ArticlePDF Available

Inflammasomes are important mediators of prostatic inflammation associated with BPH

Authors:

Abstract and Figures

Background There is mounting evidence to support the role of inflammation in benign prostate hyperplasia (BPH), and a recent study reported expression of inflammasome derived cytokine IL-18 in prostate biopsy of BPH patients. Here we examined the expression of inflammasome-derived cytokines and activation of nucleotide-binding oligomerization domain-like receptor with pyrin domain protein 1 (NLRP) 1 inflammasome in a rat model of prostatic inflammation relevant to BPH. Methods Prostatic inflammation was experimentally induced in three-month-old male Sprague–Dawley rats by intraprostatic injection (50 μL) of either 5 % formalin or saline (sham) into the ventral lobes of prostate. 7 days later, prostate and bladder tissue was harvested for analysis of inflammasome by Western blot, immunohistochemistry and downstream cytokine production by Milliplex. Results Expression of interleukins, CXC and CC chemokines were elevated 2-15 fold in formalin injected prostate relative to sham. Significant expression of NLRP1 inflammasome components and caspase-1 in prostate were associated with significant elevation of pro and cleaved forms of IL-1β (25.50 ± 1.16 vs 3.05 ± 0.65 pg/mg of protein) and IL-18 (1646.15 ± 182.61 vs 304.67 ± 103.95 pg/mg of protein). Relative to prostate tissue, the cytokine expression in bladder tissue was much lower and did not involve inflammasome activation. Conclusions Significant upregulation of NLRP1, caspase-1 and downstream cytokines (IL-18 and IL-1β) suggests that a NLRP1 inflammasome is assembled and activated in prostate tissue of this rat model. Recapitulation of findings from human BPH specimens suggests that the inflammasome may perpetuate the inflammatory state associated with BPH. Further clarification of these pathways may offer innovative therapeutic targets for BPH-related inflammation.
Content may be subject to copyright.
R E S E A R C H Open Access
Inflammasomes are important mediators of
prostatic inflammation associated with BPH
Mahendra Kashyap, Subrata Pore, Zhou Wang, Jeffrey Gingrich, Naoki Yoshimura and Pradeep Tyagi
*
Abstract
Background: There is mounting evidence to support the role of inflammation in benign prostate hyperplasia (BPH),
and a recent study reported expression of inflammasome derived cytokine IL-18 in prostate biopsy of BPH patients.
Here we examined the expression of inflammasome-derived cytokines and activation of nucleotide-binding
oligomerization domain-like receptor with pyrin domain protein 1 (NLRP) 1 inflammasome in a rat model of prostatic
inflammation relevant to BPH.
Methods: Prostatic inflammation was experimentally induced in three-month-old male SpragueDawley rats by
intraprostatic injection (50 μL) of either 5 % formalin or saline (sham) into the ventral lobes of prostate. 7 days later,
prostate and bladder tissue was harvested for analysis of inflammasome by Western blot, immunohistochemistry
and downstream cytokine production by Milliplex.
Results: Expression of interleukins, CXC and CC chemokines were elevated 2-15 fold in formalin injected prostate relative
to sham. Significant expression of NLRP1 inflammasome components and caspase-1 in prostate were associated
with significant elevation of pro and cleaved forms of IL-1β(25.50 ± 1.16 vs 3.05 ± 0.65 pg/mg of protein) and IL-18
(1646.15 ± 182.61 vs 304.67 ± 103.95 pg/mg of protein). Relative to prostate tissue, the cytokine expression in bladder
tissue was much lower and did not involve inflammasome activation.
Conclusions: Significant upregulation of NLRP1, caspase-1 and downstream cytokines (IL-18 and IL-1β)suggeststhata
NLRP1 inflammasome is assembled and activated in prostate tissue of this rat model.Recapitulation of findings from
human BPH specimens suggests that the inflammasome may perpetuate the inflammatory state associated with BPH.
Further clarification of these pathways may offer innovative therapeutic targets for BPH-related inflammation.
Keywords: Inflammasome, NLRP1, BPH, IL-18, Chemokines
Introduction
The prevalence of benign prostatic hyperplasia (BPH)/
lower urinary tract symptoms (LUTS) in US population
is expected to increase with an ageing population and an
increased prevalence of metabolic diseases [1]. Although
generally thought to be due to prostatic enlargement,
BPH/LUTS is also known to be associated with intra-
prostatic infiltration of inflammatory cells in majority of
patients [2]. The presence of inflammatory infiltrates in
the prostate biopsy predicted unfavorable outcomes in
placebo-treated BPH patients in Medical Therapy Of
Prostatic Symptoms MTOPS study [2].
Findings from large clinical studies suggest that
LUTS, especially storage symptoms such as urgency
and frequency, associated withBPHarenotonlyacon-
sequence of bladder outlet obstruction caused by pros-
tatic enlargement, but there is also a contribution of
prostatic inflammation. A large Olmsted county study
on BPH patients found that daily use of a non-steroidal
anti-inflammatory drug was inversely associated with
onset of moderate/severe urinary symptoms [3]. Several
other epidemiologic studies have also identified obesity
as an important risk factor for BPH associated inflam-
mation and most obesity related disorders are associ-
ated with excessive inflammasome activation [4]. While
the expression of IL-1βand IL-18 were shown to be el-
evated in the prostate biopsy tissue obtained from BPH
patients [5], none of the studies so far has examined
whether this expression in the prostate is associated
with inflammasome activation.
* Correspondence: prt18@pitt.edu
Department of Urology, University of Pittsburgh, Pittsburgh, USA
© 2015 Kashyap et al.; licensee BioMed Central. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain
Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article,
unless otherwise stated.
Kashyap et al. Journal of Inflammation (2015) 12:37
DOI 10.1186/s12950-015-0082-3
Inflammasomes are cytosolic oligomeric signaling plat-
forms found in myeloid cells such as monocytes, macro-
phages and epithelial cells. Inflammasomes are thought to
be responsible for initiating an inflammatory cascade in
response to endogenous or exogenous stress signals [6]. In
particular, NLRP (NLR family containing pyrin domain)
inflammasomes are functional cytosolic homologs of mem-
brane bound toll-like receptors [6] and crucial initiators of
sterile inflammation in several metabolic disorders [7] and
chronic inflammatory diseases [8, 9]. Interestingly, a recent
report demonstrated that expression of mRNA coding for
inflammasome components [10] was higher in prostate
biopsy of BPH patients relative to the expression in prostate
cancer patients. We have previously reported on a rat
model of prostatic inflammation induced by intraprostatic
formalin injection and its relevance for BPH studies [11]. In
this study, we evaluated the expression of different chemo-
kines including the activation of the inflammasome in pros-
tate and bladder tissue following the induction of prostatic
inflammation.
Methods
All animal experiments were performed in accordance
with institutional guidelines and with an approval from
the University of Pittsburgh Institutional Animal Care and
Use Committee (Protocol # 1011435). Male Sprague
Dawley rats weighing 250-320 g were anesthetized with
isoflurane. Following an abdominal incision, formalin (5 %
in saline) or saline (sham) was injected into each ventral
lobe of the prostate (50 μl per lobe) to produce chemically
induced prostatic inflammation. Ventral lobes of the pros-
tate and bladder tissue were excised 7 days after injection.
Histological analyses
A part of the prostate injected with either formalin or
saline (n= 3 per group) was embedded in OCT Tissue-
Tek compound (Sakura Finetek U.S.A, Torrance, CA),
frozen on dry ice, and kept at -80 °C until use. Samples
were serially sectioned at 8 μm thickness and stained
with hematoxylin and eosin.
Immuno-histochemical analysis
8μm cryosections were washed in PBS and fixed in
chilled acetone for 10 min at 4 °C. Sections were then
incubated with PBS containing 0.4 % Triton X-100
(PBST) and 5 % normal donkey serum (Jackson Immu-
noresearch) for 30 min at room temperature. The pri-
mary antibodies specific to NALP1 (1:200, Abcam); and
IL-18 (1:50, Santa Cruz Biotechnologies) were applied
overnight to sections in PBST containing 5 % normal
donkey serum at 4 °C. Sections were then washed 3
times with PBST containing 1.0 % BSA for 5 min at
room temperature and then incubated for 2 h with sec-
ondary donkey anti-primary Alexa Fluor 488 or Alexa
Flour 594 antibody (1: 200, Molecular Probes) at room
temperature. Sections were washed again for three times
at room temperature in PBST containing 1.0 % serum,
and then mounted with medium containing 4,6-diami-
dino-2-phenylindole DAPI (Floromount-G with DAPI,
Fischer Scientific). Sections were visualized under Olympus
BX51 microscope and the images were captured using
MagnaFire 2.1 software.
