that
doxycycline significantly
reduced the expression of IL-17,
iNOS and MMP-9 in sciatic
nerves. gibar figures show
quantified results. The unpaired
ttest was performed to compare
the differences between
doxycycline treated and PBS
control EAN rats (Graph Pad
Prism 4.0 for Windows).
** P\0.05, compared to their
respective control
1988 Neurochem Res (2011) 36:1984–1990
123
propagation of inflammation [20]. In addition, MMPs were
reported to induce the degradation of myelin or axonal
injury after injection into the brain [21], and fragments of
MMP-mediated digestion of myelin basic protein are
encephalitogenic when injected into mice. Thereby, a cas-
cade of demyelinating and pro-inflammatory events is
generated in the nervous system as a result of aberrant MMP
expression [22]. Doxycycline not only inhibits the enzy-
matic activity of MMPs, but also reduces the expression of
several other MMP family members [23]. Here, our data
showed that doxycycline reduced MMP-9 level in sciatic
nerves of EAN rats, which could not only inhibit leucocyte
infiltration but also diminish their effects on demyelination.
Doxycycline could also attenuate inflammatory cyto-
kines in sciatic nerves. Cytokines are produced and
released by many cell types and regulate inflammation and
immunity. Pro-inflammatory cytokines, such as IL-17
produced by Th cells, augment both inflammation and
subsequent immune responses [24]. IL-17 stimulates pro-
duction of IL-6, nitric oxide and prostaglandin E2 to
amplify local inflammation, mediates chemotaxis of neu-
trophils and monocytes to sites of inflammation and aug-
ments the induction of co-stimulatory molecules such as
ICAM-1 to support T cell activation [25,26]. Similar to
inflammatory cytokines, iNOS is up-regulated in EAN and
known to play important roles in inflammatory progression
of disease [27,28]. iNOS functions to produce nitric oxide
which possesses pro-inflammatory property including
vasodilation, oedema, cytotoxicity and mediates cytokine-
dependent processes that can result in tissue destruction
[29]. In EAN, upregulation of iNOS was reported and was
particularly related to pathogenesis of PNS cell-mediated
demyelination and even axonal damage [30]. In our study,
doxycycline reduced lesional iNOSs, which may inhibit
activation of immune cells, and be an important additional
part of doxycycline’s anti-inflammatory effect. Interest-
ingly, inhibition of iNOS expression is considered to be a
major mechanism of doxycycline [14]. Accumulated data
propose several potential mechanisms of inhibitory activity
of doxycycline on macrophage activity, but the exact
mechanisms are not clear yet. One of the potential mech-
anisms is inhibition of the expression of iNOS, resulting
in reduced release of NO and NO induced phosphorylation
of p38 MAPK (mitogen-activated protein kinase) [23].
Taken together, doxycycline greatly attenuated peripheral
inflammation and decreased local expression/release of
multiple inflammatory cytokines/molecules.
It is well known that another tetracycline, minocycline,
also has anti-inflammatory and neuroprotective effects
similar to doxycycline, as we had reported previously [31].
However, minocycline is also known for its side effects,
such as vestibular toxicity [32] and effects on bone and
teeth formation. Recently, decreased tolerability of
minocycline has also been raised as a concern during a
clinical trial of Parkinson’s Disease [33]. In contrast,
doxycycline has the least toxic side effects among tetra-
cyclines [34]. Although doxycycline and minocycline have
similar chemical structures, they have been reported to
exhibit differences in patterns of neuroprotection in some
models of cerebral ischemia [3,11,35] and in antioxidant
activity [36,37]. Additionally, doxycycline is a more
potent inhibitor of MMPs [37]. These data suggest that
doxycyline has more advantages in clinical applications.
In conclusion, doxycycline is inexpensive and easily
obtainable, could effectively suppress peripheral inflam-
mation to improve outcome in EAN rats. Our results sug-
gest that doxycycline should be considered a potential
candidate toward the therapeutic strategy for autoimmune
neuropathies.
Acknowledgments Chenju Yi gratefully acknowledged China
Scholarship Council. This investigation was partly supported by
National Nature Science Foundation of China (No: 81070954).
References
1. Hughes RA, Cornblath DR (2005) Guillain-Barre
´syndrome.
Lancet 366:1653–1666
2. Milner P, Lovelidge CA, Taylor WA et al (1987) P0 myelin
protein produces experimental allergic neuritis in Lewis rats.
J Neurol Sci 79:275–285
3. Yrjanheikki J, Keinanen R, Pellikka M et al (1998) Tetracyclines
inhibit microglial activation and are neuroprotective in global
brain ischemia. Proc Natl Acad Sci USA 95:15769–15774
4. Yrjanheikki J, Tikka T, Keinanen R et al (1999) A tetracycline
derivative, minocycline, reduces inflammation and protects
against focal cerebral ischemia with a wide therapeutic window.
