ArticlePDF Available

Protective effects of neem (Azadirachta indica A. Juss.) leaf extract against cigarette smoke- and lipopolysaccharide-induced pulmonary inflammation

Authors:

Abstract

Neem (Azadirachta indica A. Juss.) leaf has been reported to exert anti-inflammatory, antibacterial and antioxidant effects. The purpose of this study was to investigate the protective effects of neem leaf extract (NLE) against cigarette smoke (CS)- and lipopolysaccharide (LPS)-induced pulmonary inflammation. Treatment with NLE significantly attenuated the infiltration of inflammatory cells, such as neutrophils and macrophages in bronchoalveolar lavage fluid (BALF). NLE also reduced the production of reactive oxygen species and the activity of neutrophil elastase in BALF. Moreover, NLE attenuated the release of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 in BALF. NLE inhibited the recruitment of inflammatory cells and the expression of monocyte chemoattractant protein-1 (MCP-1) in the lungs of mice with CS- and LPS-induced pulmonary inflammation. NLE also decreased the expression of inducible nitric oxide synthase (iNOS) in the lungs of the mice CS- and LPS-induced pulmonary inflammation. Furthermore, treatment with NLE significantly attenuated the activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in the lungs mice exposed to CS and LPS. NLE also inhibited the phosphorylation of nuclear factor (NF)-κB and inhibitor of NF-κB (IκB) in the lungs of mice expose to CS and LPS. These findings thus suggest that NLE has potential for use in the treatment of chronic obstructive pulmonary disease.
INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE
Abstract. Neem (Azadirachta indica A. Juss.) leaf has
been reported to exert anti-inflammatory, antibacterial and
antioxidant effects. The purpose of this study was to investigate
the protective effects of neem leaf extract (NLE) against cigarett e
smoke (CS)- and lipopolysaccharide (LPS)-induced pulmonary
inflammation. Treatment with NLE significantly attenuated
the inltration of inammatory cells, such as neutrophils and
macrophages in bronchoalveolar lavage uid (BALF). NLE
also reduced the production of reactive oxygen species and
the activity of neutrophil elastase in BALF. Moreover, NLE
attenuated the release of pro-inf lammatory cytokines, such
as tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 in
BALF. NLE inhibite d the recr uit ment of inamm atory cells and
the expression of monocyte chemoattractant protein-1 (MCP-1)
in the lungs of mice with CS- and LPS-induced pulmonary
inammation. NLE also decreased the expression of inducible
nitric oxide synthase (iNOS) in the lungs of the mice CS- and
LPS-induced pulmonary inammation. Furthermore, treatment
with NLE signicantly attenuated the activation of extracellular
signal-regulated kinase (ERK) and c-Jun N-terminal
kinase (JNK) in the lungs mice exposed to CS and LPS. NLE
also inhibited the phosphorylation of nuclear factor (NF)-κB
and inhibitor of NF-κB (IκB) in the lungs of mice expose to CS
and LPS. These ndings thus suggest that NLE has potential for
use in the treatment of chronic obstructive pulmonary disease.
Introduction
Chronic obstructive pulmonary disease (COPD) is a chronic
airway disease that leads to difculties breathing (1), and is
characterized by chronic inflammation of the respiratory
tract with increased numbers of inflammatory cells and
molecules (2). The worldwide incidence, prevalence and
mortality of COPD are increasing (3). Cigarette smoke (CS) is
a complex mixture of chemicals generated from the burning of
tobacco (4), and is the main cause of COPD (5). CS affects the
recruitment of inammatory cells, including neutrophils into
the lungs and is associated with chronic inammation of the
airways and a decline in lung function (6).