Measurement of chemokines
Theprostateandbladdertissuesofratsinjectedwithfor-
malin or saline (n= 6 per group) were homogenized using
cold CelLyticMT Mammalian Tissue Lysis/Extraction
Reagent (sigma) containing 2 mM sodium orthovanadate,
1 mM PMSF and protein cocktail inhibitor (1X, Sigma).
The homogenate was centrifuged at 10,000 rpm for 10 min
and the resulting supernatants were stored at -80 °C until
assayed. 28 proteins including interleukins IL-1α,IL-1β,
IL-2, IL-4, IL-5, IL-6, IL-10, IL-12p70, IL-13, IL-17A and
IL-18; CXC chemokines (CXCL1, CXCL2, CXCL5 and
CXCL10), CC chemokines (CCL2, CCL3, CCL5); Growth
factors NGF, BDNF, VEGF and G-CSF, other inflammatory
mediators such as eotaxin, leptin, IFN-γand TNFαlevels
were determined on a Luminex 100 using a MILLIPLEX
MAP Rat Cytokine/Chemokine Panel (Millipore, Billerica,
MA). Levels of NGF and BDNF were determined using in-
dividual ELISA kits procured from Promega, USA. Protein
estimation was done by BCA Protein Assay Kit (Pierce,
Rockford, Illinois) to standardize the chemokine concentra-
tions relative to tissue protein levels, which are expressed as
pg/mg of total protein [12].
Western blot analysis
Tissue was homogenized using CelLyticMT Mammalian
Tissue Lysis/Extraction Reagent (Catalog no. C3228, Sigma,
USA) in the presence of phenylmethylsulfonyl fluoride
(1 mM), sodium orthovanadate(2mM)andproteininhibi-
tor cocktail (Catalog no. P8340-5ML, Sigma, USA). Protein
estimation was done by Coomassie (Bradford) Protein
Assay Kit (Thermo Scientific, USA). The lysates from the
sham and formalin injected groups were separated on
Tricine-SDS PAGE. Protein lysates (40 μg) were electro-
phoresed using 10 % Tricine-SDS Gel and then blotted
onto immobilion-P membranes (Millipore) using wet
transfer system. After blocking for 1 h at 37 °C, the
membranes were incubated overnight at 4 °C with pri-
mary antibodies specific for NALP1 (1:500, Abcam),
Caspase-1 (1; 500), IL1β(1:400) and β-Actin (1:1000, Santa
Cruz Biotechnology), in blocking buffer (pH 7.5). The
membranes were then re-incubated for 2 h at room
temperature with secondary anti-primary immunoglobulin
G (IgG)-conjugated with horseradish peroxidase (Santa
Cruz Biotechnologies, USA). Subsequently, blots were
developed using SuperSignal West Femto Maximum
Kashyap et al. Journal of Inflammation (2015) 12:37 Page 2 of 8
Sensitivity Substrate (catalog no. 34096, Thermo Scien-
tific, USA) on Versa doc imaging system (Model 4000;
BioRed, USA). Densitometry for measuring the band
specific for each protein was performed using AlphaEase
FC StandAlone V. 4.0.0 software. β-Actin was used as an
internal control to normalize the data.
Statistics
The unpaired Studentst-test was used for comparing the
values of two groups. All tests were two-sided, and p values
< 0.05 were considered statistically significant. All statistical
analyses were performed using Graphpad Prism IV.
Results
Chemokine expression in prostate & bladder
Expression of chemokines was elevated several fold in
formalin injected prostate tissue relative to the expres-
sion of respective chemokines in saline injected prostate
tissue (sham). All the chemokines belonging to CXC fam-
ily were significantly upregulated in formalin injected
prostate, with the maximum upregulation of 15 fold in
mean levels of CXCL-1 followed by 5 fold upregulation of
both CXCL-5 and CXCL-10 (Fig. 1a). Among CC chemo-
kines, CCL3 showed a 9 fold upregulation, followed by a 5
fold upregulation for CCL5 and then 2-fold upregulation
a
b
Fig. 1 aCytokine expression was elevated in prostate tissue harvested from prostatic inflammation group relative to sham. Relative to other cytokines,
expression of IL-18 was quantitatively higher and also significantly relative to the expression of IL-18 in sham prostate. There was significant upregulation of
IL-1α,IL-1β, IL-5, IL-17A, leptin, NGF, VEGF, CXC and CC chemokines (*p < 0.05, unpaired ttest) in formalin injected prostate, while expression of BDNF, IFNγ,
G-CSF, IL-2, IL-4, IL-10, IL-12p70, IL-13, TNFαremained unchanged between the groups. bOverall, expression of cytokines in bladder was lower relative to
prostate (same y-axis scale). Bladder from sham group showed significantly lower expression of IFNγ, CXCL-2, CXCL-10, CCL5, IL-5 and IL-17A compared to
the group injected with formalin (*p <0.05, unpaired ttest)
Kashyap et al. Journal of Inflammation (2015) 12:37 Page 3 of 8
for CCL2 (*p < 0.05, unpaired ttest). IL-18 expression was
quantitatively higher relative to other cytokines/chemokines
and the elevation of IL-8 in formalin injected prostate tissue
was also significant relative to sham prostate tissue.
Among other interleukins, IL-1α, IL-1β, IL-5, and
IL-17A were also significantly upregulated in formalin
prostate, with the highest fold change noted for IL-1β.
Among growth factors, prostatic inflammation induced a
significant 3 fold upregulation of VEGF, NGF and two
fold upregulation of leptin expression. The expression of
BDNF, interferon-γ(IFNγ), G-CSF, IL-2, IL-4, IL-10,
IL-12p70, IL-13, TNFαremained unchanged between
the sham and formalin injected groups, whereas the
expression of eotaxin was undetectable in prostate and
bladder tissue of both groups.
Overall, bladder tissue expression of cytokines was
lower relative to prostate of each group. Bladder tissue
obtained from rat group given intraprostatic injection of
formalin showed significantly higher expression of IFNγ,
CXCL-2, CXCL-10, CCL5, IL-5 and IL-17A (*p < 0.05,
unpaired ttest). (Fig. 1b). The expression of all other
proteins was not statistically different in the bladder
tissue of the two groups.
Western blot of inflammasome components
Western blots of prostate tissue lysate showed signifi-
cantly stronger density bands for NLRP1 (0.92 ± 0.02 vs
0.45 ± 0.01; *p < 0.05), Caspase-1 (0.97 ± 0.08 vs 0.42 ±
0.03;*p < 0.05) and mature IL-1β(0.40 ± 0.11 vs 0.08 ±
0.01;*p < 0.05) in rat group given intraprostatic injection
of formalin relative to sham prostate (Fig. 2). In contrast,
Western blot results for bladder showed absence of any
change in expression of NLRP1 and other components
following intraprostatic injection of formalin or saline.
Immuno-histochemistry of inflammasome components
Prostate from the formalin injected group showed higher
green immunoreactivity for NLRP1 and red immunore-
activity for IL-18 protein relative to the sham group
injected with the saline (Fig. 3a-f ). The yellow signal
seen in the merged image of panel F indicated the co-
localized expression of NLRP1 with IL-18 against the
blue DAPI background in the formalin injected prostate
tissue. The merged image for the sham prostate tissue in
panel E shows absence of the yellow signal, which con-
firms the lowered expression of NLRP1 and IL-18.
In contrast, there was no difference in the green immu-
noreactivity for NLRP1 and red immunoreactivity for
IL-18 in the bladder obtained from sham (Fig. 3g, i and k)
and formalin injected groups (Fig. 3h, j and l). The anti-
body for IL-18 performed better in immunohistochemistry
than in Western blot (data not shown). Therefore, a role
for inflammasome dependent cytokine expression is indi-
cated in prostate tissue, but not in bladder tissue.
Histology
Prostate tissue obtained from the sham group showed
regular shaped acini with an intact basement membrane
(Fig. 4a and c). In contrast, formalin injected prostate
tissue showed hyperplastic acini lined by tall columnar
epithelium. Epithelial pilling with budding into surround-
ing expanded stromal areas was seen in multiple foci. Infil-
tration of inflammatory cells in the formalin injected
prostate is indicated by the red color * (Fig. 4b and d).