Proc Natl Acad Sci USA 96:13496–13500
5. Popovic N, Schubart A, Goetz BD et al (2002) Inhibition of autoim-
mune encephalomyelitis by a tetracycline. Ann Neurol 51:215–223
6. Wu DC, Jackson-Lewis V, Vila M et al (2002) Blockade of
microglial activation is neuroprotective in the 1-methyl-4-phenyl-
1, 2, 3, 6-tetrahydropyridine mouse model of Parkinson disease.
J Neurosci 22:1763–1771
7. Griffin MO, Fricovsky E, Ceballos G et al (2010) Tetracyclines: a
pleitropic family of compounds with promising therapeutic
properties. Am J Physiol Cell Physiol 299:539–548
8. Paemen L, Martens E, Norga K et al (1996) The gelatinase
inhibitory activity of tetracyclines and chemically modified tet-
racycline analogues as measured by a novel microtiter assay for
inhibitors. Biochem Pharmacol 52:105–111
9. Golub LM, Ramamurthy NS, McNamara TF et al (1991) Tetra-
cyclines inhibit connective tissue breakdown: new therapeutic
implications for an old family of drugs. Crit Rev Oral Biol Med
2:297–321
10. Gabler WL, Creamer HR (1991) Suppression of human neutro-
phil functions by tetracyclines. J Periodontal Res 26:52–58
11. Jantzie LL, Cheung PY, Todd KG (2005) Doxycycline reduces
cleaved caspase-3 and microglial activation in an animal model of
neonatal hypoxia-ischemia. J Cereb Blood Flow Metab 25:314–324
12. Jantzie LL, Rauw GA, Todd KG (2006) The effects of doxycy-
cline administration on amino acid neurotransmitters in an animal
model of neonatal hypoxia-ischemia. Neurochem Int 49:717–728
Neurochem Res (2011) 36:1984–1990 1989
123
13. Zouboulis CC, Piquero-Martin J (2003) Update and future of
systemic acne treatment. Dermatology 206:37–53
14. Stirling DP, Koochesfahani KM, Steeves JD et al (2005) Mino-
cycline as a neuroprotective agent. Neuroscientist 11:308–322
15. Hartung HP, Scha
¨fer B, Heininger K et al (1988) The role of
macrophages and eicosanoids in the pathogenesis of experimental
allergic neuritis Serial clinical, electrophysiological, biochemical
and morphological observations. Brain 111:1039–1059
16. Leifeld L, Fielenbach M, Dumoulin FL et al (2002) Inducible
nitric oxide synthase (iNOS) and endothelial nitric oxide synthase
(eNOS) expression in fulminant hepatic failure. J Hepatol 37:
613–619
17. Schabet M, Whitaker JN, Schott K et al (1991) The use of pro-
tease inhibitors in experimental allergic neuritis. J Neuroimmunol
31:265–272
18. Hughes PM, Wells GM, Clements JM (1998) Matrix metallo-
proteinase expression during experimental autoimmune neuritis.
Brain 121:481–494
19. Kieseier BC, Clements JM, Pischel HB et al (1998) Matrix
metalloproteinases MMP-9 and MMP-7 are expressed in exper-
imental autoimmune neuritis and the Guillain-Barre
´syndrome.
Ann Neurol 43:427–434
20. Brundula V, Rewcastle NB, Metz LM et al (2002) Targeting
leukocyte MMPs and transmigration: minocycline as a potential
therapy for multiple sclerosis. Brain 125:1297–1308
21. Newman JP, Verity AN, Hawatmeh S et al (1996) Ciliary neu-
rotrophic factors enhances peripheral nerve regeneration. Arch
Otolaryngol Head Neck Surg 122:399–403
22. Opdenakker G, Van Damme J (1994) Cytokine-regulated prote-
ases in autoimmune diseases. Immunol Today 15:103–107
23. Curci JA, Mao D, Bohner DG et al (2001) Preoperative treatment
with doxycycline reduces aortic wall expression and activation of
matrix metalloproteinases in patients with abdominal aortic
aneurysms. J Vasc Surg 31:325–342
24. Saliba E, Henrot A (2001) Inflammatory mediators and neonatal
brain damage. Biol Neonate 79:224–227
25. Bettelli E, Korn T, Oukka M et al (2008) Induction and effector
functions of T(H)17 cells. Nature 453:1051–1057
26. Dong C (2008) TH17 cells in development: an updated view of
their molecular identity and genetic programming. Nat Rev
Immunol 8:337–348
27. Lee Y, Shin T (2002) Expression of constitutive endothelial and
inducible nitric oxide synthase in the sciatic nerve of Lewis rats
with experimental autoimmune neuritis. J Neuroimmunol 126:
78–85
28. Zhu J, Mix E, Link H (1998) Cytokine production and the
pathogenesis of experimental autoimmune neuritis and Guillain-
Barre
´syndrome. J Neuroimmunol 84:40–52
29. Abramson SB, Amin AR, Clancy RM et al (2001) The role of
nitric oxide in tissue destruction. Best Pract Res Clin Rheumatol
15:831–845
30. Conti G, Rostami A, Scarpini E et al (2004) Inducible nitric oxide
synthase (iNOS) in immune-mediated demyelination and Wal-
lerian degeneration of the rat peripheral nervous system. Exp
Neurol 187:350–358
31. Zhang ZY, Zhang ZR, Fauser U et al (2009) Improved outcome
of EAN, an animal model of GBS, through amelioration of
peripheral and central inflammation by minocycline. J Cell Mol
Med 13:341–351
32. Cunha BA (2006) New uses for older antibiotics: nitrofurantoin,
amikacin, colistin, polymyxin B, doxycycline, and minocycline
revisited. Med Clin North Am 90:1089–1107
33. NINDS NET-PD Investigators (2008) A pilot clinical trial of
creatine and minocycline in early Parkinson disease: 18-month
results. Clin Neuropharmacol 31:141–150
34. Toth A, Lesser ML, Naus G et al (1988) Effect of doxycycline on
pre-menstrual syndrome: a double-blind randomized clinical trial.