Neutrophils are the host defense inammatory cells that are
rapidly recruited to sites of infection (7). However, neutrophilic
inammation is the major cause of pulmonary inammation
in COPD pathophysiology (8). Activated neutrophils produce
several cytotoxic mediators, including reactive oxygen
species (ROS) and neutrophil elastase (NE), which aggravate
pulmonary inammation and emphysema (9). The increased
production of ROS accelerates the development of COPD through
the activation of mitogen-activated protein kinases (MAPKs)
and nuclear factor-κB (NF-κB) (10). NE activity is increased in
the lungs affected by COPD, which enhances the destruction
of alveolar structure (11). Tumor necrosis factor-α (TNF-α)
is a central inf lammatory cytokine that is associated with
Protective effects of neem (Azadirachta indica A. Juss.) leaf extract
against cigarette smoke- and lipopolysaccharide-induced
pulmonary inammation
JAE-WON LEE1, HYUNG WON RYU1, SO-YEON PARK1, HYUN AH PARK1,2,
OK-KYOUNG KWON1, HEUNG JOO YUK1, KRISHNA K. SHRESTHA3, MINWOO PARK4, JUNG HEE KIM1,
SANGWOO LEE5, SEI-RYANG OH1 and KYUNG-SEOP AHN1
1Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology,
Chungju-si, Chungbuk 363-883; 2College of Pharmacy, Chungnam National University, Daejeon 305-764,
Republic of Korea; 3Ethnobotanical Society of Nepal (ESON), Central Department of Botany, Tribhuvan University,
Kathmandu 44618, Nepal; 4SciTech Korea, Gangbuk-gu, Seoul 142-705; 5International Biological Material Research Center,
Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
Received March 30, 2016; Accepted September 25, 2017
DO I: 10. 3892/ ijm m.2017.3178
Correspondence to: Dr Kyung-Seop Ahn, Natural Medicine Research
Center, Korea Research Institute of Bioscience and Biotechnology,
30 Yeongudanji-ro, Cheongwon-gu, Chungju-si, Chungbuk 363-883,
Republic of Korea
E-mail: ksahn@kribb.re.kr
Abbreviations: COPD, chronic obstructive pulmonary disease;
CS, cigarette smoke; LPS, lipopolysaccharide; BALF, broncho-
alveolar lavage fluid; ROS, reactive oxygen species; NE, neutrophil
elastase; NLE, neem leaf extract; TNF-α, tumor necrosis factor-α;
IL-6, interleukin-6; MCP-1, monocyte chemoattractant protein-1;
iNOS, inducible nitric oxide synthase; MAPKs, mitogen-activated
protein kinases; NF-κB, nuclear factor-κB; IκB, inhibitor of NF-κB
Key wo rds: neem, chronic obstructive pulmonary disease, cigarette
smoke, lipopolysaccharide, neutrophil, mitogen-activated protein
kinases, nuclear factor-κB
LEE et al: ANTI-I NFLAMMATORY EFFECTS OF NLE AGAINST CS- AND LPS-INDUCED PULMONARY INFLAMM ATION
2
many immune-mediated diseases, including COPD (12). It
is well known that the constitutive overexpression of TNF-α
affects the recruitment of inflammatory cells and promotes
emphysema in the lungs of animals (13). Interleukin (IL)-6 is
a pro-inammatory cytokine that plays a pivotal role in the
pathogenesis of COPD by modulating pulmonary function (14).
Monocyte chemoattractant protein-1 (MCP-1) is one of the key
chemokines that contributes to the recruitment of inammatory
cells, such as neutrophils (15) and macrophages (16). MCP-1
levels are significantly increased in patients with COPD
compared with non-smokers (17). Inducible nitric oxide
synthase (iNOS) expression is induced by neutrophils and
macrophages in response to pro-inammatory stimuli (18,19)
and is known to have anti-inflammatory activity (20,21).
However, the continuous expression of iNOS is associated
with pulmonary inammation and emphysema (22). Recently,
it has also been reported that iNOS expression is higher in
the lungs of patients with COPD than non-smokers (23). The
MAPK signaling pathway promotes the inammatory response
by enhancing inammatory gene transcription (6,24). NF-κB
is a central transcription factor that plays an important role in
the expression of inammatory genes, such as iNOS, TNF-α
and IL-6 (25). CS has been shown to affect the activation of
MAPKs (26) and NF-κB (27).
Neem (Azadirachta indica A. Juss.) belonging to the family,
Meliaceae is an evergreen tree, cultivated in various parts of the
Indian subcontinent (28). The neem leaf has been reported to
exhibit various pharmacological activities, including anti-inam-
matory (29), antioxidant (30,31), antimicrobial (32) and antiviral
properties (33). Active constituents of the neem leaf include
nimbin, nimbidine, isomeldenin, β-sitosterol and quercetin (34).