Bladder tissue from the sham group (Fig. 4e) showed
histology similar to a normal, untreated rat. In contrast,
bladder tissue from (Fig. 4f) the formalin injected group
showed mild edematous changes, which were not accom-
panied by any marked infiltration of inflammatory cells.
Prostate
Sham Prostatic
Inflammation
Bladder
NLRP1, 155 KDa
CASP1, 55 KDa
Sham Prostatic
Inflammation
Fig. 2 Western blot analysis for components of inflammasome in prostate and bladder tissue obtained from sham and prostatic inflammation
group. Blots represent the activity of NLRP1, Caspase-1, parent and mature cleaved IL-1βin both tissues
Kashyap et al. Journal of Inflammation (2015) 12:37 Page 4 of 8
Discussion
Our previous study reported that clinical features, bio-
chemical and histological changes in formalin induced
non-infectious prostatic inflammation are highly similar
to those reported for clinical BPH [11]. In the present
study, we assessed the functional significance of inflam-
masome activation in the tissue specific expression of
cytokines/chemokine in the prostate and bladder tissues
of the same model. We observed that intraprostatic for-
malin injection leads to the assembly and activation of
NLRP1 inflammasomes in prostate and production of
pro-inflammatory cytokines, IL-1βand IL-18 following
the auto-proteolytic maturation of cysteine protease,
caspase-1. Meanwhile, the absence of inflammasome
derived products in bladder suggests that chemokine
expression in the bladder is induced by a stimulus,
which is different from the prostate.
A role for NLRP1 inflammasome in prostate tissue has
been previously demonstrated in another model of prostatic
inflammation [13] induced by intraprostatic injection of
carrageenan. Consideration of our findings, together with
other similar reports, led us to propose that Nod-like recep-
tor protein 1 (NALP1/NLRP1) inflammasomes mediate the
prostatic inflammation in response to irritable stimuli such
as formalin or carrageenan. Interestingly, detailed metabolic
and molecular phenotyping in clinical studies have indi-
cated that, inflammasome is a crucial link between BPH
and metabolic disorders [10], since the inflammasome
controls the energy expenditure and adipogenic gene ex-
pression [7] including that of adipocyte hormone leptin
[14]. Enhanced leptin production noted in prostate after
intraprostatic formalin injection mimics the likely endo-
crine influence from obesity in prostatic inflammation.
Visceral adiposity is correlated with circulating levels
of pro-inflammatory cytokines, and adipose tissue is
known to propagate inflammation locally and systemic-
ally, in part through chemokine mediated recruitment
of macrophages. Expectedly, chemokines chemotactic
for macrophages such as CCL2 were found elevated in
prostatic fluids of BPH patients [15]. The upregulated
expression of other CC chemokines, CCL2, CCL3 and
CCL5 noted in formalin injected prostate is consist-
ent with the prostatic infiltration of macrophages
[16, 14]. Macrophages not only release CCL3 but,
also serve as a site for inflammasome activation and
are also known to positively regulate gene expression
abgh
cd i j
efkl
Fig. 3 a-f Detection of NLRP1 inflammasomes (green) and derived cytokine IL-18 (red) in the prostate by immunohistochemistry in prostate of
sham (panel a, c and e) and formalin injected group (panel b, d and f). Merged image in panel F shows the co-localized expression of NLRP1
with IL-8 against the DAPI blue background in formalin injected prostate and its marked absence in sham prostate. g-l Detection of NLRP1
inflammasomes (green) and derived cytokine IL-18 (red) by immunohistochemistry in bladder of sham (panel g, i and k) and formalin injected group
(panel h, j and l). Merged image shows the constitutive expression of IL-18 against the DAPI blue background, but the absence of NLRP1 in bladder
Kashyap et al. Journal of Inflammation (2015) 12:37 Page 5 of 8
of CCL5 via NF-κB signaling cascades induced by IFN-γ
[16, 14].
Inflamed regions in prostate biopsy of BPH patients
are known to express upto 5-fold higher IFN-γcom-
pared to the non-inflamed regions [17]. Here, we found
that IFN-γinducible chemokines [18] were upregulated
to different extents in formalin-injected prostate tissue.
CXC chemokines including CXCL1, CXCL2, CXCL5, are
potent chemoattractants for neutrophils and can further
drive the neutrophil dependent tissue injury [8] visual-
ized on the histology of formalin injected prostate tissue.
CXCL2 induces migration of hematopoietic stem cells
and its expression is affected by FGF-2, which is impli-
cated in BPH [19]. CXCL-10 is chemotactic for lympho-
cytes and is structurally and functionally different from
CXCL-1, a rat homolog of human IL-8 [20].
The dysfunctional voiding reported in rats after intra-
prostatic formalin injection [21] is presumably linked
to the predominant expression of IFN-γand the IFN-γ
inducible expression of chemokine, CXCL-10 in bladder.
CXCL-10 is considered to be constitutivelyexpressedin
neurons and contribute to the excitability of primary
afferent neurons through transactivation of transient
receptor channels and nociceptor sensitization [22].
Clinical relevance of CXCL-10 in bladder function and
urinary symptoms can be gleaned from the upregulated
gene expression of CXCL-10 in bladder biopsy of
ulcerative cystitis patients [23]. Interestingly, a previous
report showed that systemic neutralization of CXCL-10
by monoclonal antibodies ameliorated the dysfunctional
voiding following chemically induced cystitis [24].
Increased expression of CXCL-10 in bladder tissue of
formalin injected rats accompanied a modest increase
in the expression of CXCL-2, CCL5, IL-5 and IL-17A,
which suggests a role for T-helper17 lymphocytes and
epithelium in the observed bladder expression of che-
mokines [17]. Ubiquitous expression of IL-18 by most
epithelial tissues [25] may explain the substantial IL-18
Fig. 4 a-d Prostate tissue sections from sham group (panel aand c) showed regular shaped acini with an intact basement membrane. Prostatic
inflammation in formalin injected group (panel band d) caused epithelial pilling with budding into surrounding stroma at multiple foci.
Expanded stromal area in formalin injected group showed inflammatory cells indicated by red colored * (panel band d). Magnification in panel
aand bis 10X and the region shown in white dotted line square region of panel a (sham) & b (prostatic inflammation) is magnified upto 4 fold
and shown in panel c & d, respectively. e-f Bladder tissue sections obtained from sham group (panel e) showed no histological changes, whereas
bladder from rat group with prostatic inflammation (panel f) showed slight edematous changes related to cytokine expression shown in Fig. 1b.
Magnification is 20X in both panels
Kashyap et al. Journal of Inflammation (2015) 12:37 Page 6 of 8
expression in bladder of both saline and formalin
injected group.
Over expression of CC and CXC chemokines in prostate
of formalin injected rats corroborates similar findings
obtained in another rat model of BPH induced by chronic
estradiol injection [26]. An earlier study mixed a colored
dye with formalin during intraprostatic injection in order to
check the spread of formalin outside of prostate tissue by
the external spread of dye [21]. The study did not find any
spread of the injected dye outside of the prostate, which
precludes a possible direct irritation of bladder by formalin
in this model. A severe inflammatory response typically
seen with direct irritation of bladder by formalin [27] was
also not seen in bladder histology images (Fig. 4f). Taking
histology images of bladder together with NLRP1 blot and
immunoreactivity indicates that inflammasome is not
assembled in bladder tissue following intraprostatic
injection of formalin. It is known that expression of
NLRP3 is higher than expression of NLRP1 in bladder
[6], but they both generate similar inflammasome
derived products. Therefore, lack of any change in the
expression of cleaved IL-1βor IL-18 in bladder tissue,
lend further support to the absence of the inflamma-
some in the bladder of this model.
NLRP1 are known to respond to endogenous meta-
bolic stress (ATP and fatty acids) [7], and to exogenous
stress of microbial infection [6], but the precise stimulus
triggering the activation of NLRP1 in human prostate
remains to be investigated. Recent studies on tissue
specimens of BPH patients have implicated a pivotal
functional role for IL-18 in BPH [5]. Predominant
expression of IL-18 in rat prostate of this model is sup-
ported by Western blot and immunoreactivity findings.