J Int Med Res 16:270–279
35. Fox C, Dingman A, Derugin N et al (2005) Minocycline confers
early but transient protection in the immature brain following
focal cerebral ischemia-reperfusion. J Cereb Blood Flow Metab
25:1138–1149
36. Kraus RL, Pasieczny R, Lariosa-Willingham K et al (2005)
Antioxidant properties of minocycline: neuroprotection in an
oxidative stress assay and direct radical-scavenging activity.
J Neurochem 94:819–827
37. Yao JS, Shen F, Young WL et al (2007) Comparison of doxy-
cycline and minocycline in the inhibition of VEGF-induced
smooth muscle cell migration. Neurochem Int 50:524–530
1990 Neurochem Res (2011) 36:1984–1990
123
... In addition, many dermatoses are considered to be trivial, with the result that they become ''orphan diseases'' with no medications approved for treatment of the indication. The anti-inflammatory activities of antimicrobials have been widely reported [2][3][4], but the non-antifungal activities of antifungal agents are less well known. In this review, we explore the possible non-antifungal use of itraconazole in dermatology and discuss the possible modes of action of this agent (Table 1). ...
Article
Full-text available
Introduction The anti-inflammatory and pro-kinetic properties of antibiotics have been widely reported. However, the non-antifungal properties of antifungal agents are less well known and less explored in clinical practice. The purpose of this review was to survey the literature on the non-antifungal use of itraconazole in dermatological practice and the possible modes of action of this agent. Methods The PubMed database was searched for relevant articles published up to January 2017. The references in the articles identified by the search were then hand-searched for additional relevant publications. Results Itraconazole displays a great diversity of non-antifungal activity and has been used to treat a broad spectrum of diseases. The results of our survey reveal that itraconazole has the potential to be an alternative agent for treating patients with advanced cancer (either alone or in combination with other cytotoxic chemotherapeutic drugs), especially those refractory to traditional treatments. Moreover, itraconazole acts as an anti-angiogenesis agent, induces nail growth, and modulates inflammatory or immune diseases. Conclusion Oral antifungal agents have many non-antifungal properties. However, the body of evidence on individual agents often remains limited due to the lack of large-scale randomized controlled studies. Although some of the findings published to date seem promising, pharmacological vigilance should be taken for off-label use in real-world practice.
... 140 Tetracyclines, alone or in combination, have been used for the systemic treatment of HS. 141,142 In various animal models, tetracyclines decreased Th17 differentiation and reduced IL-17 levels. [143][144][145][146] Retinoids Retinoids exert beneficial effects in the treatment of HS. 147 Huang and Kirchhof 148 showed that isotretinoin (13-cis-retinoic acid) exhibited a partial response in 32% and a complete response in 36% of patients with HS. These data support the view that at least a subgroup of patients with HS responds to isotretinoin, the prodrug of all-trans retinoic acid (ATRA). ...