Quercetin (35), β-sitoserol (36) and nimbidine (37) have been
shown to exert anti-inammatory effects. These effects are due
to the inhibition of pro-inammatory molecules, such as TNF-α,
iNOS and NF-κB. Recently, neem leaf extract (NLE) has been
reported to protect against endotoxemia in mice exposed to lipo-
polysaccharide (LPS) (38). However, to date, at least to the best
of our knowledge, the protective effects of NLE have not been
demonstrated in CS- and LPS-induced pulmonary inamma-
tion. Thus, the aim of this study was to investigate the protective
effects of NLE against cigarette smoke (CS)- and lipopolysac-
charide (LPS)-induced pulmonary inammation.
Materials and methods
Preparation of NLE. Neem leaf was collected from ward no. 11,
Hetauda, Nepal (latitude 27˚27'11.7'', longitude 85˚00'11.1'' and
531 m above sea level), and identied by Mr. M.R. Poudeyal of
the Ethnobotanical Society of Nepal (ESON). Voucher speci-
mens recorded as KRIB 0059759 and 760 have been deposited
in the herbarium of the Korea Research Institute of Bioscence
and Biotechnolgy (KRIB). After drying and grinding the
leaves of neem, the powder (52 g) was added to 100 liters of
methanol. The extraction was carried out using the method of
repercolation at room temperature. The extract was ltered and
concentrated by a rotavapor under reduced pressure, thereby
obtaining 2.99 g of neem methanolic extract. In the following
experiment, the neem leaves were dissolved in dimethyl sulf-
oxide (DMSO) at a concent ration of 20 mg/m l, and then diluted
to various concentrations prior to use.
Model of CS- and LPS-induced pulmonary inammation. CS-
and LPS-induced pulmonary inammation was induced using
a modication of the procedure described by Lee et al (6).
Briey, a total of 30 C57BL/6 mice (6 weeks old; weight, 20 g;
n=6/group) were whole-body exposed to room fresh air or CS
of 7 cigarettes for 50 min a day for 9 days. CS was generated
by 3R4F research cigarettes (Tobacco and Health Research
Institute, University of Kentucky, Lexington, KY, USA). LPS
was instilled intrana sally on day 8 (5 µg dissolved in 50 µl
distilled water). The mice were randomly divided into 5 groups
as follows: the normal control (NC), the CS + LPS (CS with
intranasal LPS instillation) group, the ROF (CS with intranasal
LPS instillation) + roumilast [10 mg/kg, per os (p.o)] group, and
the NLE 10 or 20 (CS with intranasal LPS instillation) + NLE
(10 or 20 mg/kg, p.o) groups. All the animal experiments were
approved by the Institutional Animal Care and Use Committee
of t h e Kore a Re s ea r ch I n stit u t e of Bio s c ienc e and Biot e chn o l o g y
and performed in compliance with the National Institutes of
Health Guidelines for the Care and Use of Laboratory Animals
and National Animal Welfare Law of Korea.
Measurement of inflammatory cells in bronchoalveolar
lavage uid (BALF). BALF collection was performed using
the method of Shin et al (5). In brief, the mice were admin-
istered an intraperitoneal injection of a pentobarbital (50 mg/
kg; Hanlim Pharm, Co., Seoul, Korea) 24 h after the final
challenge, and a tracheostomy was performed. To obtain the
BALF, ice-cold phosphate-buffered saline (PBS) (0.7 ml) was
infused into the lung and withdrawn via tracheal cannulation
twice (total volume, 1.4 ml). To determine differential cell
counts, 100 µl of BALF were centrifuged at 1,500 rpm for
5 min and the number of neutrophils and macrophages was
counted using Diff-Quik® staining reagent according to the
manufacturer's instructions (IMEB Inc., Deereld, IL, USA).
Measurement of ROS and NE in BALF. The effects of NLE on
the production of ROS were determined using 2',7'-dichlorou-
orescein diacetate (DCFH-DA; Sigma-Aldrich, St. Louis, MO,
USA). Briey, the in a m mator y cells were isolate d from BALF
and incubated with 20 µM DCFH-DA for 10 min at 37˚C. The
level of intracellular ROS was then determined using a uores-
cence microscope at 488 nm excitation and 525 nm emission (8).