The autocrine/paracrine actions of IL-1βsecreted by
the inflammasome are implicated by the overexpression
of CC and CXC chemokines [28] in this model, as these
mediators do not require inflammasome processing for
secretion.
CC and CXC chemokines represent a large family of
chemotactic peptides with a broad range of cellular targets
generated by stromal and epithelial tissues of prostate and
bladder. Leukocyte infiltration is the primary event in in-
flammation and expression of chemokines temporally pre-
cedes that infiltration, [29] which makes them well suited
for characterizing disease phenotypes [30]. Here, we found
organ-specific prominent molecular signatures of prostatic
inflammation. Considering that NALP1/NLRP1 is a
susceptibility gene involved in the devolvement of chronic
inflammatory diseases [9], this model can be used to
understand cellular triggers of inflammasome activation in
BPH. Taken together, it is clear that the inflammasomes
plays a role in prostatic inflammation associated with BPH
and inflammasome targeted therapies could be an option
for BPH management.
Conclusions
Significant upregulation of NLRP1, caspase and down-
stream cytokines suggest that a NLRP1 inflammasome is
assembled and activated in the prostate tissue of this rat
model.Recapitulation of findings from human BPH
specimens suggests that the inflammasome may per-
petuate the inflammatory state associated with BPH and
further clarification of the pathways may offer innovative
therapeutic targets in BPH.
Abbreviations
NLRP1: Nucleotide-binding oligomerization domain-like receptor with pyrin
domain protein 1; BPH: Benign prostatic hyperplasia; IL: Interleukins;
MTOPS: Medical therapy of prostatic symptoms; LUTS: Lower urinary tract
symptoms; NGF: Nerve growth factor; BDNF: Brain derived neurotrophic
factor; VEGF: Vascular endothelial growth factor; G-CSF: Granulocyte
stimulating factor; IFN-γ: Interferon-γ.
Competing interests
The authors declare that they have no competing interests.
Authorscontributions
MK, SP, PT drafted the manuscript. ZW, JG, NY, PT conceived of the study,
and participated in its design and coordination and helped to draft the
manuscript. MK, SP, PT performed the animal experiments, immunoassays,
blots and staining. All authors read and approved the final manuscript.
Acknowledgement
The work was partly supported by grants from NIH NIDDK 2P20 DK090919
and DK088836.
Received: 30 March 2015 Accepted: 8 May 2015
References
1. Wei JT, Calhoun E, Jacobsen SJ. Urologic diseases in america project: benign
prostatic hyperplasia. J Urol. 2008;179(5 Suppl):S7580. doi:10.1016/
j.juro.2008.03.141.
2. Roehrborn CG. BPH progression: concept and key learning from MTOPS,
ALTESS, COMBAT, and ALF-ONE. BJU Int. 2008;101 Suppl 3:1721.
doi:10.1111/j.1464-410X.2008.07497.x.
3. St Sauver JL, Jacobson DJ, McGree ME, Lieber MM, Jacobsen SJ. Protective
association between nonsteroidal antiinflammatory drug use and measures
of benign prostatic hyperplasia. Am J Epidemiol. 2006;164(8):7608.
doi:10.1093/aje/kwj258.
4. Weber K, Schilling JD. Lysosomes integrate metabolic-inflammatory cross-talk
in primary macrophage inflammasome activation. J Biol Chem.
2014;289(13):915871. doi:10.1074/jbc.M113.531202.
5. Hamakawa T, Sasaki S, Shibata Y, Imura M, Kubota Y, Kojima Y, et al.
Interleukin-18 may lead to benign prostatic hyperplasia via
thrombospondin-1 production in prostatic smooth muscle cells. Prostate.
2014;74(6):590601. doi:10.1002/pros.22773.
6. Kummer JA, Broekhuizen R, Everett H, Agostini L, Kuijk L, Martinon F, et al.
Inflammasome components NALP 1 and 3 show distinct but separate
expression profiles in human tissues suggesting a site-specific role in the
inflammatory response. J Histochem Cytochem. 2007;55(5):44352.
doi:10.1369/jhc.6A7101.2006.
7. Rossi C, Santini E, Chiarugi M, Salvati A, Comassi M, Vitolo E, et al. The
complex P2X7 receptor/inflammasome in perivascular fat tissue of heavy
smokers. Eur J Clin Invest. 2014;44(3):295302. doi:10.1111/eci.12232.
8. van Golen RF, van Gulik TM, Heger M. The sterile immune response during
hepatic ischemia/reperfusion. Cytokine Growth Factor Rev. 2012;23(3):6984.
doi:10.1016/j.cytogfr.2012.04.006.
9. Pontillo A, Catamo E, Arosio B, Mari D, Crovella S. NALP1/NLRP1 genetic
variants are associated with Alzheimer disease. Alzheimer Dis Assoc Disord.
2012;26(3):27781. doi:10.1097/WAD.0b013e318231a8ac.
10. Ponomareva L, Liu H, Duan X, Dickerson E, Shen H, Panchanathan R, et al. AIM2,
an IFN-inducible cytosolic DNA sensor, in the development of benign prostate
Kashyap et al. Journal of Inflammation (2015) 12:37 Page 7 of 8
hyperplasia and prostate cancer. Mol Cancer Res. 2013;11(10):1193202.
doi:10.1158/1541-7786.MCR-13-0145.
11. Funahashi Y, O'Malley KJ, Kawamorita N, Tyagi P, DeFranco DB, Takahashi R,
et al. Upregulation of androgen-responsive genes and transforming growth
factor-beta1 cascade genes in a rat model of non-bacterial prostatic
inflammation. Prostate. 2014;74(4):33745. doi:10.1002/pros.22668.
12. Smaldone MC, Vodovotz Y, Tyagi V, Barclay D, Philips BJ, Yoshimura N, et al.
Multiplex analysis of urinary cytokine levels in rat model of cyclophosphamide-
induced cystitis. Urology. 2009;73(2):4216. doi:10.1016/j.urology.2008.07.031.
13. Chen CS, Chang PJ, Lin WY, Huang YC, Ho DR. Evidences of the inflammasome
pathway in chronic prostatitis and chronic pelvic pain syndrome in an animal
model. Prostate. 2013;73(4):3917. doi:10.1002/pros.22580.
14. Stienstra R, van Diepen JA, Tack CJ, Zaki MH, van de Veerdonk FL, Perera D,
et al. Inflammasome is a central player in the induction of obesity and
insulin resistance. Proc Natl Acad Sci U S A. 2011;108(37):153249.
doi:10.1073/pnas.1100255108.
15. Fujita K, Ewing CM, Getzenberg RH, Parsons JK, Isaacs WB, Pavlovich CP.
Monocyte chemotactic protein-1 (MCP-1/CCL2) is associated with prostatic
growth dysregulation and benign prostatic hyperplasia. Prostate.
2010;70(5):47381. doi:10.1002/pros.21081.
16. Berube J, Bourdon C, Yao Y, Rousseau S. Distinct intracellular signaling
pathways control the synthesis of IL-8 and RANTES in TLR1/TLR2, TLR3 or
NOD1 activated human airway epithelial cells. Cell Signal. 2009;21(3):44856.
doi:10.1016/j.cellsig.2008.12.001.
17. Steiner GE, Stix U, Handisurya A, Willheim M, Haitel A, Reithmayr F, et al.
Cytokine expression pattern in benign prostatic hyperplasia infiltrating T
cells and impact of lymphocytic infiltration on cytokine mRNA profile in
prostatic tissue. Lab Invest. 2003;83(8):113146.
18. Kramer G, Steiner GE, Handisurya A, Stix U, Haitel A, Knerer B, et al. Increased
expression of lymphocyte-derived cytokines in benign hyperplastic prostate
tissue, identification of the producing cell types, and effect of differentially
expressed cytokines on stromal cell proliferation. Prostate. 2002;52(1):4358.
doi:10.1002/pros.10084.
19. Yoon KA, Cho HS, Shin HI, Cho JY. Differential regulation of CXCL5 by FGF2
in osteoblastic and endothelial niche cells supports hematopoietic stem cell
migration. Stem Cells Dev. 2012;21(18):3391402. doi:10.1089/scd.2012.0128.
20. Antoniou KM, Tzanakis N, Tzortzaki EG, Malagari K, Koutsopoulos AV, Alexandrakis
M, et al. Different angiogenic CXC chemokine levels in bronchoalveolar lavage
fluid after interferon gamma-1b therapy in idiopathic pulmonary fibrosis patients.