Article
Background Disintegration of the infundibula of terminal hair follicles (HF) in intertriginous skin areas exhibits the histological hallmark of hidradenitis suppurativa (HS)/acne inversa, featuring a dissecting terminal hair folliculitis. Elevated serum levels of interleukin 17 and local increase in the ratio of proinflammatory Th17 cells and antiinflammatory regulatory T cells (Tregs) have been reported. Perifollicular Tregs play a key role for HF stem cell homeostasis and infundibular integrity. Objectives In this review, we evaluate the Th17/Treg ratio in HS, its aggravating conditions and associated comorbidities. Furthermore, we intended to clarify whether drugs with reported beneficial effects in HS readjust the deviated Th17/Treg axis. Methods PubMed‐listed, peer‐reviewed original research articles characterizing Th17/Treg regulation in hidradentitis suppurativa/acne inversa and associated comorbidities have been selected. Results This review presents HS as a disease exhibiting an increased Th17/Treg ratio. Perifollicular deficiencies in Treg numbers or function may disturb HF stem cell homeostasis initiating infundibular dissection of terminal HFs and perifollicular inflammation. The Th17/Treg imbalance is aggravated by obesity, smoking as well as decreased Notch signalling. In addition, HS‐associated autoimmune diseases exhibit a disturbed Th17/Treg axis resulting in a Th17‐dominant state. All drugs that have beneficial effects in the treatment of HS normalise the Th17/Treg ratio. Conclusions HS immunopathogenesis is closely related to deviations of the Th17/ Treg balance, which may negatively affect Treg‐controlled HF stem cell homeostasis and infundibular integrity. Pharmacological intervention should not only attenuate Th17/IL‐17 signalling but should improve Treg function to stabilize HF stem cell homeostasis and infundibular integrity This article is protected by copyright. All rights reserved.
... Tetracyclines have been shown to have immunomodulatory and anti-inflammatory effects in several inflammatory conditions, such as multiples sclerosis, Parkinson's disease and rheumatoid arthritis [57][58][59][60] . Furthermore, doxycycline suppressed the proliferation of lymphocytes 61 and minocycline inhibited proliferation and reduced production of IL-2, IFNγ and TNFα in human T-cells 62 . ...
Article
Full-text available
Inflammatory bowel disease (IBD) may develop due to an inflammatory response to commensal gut microbiota triggered by environmental factors in a genetically susceptible host. Isotretinoin (acne therapy) has been inconsistently associated with IBD onset and flares but prior treatment with antibiotics, also associated with IBD development, complicates the confirmation of this association. Here we studied in mice whether doxycycline, metronidazole or isotretinoin induce epigenetic modifications, and consequently change T-cell mRNA expression and/or function directly after treatment and after a 4 week recovery period. Isotretinoin induced IL-10 signaling in Tregs and naive T-cells directly after treatment and reduced effector T-cell proliferation alone and in co-culture with Tregs. Metronidazole activated processes associated with anti-inflammatory pathways in both T-cell subsets directly after the treatment period whereas doxycycline induced an immediate pro-inflammatory expression profile that resolved after the recovery period. Long-term changes indicated an inhibition of proliferation by doxycycline and induction of beneficial immune and metabolic pathways by metronidazole. Persistent alterations in microRNA and mRNA expression profiles after the recovery period indicate that all three medications may induce long-term epigenetic modifications in both T-cell subsets. Yet, our data do not support the induction of a long-term pro-inflammatory phenotype in murine Tregs and naive T-cells.
... In this study, in rats treated with Dox, bicyclam, and their combination, the withdrawal responses to both Von Frey filaments and pressure paw test in operated hind limb were significantly elevated compared to the saline group. These findings confirm earlier observation in experimental autoimmune neuritis model of the sciatic nerve by Yi and his coworkers [42], who reported that administration of Dox for 5 days reduced inflammatory cells, e.g. macrophages, B-and T-lymphocytes infiltration, and demyelination in sciatic nerves due to matrix metalloproteinase-9 inhibition . ...
Article
Chemokines have been recently recognized to play a role in chronic pain syndromes’ pathophysiology. This study investigated the role of monocyte chemoattractant protein-1 (MCP-1), stromal cell derived factor-1 (SDF-1), and retinoic acid (RA) as targets for the therapeutic approach of neuropathic pain. A chronic constriction injury (CCI) model of neuropathic pain by unilateral ligation of left sciatic nerve was performed in adult female Wistar rats. The effects of doxycycline (Dox, 50 mg/kg/day i.p. for 7 days), single dose of bicyclam (5 mg/kg i.p.), RA (15 mg/kg/day i.p. for 7 days), and their combination(s) on behavioral tests of nociception (Von Frey filaments; paw pressure test) on days 0, 1, 3, 5, and 7 of operation were studied. Serum concentrations of MCP-1 and SDF-1 were measured by ELISA. Histological examination of the sciatic nerve was investigated. CCI of sciatic nerve significantly induced mechanical allodynia and hyperalgesia and an increase of MCP-1 and SDF-1 serum levels. Dox-treated groups (Dox, Dox+bicyclam, Dox+RA, Dox+bicyclam+RA) and bicyclam-treated groups (bicyclam, Dox+bicyclam, bicyclam+RA, Dox+bicyclam+RA) attenuated CCI-induced behavioral and biochemical changes. RA inhibited CCI-induced mechanical hyperalgesia but produced a time-dependent reversal of allodynia. Histological findings showed degenerative changes of sciatic nerve after CCI that were partially recovered in Dox-treated groups. These findings demonstrate an association between serum MCP-1 and SDF-1 concentrations and behavioral manifestations of neuropathic pain. RA administration decreased neuropathic pain (antihyperalgesic effect) but did not cause any improvement in sciatic nerve tissues, either alone or in combination with chemokine antagonists. Thus, chemokines may serve as potential targets for drug development in neuropathic pain treatment.