The activity of NE was examined using N-succinyl-(Ala)3-p-
nitroanilide (Sigma-Aldrich) in 37˚C for 90 min, according to
the protocol described by Sakuma et al (39).
Measurement of the level of pro-inammatory cytokines in
BA L F. The levels of pro-inammatory cytokines (TNF-α and
IL-6) in BALF were determined using ELISA according to the
manufacturer's instructions (R&D Systems, Shanghai, China).
The absorbance was measured at 450 nm using a microplate
reader (Molecular Devices, Sunnyvale, CA, USA), as previ-
ously described (4).
Western blot analysis. Lung tissues were homogenized using
a homogenizer with a lysis buffer (Intron Biotechnology, Inc.,
Seoul, Korea). Protein samples were denatured and resolved
on 10% SDS-polyacrylamide gels and transferred onto a
nitrocellulose membrane. The membrane was incubated
with blocking solution for 1 h. Specific antibodies against
INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 3
MCP-1 (1;1,000; ab25124; Abcam, Cambridge, MA,
USA), iNOS (1;1,000; ADI-905-431; Enzo Life Sciences,
Farmingdale, NY, USA), p-ERK (1:1,000; #9101; Cell
Signaling Technology, Inc., Danvers, MA, USA), ERK
(1:1,000; sc-154; Santa Cruz Biotechnology, Santa Cruz, CA,
USA), p-J N K (1:1,000; KAP-SA011; Enz o Life Sciences), JNK
(1:1,000; sc-474; Santa Cruz Biotechnology), p-p38 (1:1,000;
ADI-KAP-MA022; Enzo Life Sciences), p-38 (1:1,000;
sc-7149; Santa Cruz Biotechnology), p-p65 (1:1,000; #3033;
Cell Signaling Technology, Inc.), p65 (1:1,000; sc-372; Santa
Cruz Biotechnology), p-inhibitor of NF-κB (IκB; 1:1,000;
sc-371; Santa Cruz Biotechnology) and β-actin (1;2,500;
#4967; Cell Signaling Technology, Inc.) were incubated over-
night at 4˚C with 5% skim milk. The membranes were washed
in Tris-buffered saline with Tween 20 (TBST) and incubated
with the Peroxidase-AfniPure goat anti-mouse IgG (H+L)
(1:2,000; 115-035-003; Jackson ImmunoResearch
Laboratories, Inc., West Grove, PA, USA) and the Peroxidase-
AfniPure goat anti-rabbit IgG (H+L) (1:2,000; 111-035-003;
Jackson ImmunoResearch Laboratories, Inc.) for 2 h at room
temperature. The blots were washed 3 times with TBST, and
then developed with an enhanced chemiluminescence (ECL)
kit (Amersham Biosciences, Piscataway, NJ, USA).
Histological analysis. After the BALF samples were collected,
lung tissues were xed in 10% (v/v) neutral-buffered formalin
solution. For histological examination, the lung tissues were
embedded in parafn, sectioned at 4 µm thickness, and stained
with a hematoxylin and eosin (H&E) solution (Sigma-Aldrich)
to estimate the inammatory response.
Statistical analysis. All values shown in the figures are
expressed as the means ± SD obtained from at least 3 indepen-
dent experiments. Statistical signicance was carried out using
a two-tailed Student's t-test. A p-value <0.05 was considered to
indicate a statistically signicant difference.
Results
NLE inhibits the inltration of inammatory cells in the BALF
of mice with CS- and LPS-induced pulmonary inammation.
Given the fact that the inltration of inammatory cells, such as
neutrophils and macrophages is increased in the BALF of mice
with CS- and LPS-induced pulmonary inflammation (9), we
investigated whether NLE inhibits the inltration of neutrophils
and macrophages in BALF. As shown in Fig. 1, we observed that
increased numbers of neutrophils and macrophages were detected
in the BALF of mice in the CS and LPS group compared with
those in the normal control group. However, treatment with NLE
signicantly attenuated the numbers of neutrophils and macro-
phages in BALF, compared with the CS and LPS group in a
concentration-dependent manner (Fig. 1). The effect of 20 mg/kg
NLE was similar to that of treatment with 10 mg/kg ROF.