Pulm Pharmacol Ther. 2008;21(6):8404. doi:10.1016/j.pupt.2008.06.005.
21. Vera PL, Meyer-Siegler KL. Inflammation of the rat prostate evokes release of
macrophage migration inhibitory factor in the bladder: evidence for a
viscerovisceral reflex. J Urol. 2004;172(6 Pt 1):24405.
22. Bhangoo S, Ren D, Miller RJ, Henry KJ, Lineswala J, Hamdouchi C, et al.
Delayed functional expression of neuronal chemokine receptors following
focal nerve demyelination in the rat: a mechanism for the development of
chronic sensitization of peripheral nociceptors. Mol Pain. 2007;3:3858.
doi:10.1186/1744-8069-3-38.
23. Ogawa T, Homma T, Igawa Y, Seki S, Ishizuka O, Imamura T, et al. CXCR3
binding chemokine and TNFSF14 over expression in bladder urothelium of
patients with ulcerative interstitial cystitis. J Urol. 2010;183(3):120612.
doi:10.1016/j.juro.2009.11.007.
24. Sakthivel SK, Singh UP, Singh S, Taub DD, Novakovic KR, Lillard Jr JW.
CXCL10 blockade protects mice from cyclophosphamide-induced cystitis.
J Immune Based Ther Vaccines. 2008;6:620. doi:10.1186/1476-8518-6-6.
25. Knodler LA, Crowley SM, Sham HP, Yang H, Wrande M, Ma C, et al.
Noncanonical inflammasome activation of caspase-4/caspase-11 mediates
epithelial defenses against enteric bacterial pathogens. Cell Host Microbe.
2014;16(2):24956. doi:10.1016/j.chom.2014.07.002.
26. Sugimoto M, Oka M, Tsunemori H, Yamashita M, Kakehi Y. Effect of a
phytotherapeutic agent, Eviprostat(R), on prostatic and urinary cytokines/
chemokines in a rat model of nonbacterial prostatitis. Prostate.
2011;71(4):43844. doi:10.1002/pros.21299.
27. Dupont MC, Spitsbergen JM, Kim KB, Tuttle JB, Steers WD. Histological and
neurotrophic changes triggered by varying models of bladder
inflammation. J Urol. 2001;166(3):11118.
28. Kogan-Sakin I, Cohen M, Paland N, Madar S, Solomon H, Molchadsky A,
et al. Prostate stromal cells produce CXCL-1, CXCL-2, CXCL-3 and IL-8 in
response to epithelia-secreted IL-1. Carcinogenesis. 2009;30(4):698705.
doi:10.1093/carcin/bgp043.
29. Perez de Lema G, Maier H, Nieto E, Vielhauer V, Luckow B, Mampaso F, et al.
Chemokine expression precedes inflammatory cell infiltration and
chemokine receptor and cytokine expression during the initiation of murine
lupus nephritis. J Am Soc Nephrol. 2001;12(7):136982.
30. Tyagi P, Killinger K, Tyagi V, Nirmal J, Chancellor M, Peters KM. Urinary
chemokines as noninvasive predictors of ulcerative interstitial cystitis. J Urol.
2012;187(6):22438. doi:10.1016/j.juro.2012.01.034.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Kashyap et al. Journal of Inflammation (2015) 12:37 Page 8 of 8
... Once stimulated, the N-terminal PYD domain of NLRP1 binds to ASC, initiating a cascade reaction and participating in numerous inflammatory diseases by regulating the innate and adaptive immune responses (30). In a previous study of prostatitis and prostatic hyperplasia in rats, Kashyap et al (31) reported that assembly and activation of NLRP1 in the prostate promoted the production of proinflammatory cytokines IL-1β and IL-18 after the autoproteolysis of caspase-1 and maturation. Glinskii et al (32) reported that the expression of the NLRP1 inflammasome is involved in the occurrence of prostate tumors, and NLRP1 expression is enhanced in highly metastatic prostate cancer cells in prostate cancer. ...
... T he N L RC4 i n f la m ma some, a lso k now n a s ICE-protease-activating factor, belongs to the NLR protein family (31). NLRC4 is comprised of CARD, neuronal apoptosis inhibitory protein (NAIP), MHC class II transcription activator, incompatibility locus protein from Podospora anserina and telomerase-associated protein and LRR domains (31). ...
... T he N L RC4 i n f la m ma some, a lso k now n a s ICE-protease-activating factor, belongs to the NLR protein family (31). NLRC4 is comprised of CARD, neuronal apoptosis inhibitory protein (NAIP), MHC class II transcription activator, incompatibility locus protein from Podospora anserina and telomerase-associated protein and LRR domains (31). NLRC4 can directly bind to pro-caspase-1 through CARD-CARD interactions, which triggers caspase-1 processing and activation (33). ...
Article
The present study explored the clinical value of the protein expression levels of nucleotide binding oligomerization-like receptor family pyrin domain containing 1 (NLRP1) and nucleotide-binding oligomerization domain leucine-rich repeat and caspase recruitment domain-containing 4 (NLRC4) inflammasomes in the diagnosis and treatment of prostate cancer. A total of 54 patients with prostatic hyperplasia and 58 patients with prostate cancer were recruited at The First People's Hospital of Pinghu between January and May 2022. Immunohistochemical staining was used to determine the protein expression levels of the NLRP1 and NLRC4 inflammasomes in addition to the proinflammatory cytokines IL-18 and IL-1β in the two groups of patients. The protein expression levels of NLRP1 and NLRC4 inflammasome were significantly increased in patients with prostate cancer compared with patients with prostate hyperplasia. The differences in expression of NLRP1 and NLRC4 inflammatory vesicles in prostate cancer of different stages were also compared based on data from The Cancer Genome Atlas. The protein expression level of NLRP1 demonstrated a significant positive correlation with IL-1β and IL-18 expression, and the protein expression level of the NLRC4 inflammasome was significantly positively correlated with IL-18 expression. The protein expression levels of both NLRP1 and NLRC4 demonstrated a significant positive correlation with the Gleason score of prostate cancer. The expression of NLRP1 in tumor (T)3/T4 was significantly higher compared with T1 and expression of the NLRC4 inflammasome in T2 and T3/T4 was significantly higher compared with T1. Expression of the NLRP1 and NLRC4 inflammasomes was significantly higher in patients with prostate cancer, compared with patients with prostatic hyperplasia. Therefore, expression of NLRP1 and NLRC4 may promote tumorigenesis by promoting the maturation and release of proinflammatory cytokines IL-1β and IL-18. Expression of the NLRP1 and NLRC4 inflammasomes demonstrated a significant positive correlation with the risk of prostate cancer. Expression of the NLRP1 and NLRC4 inflammasomes in middle- and advanced-stage tumors was higher compared with early-stage tumors. These results suggested that inflammasome expression may serve a significant role in the progression of tumors and could provide a fixed value for the risk assessment and prognosis prediction of prostate cancer.
... , also called gamma interferon induction factor (IFN-γ), is a pleiotropic cytokine belonging to the interleukin 1 family [18]. The formation of an active and mature form of IL-18 depends on caspase-1 resulting from the assembly of protein complexesinflammasomes, which are crucial for the initiation of sterile inflammation in metabolic disorders and chronic inflammatory diseases [19]. Interleukin 18 is secreted from monocytes/ macrophages, however more than 80% of the precursor form of IL-18 remains in this form inside the cell. ...
... The role of the inflammasome and the secretion of IL-18 with it was first confirmed in studies conducted in rats. It has been shown that prostate inflammation in the course of BPH is associated with the activation of the inflammasome, and through the secretion of pro-inflammatory interleukins, it is involved in maintaining the inflammation of the prostate gland [19]. ...
Article
Recent studies indicate that inflammation is one of the causes of the development of benign prostatic hyperplasia (BPH). Inflammation may result from past infections, metabolic disorders, but also from the state of functioning of the intestinal microbiota. The aim of the study was to assess whether the diagnostic lipid parameters for metabolic syndrome and short-chain fatty acids (SCFAs) are related to the immunoexpression of interleukins in prostate tissue with benign hyperplasia. The study involved 103 men with BPH, who were divided into two groups depending on the presence of MetS. We analysed tissue immunoexpression of two proinflammatory interleukins: IL-6, which is known to be involved in the development of BPH, and IL-18, which has not been analysed so far. The results of our study indicate that men with BPH + MetS in the stroma of the prostate have a significantly higher overall percentage of IL-6+ cells compared to men without MetS (p = 0.034). The analysis of IL-18 immunoexpression in prostate tissue indicated that in men with BPH + MetS, the glandular part of the prostate had a significantly higher percentage of cells with strong IL-18 expression (p = 0.040). We also noticed a relationship between tissue expression of IL-6 and IL-18 and lipid parameters (TG and HDL). We conclude that lipid disorders occurring in men with BPH increase inflammation in the prostate gland. Moreover, it has also been demonstrated for the first time that, indirectly, through SCFAs, the gut microbiota can act to prevent or create an inflammatory microenvironment in the prostate gland.