Article
Full-text available
Multiple sclerosis (MS) is a chronic, autoimmune, demyelinating disease of the central nervous system (CNS). Microbes, including bacteria and certain viruses, particularly Epstein–Barr virus (EBV), have been linked to the pathogenesis of MS. While there is currently no cure for MS, antibiotics and antivirals have been studied as potential treatment options due to their immunomodulatory ability that results in the regulation of the immune process. The current issue addressed in this systematic review is the effect of antimicrobials, including antibiotics, antivirals, and antiparasitic agents in animals and humans. We performed a comprehensive search of PubMed, Google Scholar, and Scopus for articles on antimicrobials in experimental autoimmune encephalomyelitis animal models of MS, as well as in people with MS (pwMS). In animal models, antibiotics tested included beta-lactams, minocycline, rapamycin, macrolides, and doxycycline. Antivirals included acyclovir, valacyclovir, and ganciclovir. Hydroxychloroquine was the only antiparasitic that was tested. In pwMS, we identified a total of 24 studies, 17 of them relevant to antibiotics, 6 to antivirals, and 1 relevant to antiparasitic hydroxychloroquine. While the effect of antimicrobials in animal models was promising, only minocycline and hydroxychloroquine improved outcome measures in pwMS. No favorable effect of the antivirals in humans has been observed yet. The number and size of clinical trials testing antimicrobials have been limited. Large, multicenter, well-designed studies are needed to further evaluate the effect of antimicrobials in MS.
Article
Full-text available
Introduction Limited data from clinical trials in multiple sclerosis (MS) reported that minocycline, a widely used antibiotic belonging to the family of tetracyclines (TCs), exerts a beneficial short-lived clinical effect A similar anti-inflammatory effect of minocycline attributed to a deviation from Th1 to Th2 immune response has been reported in experimental models of MS. Whether such an immunomodulatory mechanism is operated in the human disease remains largely unknown. Aim To assess the in vitro immunomodulatory effect of tetracyclines, and in particular minocycline and doxycycline, in naïve and treated patients with MS. Material and Methods Peripheral blood mononuclear cells from 45 individuals (35 MS patients, amongst which 15 naïve patients and 10 healthy controls, HCs) were cultured with minocycline or doxycycline and conventional stimulants (PMA/Ionomycin or IL-12/IL-18). IFN-γ and IL-17 producing T-, NK- and NKT cells were assessed by flow cytometry. The effect of TCs on cell viability and apoptosis was further assessed by flow cytometry with Annexin V staining. Results Both tetracyclines significantly decreased, in a dose dependent manner, IFN-γ production in NKT and CD4⁺ T lymphocytes from MS patients (naïve or treated) stimulated with IL-12/IL-18 but did not decrease IFN-γ producing CD8⁺ T cells from naive MS or treated RRMS patients. They also decreased IL-17⁺ T and NKT cells following PMA and Ionomycin-stimulation. Tetracyclines did not affect the viability of cell subsets. Conclusion Tetracyclines can in vitro suppress IFN-γ and IL-17- producing cells from MS patients, and this may explain their potential therapeutic effect in vivo.
Article
Full-text available
Objective(s): Prevention of inflammation in early stages will be useful in maintaining vitality of the organism. The objective of this study was to evaluate the effects of doxycycline (DOX) or meloxicam (MLX) monotherapy and combination therapy on the levels of inflammatory mediators in the brain tissues of rats with Escherichia coli lipopolysaccharide (LPS)-induced brain inflammation. Materials and Methods: Seventy-eight rats were divided into the following groups: control (n=6), LPS (0.5 µg/10 µl intracranial) (n=18), LPS (0.5 µg/10 µl intracranial)+DOX (40 mg/kg intraperitoneal) (n=18), LPS (0.5 µg/10 µl intracranial)+MLX (2 mg/kg intraperitoneal) (n=18) and LPS (0.5 µg/10 µl intracranial)+DOX (40 mg/kg intraperitoneal)+MLX (2 mg/kg intraperitoneal) (n=18) groups. Brain tissues were harvested from all rats in the control group and from six rats each in the four experimental groups at 1, 3 and 6 hr under anaesthesia. The levels of tumor necrosis factor α (TNFα), interleukin 4 (IL-4), IL-6, IL-10, IL-17, brain-derived neurotrophic factor (BDNF), matrix metalloproteinase 3 (MMP-3), tissue inhibitor of metalloproteinase 3 (TIMP-3) and cyclooxygenase 2 (COX-2) in the brain tissues were measured using ELISA kits with ELISA device. Results: LPS administration increased proinflammatory cytokines (TNF, IL-6, IL-17), and MMP-3 levels and decreased anti-inflammatory cytokines (IL-10, IL-4), and BDNF levels. The lowest TNFα levels were detected in the LPS+MLX group (P<0.05). All the drug treatment groups showed decreased IL-17 and COX-2 levels compared to the LPS groups. Conclusion: DOX or MLX monotherapy exerts neuroprotective effects against brain inflammation by decreasing proinflammatory cytokine levels and by increasing anti-inflammatory cytokines levels.