NLE attenuates the production of ROS and NE in BALF.
It is well known that ROS production and NE activity are
Figure 1. Effect of neem leaf extract (NLE) on the inltration of neutrophils
and macrophages in the bronchoalveolar lavage uid (BALF) of mice with
cigarette smoke (CS)- and lipopolysacchar ide (LPS)-induced pulmona ry
inammation. (A and B) The BALF differential cell count was determined
using the Diff-Quick® staining reagent (x400 magnication). The values are
expressed as means ± SD (n=6 mice per g roup). NC, normal control mice with
PBS only; CS + LPS, cigarette smoke (CS) and lipopolysaccharide (LPS);
ROF, roumilast (10 mg/kg) + CS and LPS; NLE 10 or 20, NLE (10 or 20 mg/
kg) + CS and LPS. #p<0.01 indicates a statistically signicant difference from
the normal control group. *p<0.05 and **p<0.01 indicate statistically signicant
differences compared to t he CS and LPS group.
Figure 2. Effect of neem leaf extract (NLE) on the production of reactive
oxygen species (ROS) and neutrophil elastase (NE) in bronchoalveolar lavage
uid (BALF). (A) ROS production and (B) NE activity. Data are expressed
as the means ± SD. #p<0.01 indicates a statistically signicant difference
from the normal control group. *p<0.05 and **p<0.01 indicate statistically
signicant differences compared to the cigarette smoke (CS) and lipopolysac-
charide (LPS) group.
LEE et al: ANTI-I NFLAMMATORY EFFECTS OF NLE AGAINST CS- AND LPS-INDUCED PULMONARY INFLAMM ATION
4
increased in the BALF of mice with CS- and LPS-induced
pulmonary inammation (5,6). Thus, in this study, the levels of
ROS and NE were examined in the BALF of mice with CS and
LPS-induced pulmonary inammation. As shown in Fig. 2,
the levels of ROS and NE were signicantly increased in the
CS and LPS group. However, treatment with NLE signicantly
decreased the levels of ROS and NE (Fig. 2). In particular,
treatment with 20 mg/kg NLE more effectively attenuated the
levels of those molecules compared with 10 mg/kg ROF.
NLE decreases the levels of TNF-α and IL- 6 in BALF. The
increased release of TNF-α and IL-6 in BALF is one of the
major characteristics of COPD (5). Thus, to determine whether
NLE affects the release of pro-inflammatory cytokines in
Figure 4. Effect of neem leaf extract (NLE) on the inltration of inammatory cells in lungs of mice with cigarette smoke (CS)- and lipopolysaccharide (LPS)-induced
pulmonar y inam mation. (A) Peribronchial lesion (x400 magnication): (a) negative control, (b) CS + LPS, (c) roumilast, (d) NLE 10 mg/kg and (e) NLE 20 mg/
kg. (B) Quantitative analysis of airway inammation in lung tissue stained with H&E solution. (C) Monocyte chemoattractant protein-1 (MCP-1) expression was
detected by western blot analysis. NC, normal control mice with PBS only; CS + LPS, cigarette smoke (CS) and lipopolysaccharide (LPS); ROF, roumilast (10 mg/
kg) + CS and LPS; NLE 10, NLE (10 mg/kg) + CS and LPS; NLE 20, NLE (20 mg /kg) + CS an d LPS. Data are expressed as the mean s ± SD. #p<0.01 indicates a sta-
tistically signicant difference from the ormal control group. *p<0.05 and **p<0.01 indicate statistically signicant differences compared with the CS and LPS group.
Figure 3. Effect of neem leaf extract (NLE) on the levels of pro-inammatory cytokines, such as tumor necrosis factor-α (T NF-α) and interleukin-6 (IL- 6)
in bronchoalveolar lavage uid (BALF). (A) The levels of TNF-α and (B) IL-6 were measured by ELISA. The absorbance was measured at 450 nm using a
microplate reader. Data are expressed as the means ± SD. #p<0.01 indicates a statistically signicant differencefrom the normal control group. **p<0.01 indicate
statistically signicant differences compa red to the cigarette smoke (CS) and lipopolysaccharide (LPS) group.
INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 5
BALF, the levels of TNF-α and IL-6 were examined by ELISA.