... A further interesting result that emerged from our immunohistochemical results is the evidence of a significant downregulation of the NLRP3 inflammasome belonging to the innate immune system and its downstream pathway, caspase-1, NFkB, and IL-1β in the prostates of CR rats compared with ND-rats. Different animal models of prostatic inflammation and BHP reported a significant upregulation of NLRP3, caspase, and downstream cytokines, IL-1β and IL-18, suggesting that inflammasomes may perpetuate the inflammatory state associated with BPH [45][46][47]. Furthermore, inflammasomes, by triggering inflammatory cascades, promote the formation of an inflammatory microenvironment that is conducive to tumor cell growth [48,49]. In support of this, the molecular mechanism by which the overactivation of the NLRP3 inflammasome promotes the malignant progression of prostate cancer has been defined, proposing this inflammasome as a new prognostic biomarker and potential therapeutic target for this cancer [22]. ...
Article
Full-text available
Numerous animal models have demonstrated that caloric restriction (CR) is an excellent tool to delay aging and increase the quality of life, likely because it counteracts age-induced oxidative stress and inflammation. The aging process can affect the prostate in three ways: the onset of benign prostatic hyperplasia, prostatitis, and prostate cancer. In this study, we used 14 aged male Sprague Dawley rats, which were allocated into two groups, at the age of 18 months old. One group was fed ad libitum (a normal diet (ND)), and the other group followed a caloric restriction diet with a 60% decrease in intake. The rats were sacrificed at the age of 24 months. By immunohistochemical (IHC) and Western blot (WB) analyses, we studied the variations between the two groups in immune inflammation and fibrosis-related markers in aged prostate tissues. Morphological examinations showed lower levels of prostatic hyperplasia and fibrosis in the CR rats vs. the ND rats. The IHC results revealed that the prostates of the CR rats exhibited a lower immune proinflammatory infiltrate level and a reduced expression of the NLRP3 inflammasome pathway, together with significantly reduced expressions of mesenchymal markers and the profibrotic factor TGFβ1. Finally, by WB analysis, we observed a reduced expression of ERα, which is notoriously implicated in prostate stromal proliferation, and increased expressions of SOD1 and Hsp70, both exerting protective effects against oxidative stress. Overall, these data suggest that CR brings potential benefits to prostatic tissues as it reduces the physiological immune–inflammatory processes and the tissue remodeling caused by aging.
... There is much evidence to suggest that inflammation plays an important role in BPH. It has been reported that the expression levels of NLRP1 and caspase-1, IL-18 and IL-1β are elevated in BPH [45]. Therefore, the NLRP1/caspase-1 pathway is activated and participates in the development of BPH. ...
Article
Full-text available
Pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, is thought to be closely associated with the pathogenesis of diseases. Recently, the association between pyroptosis and urinary diseases has attracted considerable attention, and a comprehensive review focusing on this issue is not available. In this study, we reviewed the role of pyroptosis in the development and progression of benign urinary diseases and urinary malignancies. Based on this, pyroptosis has been implicated in the development of urinary diseases. In summary, this review sheds light on future research directions and provides novel ideas for using pyroptosis as a powerful tool to fight urinary diseases.
... A clinical study indicated that inflammasome components were elevated in prostate tissues from patients with BPH, suggesting that inflammasomes are important mediators of prostate inflammation associated with BPH [12]. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is associated with a wide range of diseases, including chronic inflammatory disorders [13]. ...
Article
Full-text available
Mitoquinone (MitoQ), a mitochondria-targeted antioxidant, has been used to treat several diseases. The present study aimed to investigate the therapeutic effects of MitoQ in benign prostatic hyperplasia (BPH) models and their underlying molecular mechanisms. In this study, we determined that MitoQ inhibited dihydrotestosterone (DHT)-induced cell proliferation and mitochondrial ROS by inhibiting androgen receptor (AR) and NOD-like receptor family pyrin domain-containing 3 (NLRP3) signaling in prostate epithelial cells. Molecular modeling revealed that DHT may combine with AR and NLRP3, and that MitoQ inhibits both AR and NLRP3. AR and NLRP3 downregulation using siRNA showed the linkage among AR, NLRP3, and MitoQ. MitoQ administration alleviated pathological prostate enlargement and exerted anti-proliferative and antioxidant effects by suppressing the AR and NLRP3 signaling pathways in rats with BPH. Hence, our findings demonstrated that MitoQ is an inhibitor of NLPR3 and AR and a therapeutic agent for BPH treatment.
... The NLRP3 inflammasome is involved in the production of IL-1β, a mediator of prostate inflammation associated with BPH, chronic prostatitis, and chronic pelvic pain syndrome [28,29]. The secretion of IL-1β is tightly regulated [30]. ...
Article
Full-text available
i>Trichomonas vaginalis is a flagellated protozoan that causes trichomoniasis, a common nonviral sexually transmitted infection. T. vaginalis infection is asymptomatic in most infected men but can lead to chronic infection. The inflammatory response to chronic T. vaginalis infection may contribute to prostatic diseases, such as prostatitis and benign prostatic hyperplasia (BPH); however, studies on the relationship between T. vaginalis infection and prostate diseases are scarce. In this review, we discuss evidence from our studies on the involvement of T. vaginalis in the pathogenesis of prostate diseases, such as prostatitis and BPH. Studies of prostatitis have demonstrated that the attachment of T. vaginalis trophozoite to prostate epithelial cells (PECs) induces inflammatory cytokine production and inflammatory cell migration, leading to prostatitis. T. vaginalis also causes pathological changes, such as inflammatory cell infiltration, acinar changes, interstitial fibrosis, and mast cell infiltration, in prostate tissues of infected rats. Thus, T. vaginalis is considered an infectious agent that triggers prostatitis. Meanwhile, studies of prostatic hyperplasia revealed that mast cells activated by T. vaginalis -infected prostate cells secreted inflammatory mediators, such as β-hexosaminidase and tryptase, which promoted proliferation of prostate stromal cell (PSC). Moreover, interleukin-6 produced by proliferating PSCs induced the multiplication of BPH-1 epithelial cells as a result of stromal–epithelial interaction, suggesting that the proliferation of T. vaginalis -infected prostate cells can be induced through crosstalk with mast cells. These collective findings suggest that T. vaginalis contributes to the progression of prostatitis and prostatic hyperplasia by creating an inflammatory microenvironment involving PECs and PSCs.
... In the process of activation of inflammatory innate immune system, in addition to cytokines playing an important role in the inflammatory process, inflammasome is also indispensable in promoting the formation and transformation of cytokines. Chronic bacterial prostatitis is through the upregulation of inflammasomes NLRP1 and NLRP3, and the increased expression of ASC and caspase-1, which together promote the conversion of downstream IL-1b and IL-18 precursors, promote the release of mature cytokines, and participate in the inflammatory and immune processes (99)(100)(101). Therefore, considering the role of inflammasome can provide a new target for the treatment of prostatitis. ...
Article
Full-text available
Up to 50% of infertility is caused by the male side. Varicocele, orchitis, prostatitis, oligospermia, asthenospermia, and azoospermia are common causes of impaired male reproductive function and male infertility. In recent years, more and more studies have shown that microorganisms play an increasingly important role in the occurrence of these diseases. This review will discuss the microbiological changes associated with male infertility from the perspective of etiology, and how microorganisms affect the normal function of the male reproductive system through immune mechanisms. Linking male infertility with microbiome and immunomics can help us recognize the immune response under different disease states, providing more targeted immune target therapy for these diseases, and even the possibility of combined immunotherapy and microbial therapy for male infertility.