Article
Background: Glycolic acid (GA) and salicylic acid (SA) peels have been used separately for acne treatment, not as a sequential peel. Aim: To evaluate the efficacy and safety of sequential peeling with 70% GA and 20% SA as a monotherapy and as an adjuvant to systemic doxycycline in treatment of mild to moderate acne and the effect on serum interleukin (IL) 17 and tissue IL-1α. Patients/methods: Forty-five mild to moderate acne vulgaris patients were randomly assigned into three groups. Group [A] underwent sequential application of 70% GA followed by 20% SA biweekly for three months. Group [B] underwent sequential peeling and doxycycline PO100 mg BD for 1 month followed by 100 OD for 2 months. Group [C] received oral doxycycline. Acne grading, lesion counting, and patient satisfaction were assessed. Serum samples and perilesional skin biopsies were obtained at onset and 2 weeks after finishing the treatment for assessment of serum IL-17 and tissue IL-1α. Results: All groups showed statistically significant decrease in acne grading and lesion count, increase in patient satisfaction, and decrease in serum IL-17 and tissue IL-1 α after treatment. There was no significant difference between the 3 groups before or after treatment, except regarding patient satisfaction after treatment, which was significantly higher in groups [A] and [B] than group [C] (P = .001). Conclusions: This study recommends using sequential GA 70% and SA 20% peels in the treatment of mild or moderate acne vulgaris as a new cost-effective mode, with low-down time and potential safety, in noncompliant patients on medical therapy.
Article
Experimental autoimmune (allergic) neuritis (EAN), which is induced in various animal strains by immunization with peripheral nerve constituents, has been recognized as an animal model of human immune‐mediated neuritis; that is, Guillain–Barré syndrome or chronic inflammatory demyelinating neuritis. At first, EAN was reported in rabbits, and soon thereafter, in mice and rats. Various antigens relating to the peripheral nervous system (PNS) have also been reported. Symptomatic characteristics of EAN are quite similar despite different strains/antigens; that is, acute, self‐limiting, monophasic flaccid paralysis. However, modification of the antigen or administration of immune suppressants to EAN animals are able to induce chronic, relapsing demyelinating neuropathy. The cellular response and humoral kinetics, such as cytokine/chemokine/cell adhesion molecules in PNS of EAN animals, have been extensively studied. In the early 1990s, the dichotomy of type 1 T helper cells and type 2 T helper cells cellular response, along with the cytokine/chemokine milieu in EAN were investigated robustly; thereafter, type 17 T helper cells and regulating T cells followed. This sequence is very similar to what had been observed in human multiple sclerosis and experimental autoimmune encephalomyelitis, an animal model for multiple sclerosis and somewhat counterpart of EAN. However, the PNS and central nervous system have several differences in respect to their cellular components, thus, the immune response that appears in EAN has some unique aspects. Investigation of these differences, and the mechanism of recovery and regeneration of PNS might provide us with better therapeutic options for PNS diseases, especially inflammatory neuropathy. History, overview of phenotype, pathogenesis of experimental autoimmune neuritis was reviewed.
Article
Recent studies have shown that autophagy is involved in peripheral nervous system disease. However, the role of autophagy in the pathogenesis of experimental autoimmune neuritis (EAN) remains unclear. Therefore, EAN was induced by a subcutaneous injection into both hind footpads of synthetic neuritogenic P257-81 peptide in male Lewis rats. The clinical evaluation was completed using a 10-point scale method. The histological alteration of sciatic nerves was analyzed by hematoxylin and eosin and luxol fast blue staining. The ultrastructure of sciatic nerves was analyzed by transmission electron microscopy. Expressions of beclin-1 and microtubule-associated protein light chain-3 (LC3) and p62/SQSTM1 were determined by western blot. 3-Methyladenine, the inhibitor of autophagy, was used in this research. Results showed that the clinical scores were significantly increased from day 6 to day 16 after immunization compared with the control group. Compared with the control group, the number of inflammatory cells and the histological score of sciatic nerves were significantly increased, expressions of beclin-1 and LC3-II and the ratio of LC3-II/LC3-I in the sciatic nerve were significantly increased, and the expression of p62 was significantly decreased in the EAN model group. Considerable double-membrane autophagosomes in axons and myelin sheaths of sciatic nerves were observed and the number of autophagosomes in axons and myelin sheaths of sciatic nerves in the EAN model group was obviously increased compared with the control group. 3-Methyladenine ameliorated the neurologic severity of EAN. Our results suggest that autophagy activity in nerve tissue of EAN rats is increased, which may be associated with the pathogenesis of EAN.