As shown in Fig. 3, treatment with NLE effectively inhibited the
release of these cytokines in BALF.
NLE reduces the recruitment of inflammatory cells and
the expression of MCP-1 in the lungs of mice with CS- and
LPS-induced pulmonary inammation. To examine whether
NLE affects the recruitment of inflammatory cells and the
expression of MCP-1 in the lungs of mice with CS- and
LPS-induced pulmonary inf lammation, the infiltration of
inammatory cells was determined by H&E staining. As shown
in Fig. 4A and B, the mice in the CS and LPS group exhibit ed an
increased inltration of inammatory cells. However, treatment
with NLE signicantly reduced the recruitment of inamma-
tory cells in a concentration-dependent manner. Consistent with
the decrease in inammatory cell recruitment, treatment with
NLE also signicantly decreased the expression of MCP-1 in
the lungs, suggesting that NLE attenuated the recruitment of
inammatory cells (Fig. 4C). Similar to the results shown above,
the effect of 20 mg/kg NLE was similar to that of treatment with
10 mg/kg ROF.
NLE inhibits the expression of iNOS in lungs of mice with CS-
and LPS-induced pulmonary inammation. As the increased
expression of iNOS induced by neutrophils (40) and macro-
phages (2) is an important in the pathologenesis of COPD, we
investigated whether NLE affects the level of iNOS in the lungs
of mice with CS- and LPS-induced pulmonary inammation.
As shown in Fig. 5, iNOS expression was increased in the lungs
of mice in the CS and LPS group. However, treatment with
NLE effectively inhibited the expression of iNOS, compared
with normal control mice.
NLE attenuates the activation of ERK and JNK in the lungs
of mice with CS- and LPS-induced pulmonary inammation.
MAPK activation plays an important role in the inammatory
response regulating the release of pro-inammatory cytokines
and mediators. Thus, we investigated whether NLE treatment
attenuates the activation of MAPKs in the lungs of mice with
CS- and LPS-induced pulmonary inflammation. As shown
in Fig. 6, the activation of MAPKs (ERK, JNK and p38) was
signicantly increased in the lungs of mice in the CS and LPS
group. However, treatment with NLE signicantly decreased
the activation of ERK and JNK in a concentration-dependent
manner (Fig. 6A and B). The inhibitory effect of 20 mg/kg
Figure 6. Effect of neem leaf extract ( NLE) on the activation of ERK and
JNK in lungs of mice. (A) The activation of ERK, (B) JNK and (C) p38 was
detected by western blot analysis. Data a re expressed as the means ± SD.
#p<0.01 indicates a statistically signicant difference from the normal con-
trol group. *p<0.05 and **p<0.01 indicate statistically signicant differences
compared to the cigarette smoke (CS) and lipopolysaccharide (LPS) group.
Figure 5. Effect of neem leaf extract (NLE) on the expression of inducible
nitric oxide synthase (iNOS) in lungs of mice. The expression of iNOS was
detected by western blot analysis. Data a re expressed as the means ± SD.
#p<0.01 indicates a statistically signicant difference from the normal control
group. *p<0.05 indicate statistically signicant differences compared to the
cigarette smoke (CS) and lipopolysaccharide (LPS) group.
LEE et al: ANTI-I NFLAMMATORY EFFECTS OF NLE AGAINST CS- AND LPS-INDUCED PULMONARY INFLAMM ATION
6
NLE on ERK and JNK activation was similar to that of treat-
ment with 10 mg/kg ROF. No signicant attenuation of p38
activation was observed with NLE (Fig. 6C).
NLE decreases the phosphorylation of NF-κB and IκB in
lungs of mice with CS- and LPS-induced pulmonary inam-
mation. NF-κB is activated by a number of stimuli, including
pro-inammatory mediators and LPS. In response to these
molecules, IκB is phosphorylated, ubiquitinated and degraded,
resulting in the phosphorylation and nuclear translocation of
NF-κB (20,41). In the present study, treatment with NLE inhib-
ited the phosphorylation of NF-κB and IκB in the lungs of mice
with CS- and LPS-induced pulmonary inammation (Fig. 7).