Article
Background: Autophagy is a well-conserved catabolic process that plays a key role in cell homeostasis. In the prostate, defective autophagy has been implicated in the genesis and progression of several pathological conditions. Aim: The present review explored the autophagy pathway in prostate-related dysfunctions, focusing on prostate cancer (PCa), benign prostatic hyperplasia (BPH) and prostatitis. Results: Impaired autophagy activity has been shown in animal models of BPH and prostatitis. Moreover, autophagy activation by specific and non-specific drugs improved both conditions in pre-clinical studies. Conversely, the efficacy of autophagy inducers in PCa remains controversial, depending on intrinsic PCa characteristics and stage of progression. Intriguingly, autophagy inhibitors have shown beneficial effects in PCa suppression or even to overcome chemotherapy resistance. However, there are still open questions regarding the upstream mechanisms by which autophagy is deregulated in the prostate and the exact role of autophagy in PCa. The lack of specificity and increased toxicity associated with the currently autophagy inhibitors limits its use clinically, reflecting in reduced number of clinical data. Conclusion: New therapeutic strategies to treat prostatic diseases involving new autophagy modulators, combination therapy and new drug formulations should be explored. Understanding the autophagy signaling in each prostatic disease is crucial to determine the best pharmacological approach.
Article
Full-text available
The largest solid organ of the male genitalia, the prostate gland, is comprised of a variety of cells such as prostate epithelial cells, smooth muscle cells, fibroblasts, and endothelial cells. Prostate diseases, especially prostate cancer and prostatitis, are often accompanied by acute/chronic inflammatory responses or even cell death. Pyroptosis, a cell death distinct from necrosis and apoptosis, which mediate inflammation may be closely associated with the development of prostate disease. Pyroptosis is characterized by inflammasome activation via pattern recognition receptors (PRR) upon recognition of external stimuli, which is manifested downstream by translocation of gasdermin (GSDM) protein to the membrane to form pores and release of inflammatory factors interleukin (IL)-1β and IL-18, a process that is Caspase-dependent. Over the past number of years, many studies have investigated the role of inflammation in prostate disease and have suggested that pyroptosis may be an important driver. Understanding the precise mechanism is of major consequence for the development of targeted therapeutic strategies. This review summarizes the molecular mechanisms, regulation, and cellular effects of pyroptosis briefly and then discuss the current pyroptosis studies in prostate disease research and the inspiration for us.
Article
Full-text available
Macrophage dysfunction and inflammasome activation have been implicated in the pathogenesis of diabetes and its complications. Prolonged inflammation and impaired healing are hallmarks of the diabetic response to tissue injury, and excessive inflammasome activation has been implicated in these phenotypes. However, the mechanisms that regulate the inflammasome in response to lipid metabolic and inflammatory stress are incompletely understood. We have previously shown that IL-1β secretion is induced in primary macrophages exposed to the dietary saturated fatty acid (SFA) palmitate in combination with LPS. In this study, we sought to unravel the mechanisms underlying the activation of this lipotoxic inflammasome. We demonstrate that palmitate-loaded primary macrophages challenged with LPS activate the NLRP3 inflammasome through a mechanism that involves the lysosome. Interestingly, the lysosome was involved in both the regulation of pro-IL-1β levels and its subsequent cleavage/release. The lysosomal protease cathepsin B was required for IL-1β release, but not pro-IL-1βproduction. In contrast, disrupting lysosomal calcium regulation decreased IL-1β release by reducing pro-IL-1β levels. The calcium pathway involved the calcium activated phosphatase calcineurin, which stabilized IL-1β mRNA. Our findings provide evidence that the lysosome plays a key role in both the priming and assembly phases of the lipostoxic inflammasome. These findings have potential relevance to the hyper-inflammatory phenotypes observed in diabetics during tissue damage or infection and identify lysosomes and calcineurin as potential therapeutic targets.
Article
Full-text available
Unlabelled: Close links have been noted between chronic inflammation of the prostate and the development of human prostatic diseases such as benign prostate hyperplasia (BPH) and prostate cancer. However, the molecular mechanisms that contribute to prostatic inflammation remain largely unexplored. Recent studies have indicated that the IFN-inducible AIM2 protein is a cytosolic DNA sensor in macrophages and keratinocytes. Upon sensing DNA, AIM2 recruits the adaptor ASC and pro-CASP1 to assemble the AIM2 inflammasome. Activation of the AIM2 inflammasome cleaves pro-interleukin (IL)-1β and pro-IL-18 and promotes the secretion of IL-1β and IL-18 proinflammatory cytokines. Given that human prostatic infections are associated with chronic inflammation, the development of BPH is associated with an accumulation of senescent cells with a proinflammatory phenotype, and the development of prostate cancer is associated with the loss of IFN signaling, the role of AIM2 in mediating the formation of prostatic diseases was investigated. It was determined that IFNs (α, β, or γ) induced AIM2 expression in human prostate epithelial cells and cytosolic DNA activated the AIM2 inflammasome. Steady-state levels of the AIM2 mRNA were higher in BPH than in normal prostate tissue. However, the levels of AIM2 mRNA were significantly lower in clinical tumor specimens. Accordingly, constitutive levels of AIM2 mRNA and protein were lower in a subset of prostate cancer cells as compared with BPH cells. Further, the cytosolic DNA activated the AIM2 inflammasome in the androgen receptor-negative PC3 prostate cancer cell line, suggesting that AIM2-mediated events are independent of androgen receptor status. Implications: The AIM2 inflammasome has a fundamental role in the generation of human prostatic diseases.
Article
Full-text available
The mechanism of non-bacterial chronic prostatitis (CP/CPPS) has long been investigated but remains unclear. Under the hypothesis that abnormal response of innate immunity may be a cause of CP/CPPS, this study evaluated inflammasome, as part of innate immunity, and its effects on persist inflammation and CP/CPPS. Carrageenan was used to induce CP/CPPS in a rat animal model. After confirming tactile hyper-algesia in the rats, their local prostate inflammation status, and inflammasome expression were determined. The amount of inflammasome and its downstream protein was checked, along with prostate localization. Chlorogenic acid (CHA), an active ingredient of Chinese herbal remedy for CP/CPPS treatment, was used as treatment. The rats had CP/CPPS once scrotal static tactile allodynia developed and CHA treatment relieved the scrotal hypersensitivity. Downstream inflammasome proteins like IL-1β and caspase 1 increased within the prostate and decreased with CHA treatment. Inflammasome, NALP1 but not NALP3, was significantly increased in the prostate glandular endothelial cells. Treatment with CHA also changed the distribution pattern of NALP1 in the prostate. There is a close relationship between activation of inflammasome and patho-physiologic changes of CP/CPSS in rats. Increased inflammasome may be a possible mechanism of CP/CPPS and clinically active regimen may inhibit the inflammasome-related pathway. This provides a new therapeutic rationale and approach for CP/CPPS treatment. Prostate 73: 391–397, 2013.
Article
Inflammation plays a key role in the pathogenesis of obesity. Chronic overfeeding leads to macrophage infiltration in the adipose tissue, resulting in proinflammatory cytokine production. Both microbial and endogenous danger signals trigger assembly of the intracellular innate immune sensor Nlrp3, resulting in caspase-1 activation and production of proinflammatory cytokines IL-1 beta and IL-18. Here, we showed that mice deficient in Nlrp3, apoptosis-associated speck-like protein, and caspase-1 were resistant to the development of high-fat diet-induced obesity, which correlated with protection from obesity-induced insulin resistance. Furthermore, hepatic triglyceride content, adipocyte size, and macrophage infiltration in adipose tissue were all reduced in mice deficient in inflammasome components. Monocyte chemoattractant protein (MCP)-1 is a key molecule that mediates macrophage infiltration. Indeed, defective inflammasome activation was associated with reduced MCP-1 production in adipose tissue. Furthermore, plasma leptin and resistin that affect energy use and insulin sensitivity were also changed by inflammasome-deficiency. Detailed metabolic and molecular phenotyping demonstrated that the inflammasome controls energy expenditure and adipogenic gene expression during chronic overfeeding. These findings reveal a critical function of the inflammasome in obesity and insulin resistance, and suggest inhibition of the inflammasome as a potential therapeutic strategy.