Article
Full-text available
One of the reasons for the insufficient effectiveness of treatment of acute ischemic stroke may be secondary inflammation of the brain tissue, which, according to the results of modern studies, significantly worsens the consequences and outcome of the disease.
Article
Full-text available
Experimental autoimmune neuritis (EAN) is an animal model of Guillain-Barré syndrome. We have shown recently that BB-1101, a broad-spectrum matrix metalloproteinase (MMP) inhibitor, prevents development of EAN when given from the day of immunization and, more important clinically, reduces disease severity when given from symptom onset. This suggests the involvement of MMP activity in the pathogenesis of EAN. However, the exact function and expression patterns of MMPs in acute inflammation of the PNS have not been investigated. MMP-like enzymes are also involved in the processing of tumour necrosis factor-alpha (TNF-alpha), which has been implicated previously in the pathology associated with EAN. In the present study we investigated the profile of MMP and TNF-alpha expression and their localization in sciatic nerve tissue during EAN, using a semiquantitative competitive reverse transcriptase-coupled polymerase chain reaction and immunohistochemistry. In the normal rat PNS, four of the 10 MMPs studied were constitutively expressed and four MMPs were differentially regulated during EAN. Expression of TNF-alpha was elevated at peak disease severity and localized to Schwann cells, macrophages and endoneurial blood vessels. Expression levels of 92 kDa gelatinase and stromelysin-1 were significantly increased early in the development of EAN and continued to rise, peaking at day 15 coincident with maximum disease severity. Schwann cells and endothelial cells were the main cellular source of these enzymes. Prominent infiltration of inflammatory cells into the sciatic nerve was concordant with a significant increase in the expression levels of matrilysin and macrophage metalloelastase. Both matrilysin and macrophage metalloelastase were detected in invading macrophages, T lymphocytes and resident Schwann cells. The selective upregulation of specific MMPs during EAN and their varied cellular localization suggests that MMPs play a multifactorial role in the aetiology of EAN. Activity of MMPs could participate in the disruption of the blood-nerve barrier, breakdown of the myelin sheath, the release of TNF-alpha, and facilitate leukocyte invasion into the PNS. These observations highlight MMPs as potential targets for therapeutic intervention in acute peripheral neuropathies, such as Guillain-Barré syndrome.
Article
Full-text available
The purpose of this study is to make an approach regarding an autoimmune pathology of an ethiology not clearly defined, whose incidence has increased, being characterized by an acute inflammatory process of the peripheral and cranial nerves, leading to the progressive and increasing symmetric debility of the members, and which might also be able to show atypical forms of evolution, called Guillain-Barré Syndrome (GBS). This study contains definitions, history, ethiology, epidemiology, evolution, pathogeny, as well as the involvement of the Campylobacter bacteria with the syndrome, diagnosis, recovery, prognosis and treatment, providing a perspective of what the syndrome consists of and which variants are involved with it. It provides information on this pathology so that the population in general gets to know and understand a little more about the syndrome. Este estudo tem por finalidade discutir uma patologia autoimune de etiologia não claramente definida, sendo tal caracterizada por comprometimento inflamatório agudo dos nervos periféricos e craniais, levando à debilidade simétrica progressiva e ascendente dos membros, podendo também apresentar formas atípicas de evolução. Esta patologia é denominada de Síndrome de Guillain-Barré (SGB) e sua incidência vem aumentando. Este estudo será realizado por meio de levantamento bibliográfico e abrange definições; histórico; etiologia; epidemiologia; evolução; patogenia, bem como o envolvimento de bactéria do gênero Campylobacter com a síndrome; diagnóstico; recuperação; prognóstico e tratamento, dando uma perspectiva do que consiste a síndrome e quais as variantes envolvidas com a mesma, disponibilizando desta forma, informações sobre a referida patologia, para que a população de uma forma geral comece a conhecer e entender um pouco mais a síndrome.
Article
The incidence of neonatal stroke is high and currently there are no strategies to protect the neonatal brain from stroke or reduce the sequelae. Agents capable of modifying inflammatory processes hold promise. We set out to determine whether delayed administration of one such agent, minocycline, protects the immature brain in a model of transient middle cerebral artery (MCA) occlusion in 7-day-old rat pups. Injury volume in minocycline (45mg/kg/dose, beginning at 2 h after MCA occlusion) and vehicle-treated pups was determined 24 h and 7 days after onset of reperfusion. Accumulation of activated microglia/macrophages, phosphorylation of mitogen-activated protein kinase (MAPK) p38 in the brain, and concentrations of inflammatory mediators in plasma and brain were determined at 24 h. Minocycline significantly reduced the volume of injury at 24 h but not 7 days after transient MCA occlusion. The beneficial effect of minocycline acutely after reperfusion was not associated with changed ED1 phenotype, nor was the pattern of MAPK p38 phosphorylation altered. Minocycline reduced accumulation of IL-1 beta and CINC-1 in the systemic circulation but failed to affect the increased levels of IL-1 beta, IL-18, MCP-1 or CINC-1 in the injured brain tissue. Therefore, minocycline provides early but transient protection, which is largely independent of microglial activation or activation of the MAPK p38 pathway.
Article
Experimental autoimmune neuritis (EAN) is an animal model of Guillain-Barré syndrome. We have shown recently that BB-1101, a broad-spectrum matrix metalloproteinase (MMP) inhibitor, prevents development of EAN when given from the day of immunization and, more important clinically, reduces disease severity when given from symptom onset. This suggests the involvement of MMP activity in the pathogenesis of EAN. However, the exact function and expression patterns of MMPs in acute inflammation of the PNS have not been investigated. MMP-like enzymes are also involved in the processing of tumour necrosis factor-alpha (TNF-alpha), which has been implicated previously in the pathology associated with EAN. In the present study we investigated the profile of MMP and TNF-alpha expression and their localization in sciatic nerve tissue during EAN, using a semiquantitative competitive reverse transcriptase-coupled polymerase chain reaction and immunohistochemistry. In the normal rat PNS, four of the 10 MMPs studied were constitutively expressed and four MMPs were differentially regulated during EAN. Expression of TNF-alpha was elevated at peak disease severity and localized to Schwann cells, macrophages and endoneurial blood vessels. Expression levels of 92 kDa gelatinase and stromelysin-1 were significantly increased early in the development of EAN and continued to rise, peaking at day 15 coincident with maximum disease severity. Schwann cells and endothelial cells were the main cellular source of these enzymes. Prominent infiltration of inflammatory cells into the sciatic nerve was concordant with a significant increase in the expression levels of matrilysin and macrophage metalloelastase. Both matrilysin and macrophage metalloelastase were detected in invading macrophages, T lymphocytes and resident Schwann cells. The selective upregulation of specific MMPs during EAN and their varied cellular localization suggests that MMPs play a multifactorial role in the aetiology of EAN. Activity of MMPs could participate in the disruption of the blood-nerve barrier, breakdown of the myelin sheath, the release of TNF-alpha, and facilitate leukocyte invasion into the PNS. These observations highlight MMPs as potential targets for therapeutic intervention in acute peripheral neuropathies, such as Guillain-Barré syndrome.
Article
Multiple sclerosis is characterized by the infiltration of leukocytes into the CNS. As matrix metalloproteinases (MMPs) facilitate the passage of leukocytes across matrix barriers, we tested the hypothesis that targeting MMPs could attenuate neuro‐inflammation. We report that minocycline, a widely used generic drug with a good safety record, inhibited MMP activity, reduced production of MMP‐9 and decreased the transmigration of T lymphocytes across a fibronectin matrix barrier. In addition, minocycline was efficacious against both mild and severe experimental autoimmune encephalomyelitis (EAE) in mice, an animal model of multiple sclerosis. When severe EAE was produced, minocycline pre‐treatment delayed the course of the disease: when maximal disease activity occurred in vehicle‐treated EAE mice, minocycline animals were relatively normal and had minimal signs of inflammation and demyelination in the CNS. When tested in mice afflicted with mild EAE, minocycline attenuated the clinical severity of disease throughout the course of treatment. These results indicate that minocycline may constitute a safe and inexpensive therapy for multiple sclerosis.
Article
There must be something unique about a class of drugs (discovered and developed in the mid-1940s) where there are more than 130 ongoing clinical trials currently listed. Tetracyclines were developed as a result of the screening of soil samples for antibiotic organisms. The first of these compounds chlortetracycline was introduced in 1948. Soon after their development tetracyclines were found to be highly effective against various pathogens including rickettsiae, Gram-positive, and Gram-negative bacteria, thus, becoming a class of broad-spectrum antibiotics. The mechanism of action of tetracyclines is thought to be related to the inhibition of protein synthesis by binding to the 30S bacterial ribosome. Tetracyclines are also an effective anti-malarial drug. Over time, many other "protective" actions have been described for tetracyclines. Minocycline, which can readily cross cell membranes, is known to be a potent anti-apoptotic agent. Its mechanism of action appears to relate to specific effects exerted on apoptosis signaling pathways. Another tetracycline, doxycycline is known to exert antiprotease activities. Doxycycline can inhibit matrix metalloproteinases, which contribute to tissue destruction activities in diseases such as gingivitis. A large body of literature has provided additional evidence for the "beneficial" actions of tetracyclines, including their ability to act as oxygen radical scavengers and anti-inflammatory agents. This increasing volume of published work and ongoing clinical trials supports the notion that a more systematic examination of their possible therapeutic uses is warranted. This review provides a summary of tetracycline's multiple mechanisms of action and while using the effects on the heart as an example, this review also notes their potential to benefit patients suffering from various pathologies such as cancer, Rosacea, and Parkinson's disease.