Article
Purpose: We determined whether bladder inflammation causes elevated expression of nerve growth factor by bladder parenchymal cells, leading to alterations in neurons innervating the bladder. To answer this question biochemical, histological and neuronal size data were obtained in rats following various experimental models of bladder inflammation. Materials and Methods: Chemical (2.5% formalin), immune (lipopolysaccharide 2 x 10(4) cfu/ml.) and mechanical (chromic catgut) inflammation was evaluated at various times and compared to control bladders. Hematoxylin and eosin, and Giemsa staining was done to characterize inflammation and quantify mast cells in the bladder. Nerve growth factor protein and messenger RNA were assayed in the bladder and major pelvic ganglion using 2-site enzyme-linked immunosorbent assay and reverse transcriptase-polymerase chain reaction, respectively. Retrograde axonal tracing was done to size bladder neurons in the major pelvic and dorsal root ganglia. Results: All forms of inflammation increased bladder weight and produced diffuse hyperplasia, intramural edema, acute and chronic inflammatory cells, infiltration and mastocytosis. Generally bladder inflammation resulted in a 50% increase in nerve growth factor and 52% to 58% enlargement of peripheral neurons. Conclusions: Inflammation results in altered nerve growth factor content of the bladder, and morphological changes in sensory and motor neurons innervating the bladder. Such neuroplasticity may be a possible explanation for the association of bladder inflammation with long-term symptoms and pain after inflammation subsides.
Article
Purpose: Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine, is found in preformed stores in bladder epithelium. We examined the effects of prostatic inflammation on micturition frequency, bladder histology and bladder MIF content as a model in which to study viscerovisceral reflexes mediating pelvic visceral inflammation. Materials and Methods: Cystometry was performed in urethane anesthetized male rats. Formalin or saline was injected into the ventral lobe of the prostate to induce inflammation. Cystometry continued 1 hour after injection. The bladder, ventral lobes of the prostates and lumbosacral spinal cord were then removed, and protein levels and gene expression of MIF, cyclooxygenase-2 (COX-2) and nerve growth factor (NGF) were examined. Edema was verified histologically in the bladder and prostate. Results: Intraprostatic formalin produced almost immediate bladder hyperreflexia, which was maintained during the observation period. Bladder edema was noted during histological examination. Bladder MIF protein amounts decreased, while COX-2 and NGF increased after prostatic injection. Bladder MIF, COX-2 and NGF mRNA increased. In the lumbosacral spinal cord protein and mRNA amounts increased for all factors examined in animals that received intra-prostatic formalin. No changes were observed in the cervical cord. Rats injected with formalin mixed with dye showed restriction of the dye to the prostate. Conclusions: A viscerovisceral reflex in the rat, probably mediated by the lumbosacral spinal cord, produced bladder hyperreflexia and bladder edema, and evoked MIF release from the bladder and the induction of other inflammatory mediators. This supports our hypothesis that MIF is involved in neurogenic inflammation in the pelvic viscera and it may represent an interesting therapeutic target.
Article
Inflammasome-mediated host defenses have been extensively studied in innate immune cells. Whether inflammasomes function for innate defense in intestinal epithelial cells, which represent the first line of defense against enteric pathogens, remains unknown. We observed enhanced Salmonella enterica serovar Typhimurium colonization in the intestinal epithelium of caspase-11-deficient mice, but not at systemic sites. In polarized epithelial monolayers, siRNA-mediated depletion of caspase-4, a human ortholog of caspase-11, also led to increased bacterial colonization. Decreased rates of pyroptotic cell death, a host defense mechanism that extrudes S. Typhimurium-infected cells from the polarized epithelium, accounted for increased pathogen burdens. The caspase-4 inflammasome also governs activation of the proinflammatory cytokine, interleukin (IL)-18, in response to intracellular (S. Typhimurium) and extracellular (enteropathogenic Escherichia coli) enteric pathogens, via intracellular LPS sensing. Therefore, an epithelial cell-intrinsic noncanonical inflammasome plays a critical role in antimicrobial defense at the intestinal mucosal surface.
Article
Although inflammation plays an important role in the development of benign prostatic hyperplasia (BPH), little is known about the exact mechanism underlying this pathogenesis. Here, we investigated the relationship between the inflammatory reaction and BPH. cDNA microarray analysis was used to identify changes in inflammation-related gene expression in a recently established rat model that mimics human BPH. To investigate the genes identified in the analysis, quantitative (q)RT-PCR, Western blotting, immunostaining, and a cell proliferation assay were conducted using BPH model tissues, human prostate tissues, and normal human prostate cultured cells. Of the 31,100 genes identified in the cDNA analysis, seven inflammatory-response-related genes were expressed at a >2-fold higher level in rat BPH tissues than in normal rat prostate tissues. The levels of the most commonly expressed pro-inflammatory cytokine, IL-18, significantly increased in rat BPH tissues. In humans, IL-18 was localized in the epithelial and stromal components, while its receptor was strongly localized in smooth muscle cells. Furthermore, in human prostate smooth muscle cell line (PrSMC), IL-18 effected dose-dependent increases in the phosphorylated Akt and thrombospondin-1 (TSP-1) levels. TSP-1 promoted proliferation of the human prostate stromal cells (PrSC). IL-18 may act directly in BPH pathogenesis by inducing TSP-1 production in prostatic smooth muscle cells via Akt phosphorylation. Prostate. © 2014 Wiley Periodicals, Inc.
Article
Prostatic inflammation is associated with the development of prostatic hyperplasia. We investigated the effects of prostatic inflammation on expression levels of androgen-responsive genes and growth factors in the prostate. Prostatic inflammation was induced by formalin injection into bilateral ventral lobes of the prostate of male SD rats. After 28 days, the prostate was harvested for analyses of proinflammatory cytokines, androgen-responsive genes in the epithelium, and TGF-β1 cascade genes in the stroma. Some rats were given a COX-2 inhibitor (celecoxib; 10 mg/kg/day) by oral gavage for 28 days. The formalin-injected prostate exhibited widespread low-grade inflammation (<50 leukocytes/10,000 μm(2) ) along with focal high-grade inflammation (>100 leukocytes/10,000 μm(2) ) in limited areas. Compared to control, formalin-injected prostate exhibited a 2.5-fold to sixfold increased protein expression of IL-1α, IL-1β, and IL-6. In the low-grade inflammatory regions, threefold to ninefold and twofold to threefold upregulations of mRNA levels of androgen receptors/androgen-responsive genes and TGF-β1 cascade genes were respectively, observed in the epithelium and stroma obtained by laser-capture microdissection. Positive staining for androgen receptors in the epithelial nuclei, and TGF-β1, IL-6, and COX-2 in the stroma was increased in the low-grade inflammation area. COX-2 inhibitor treatment suppressed these upregulations of cytokines, androgen-responsive, and TGF-β1 cascade genes. Prostatic inflammation induced increased expression of androgen-responsive genes in the epithelium and TGF-β1 cascade genes in the stroma, which were suppressed by COX-2 inhibitors, suggesting that activation of these genes in the low-grade inflammatory region might be involved in the development of symptomatic BPH. Prostate 74:337-345, 2014. © 2013 Wiley Periodicals, Inc.
Article
Smoking is a recognized cardiovascular risk factor. Perivascular visceral adipose (PVAT) is a source of inflammatory molecules, thus contributing to atherosclerosis progression. The P2X7 receptor (P2X7 R)-inflammasome complex, crucial in determining IL-1β and IL-18 and release, participates in this scenario. We evaluated whether smoking might affect the PVAT inflammatory phenotype and explored the putative role of the axis P2X7 R-inflammasome in this picture. TNFα, IL-6, RBP4, MCP-1, as well as P2X7 R and inflammasome components NLRP3, ASC, caspase-1 and IL-1β and IL-18 expression was determined in adipocytes isolated by PVAT of healthy smokers (Smok) and non-smokers (No-Smok) subjects. Plasma and culture medium levels of these cytokines were also determined. PVAT of Smok had a higher expression of P2X7 R and inflammasome components; via P2X7 R activation, it released more IL-1β and IL-18, whose serum levels were also higher in Smok than in No-Smok. Linear correlations of NLRP3 with P2X7 R and IL-18 expression and release emerged. Smok also had a higher PVAT expression of the chemotactic factor MCP-1. However, no difference was observed in the PVAT expression of genes more strictly related to insulin resistance, like TNFα, RBP4, IL-6; this was coupled with similar plasma levels of TNFα and RBP4 in the two groups. Smoking contributes to the pro-inflammatory status of the perivascular adipose tissue by enhancing expression and activity of the P2X7 R-inflammasome complex; the effect on adipocytokines more related with insulin resistance and metabolic abnormalities appears trivial. This article is protected by copyright. All rights reserved.