Discussion
In the present study, we examined the protective effects of NLE
against CS- and LPS-induced pulmonary inammation. NLE
sign i cantly in h i bited the inlt ra t i on of in a m m a t o r y cells, such
as neutrophils and macrophages in BALF. NLE also reduced
the production of ROS and NE, and decreased the release of
pr o -in a m mato r y cyto k ines in BAL F. NLE at t enu ated th e accu-
mulation of inammatory cells and the expression of MCP-1 in
the lungs of mice with CS- and LPS-induced pul monar y inam-
mation. Furthermore, NLE inhibited the expression of iNOS in
the lungs of mice with CS- and LPS-induced pul monar y inam-
mation. NLE also attenuated the activation of MAPKs (ERK
and JNK) and NF-κB in the lung tissue.
COPD is a global health epidemic the incidence of which is
increasing (42), and it is associated with a high risk of morbidity
and mortality (43). COPD is characterized by chronic airway
inflammation and mucus hypersecretion (44). It is also well
known that CS exposure and bacterial infection are associated
with the development of COPD (4,45,46). CS is the most impor-
tant risk factor that increases the recruitment of inammatory
cells in the lungs and the number of goblet cells in the small
airway (47). LPS is a major constituent of the Gram-negative
bacterial cell wall that stimulates the inammatory response (48).
The recruitment of inflammatory cells, such as neutrophils
and macrophages in the airways is a characteristic sign of
COPD (6,49,50). ROS production promote the inammatory
response in the lungs via the activation of transcription factors,
such as NF-κB and MAPK signal transduction pathways (10).
Increased ROS production induced by neutrophils has been
reported to promote the oxidation of proteins, DNA and lipids
which leads to lung damage (10,51). A number of studies have
reported that NE levels are increased in response to CS (52,53) or
CS and LPS (5,6), which increases the inammatory cell recruit-
ment, emphysema and the production of mucus in the lungs (54).
CS is the most important source of elevated levels of ROS and
NE in COPD (55). In the present study, treatment with NLE
signicantly inhibited inammatory cell inltration in BALF
and in the lungs of mice with CS- and LPS-induced pulmonary
inammation (Figs. 1 and 4A and B). NLE also attenuated the
production of ROS and the activity of NE (Fig. 2).
Pro-inammatory cytokines, including TNF-α and IL-6 play
an important role in the pathological processes of COPD (56).
TNF-α is a central cytokine that regulates inammation through
neutrophil recruitment and endothelial activation (57). IL-6 is
involved in the pathogenesis of lung diseases, such as COPD (58).
It has also been reported that exposure to CS increases macro-
phage accumulation that contributes to the development of
COPD by increasing the levels of IL-6 (59). Recently, TNF-α
and IL-6 were identied to be involved in CS- and LPS-induced
COPD (4,5,60). In the present study, NLE decreased the release
of TNF-α and IL-6 in BALF (Fig. 3). MCP-1 is a chemokine
that plays a key role in the migration of neutrophils and macro-
phages (15,16). Recently, the increased expression of MCP-1 was
detected in the lungs of mice exposed to CS (61). The present
data demonstrated benecial effects of NLE against the CS- and
LPS-induced expression of MCP-1 (Fig. 4C). iNOS has been
implicated in the pathophysiology of inammatory diseases,
including COPD (23), and the high expression of iNOS has been
reported to affect pulmonary inammation (62). It has also been
reported that the inhibition of iNOS exerts protective effects in
a wide variety of respiratory diseases (63). The present study
dem onstrated that N L E sig n ic a ntly sup p r e ssed the ex p r essio n of
iNOS in the lungs of mice with CS and LPS-induced pulmonary
inammation in a concentration-dependent manner (Fig. 5).
MAPKs have been reported to regulate pro-inammatory
molecules (64,65) and have been widely studied in pulmonary
inammation (4,5,66). MAPKs (ERK, JNK and p38) mediate
pro-inammatory gene transcription in response to cytokines
and LPS (5,67). CS leads to the activation of MAPKs (68-71).
It has also been reported that MAPK activation affects ROS
production in lungs affected by COPD (10). The present data
demonstrated that the activation of MAPKs (ERK, JNK
and p38) was induced by CS and LPS in the lungs of mice.
However, NLE treatment signicantly inhibited the activation
or ERK and JNK (Fig. 6A and B). No signicant inhibition of
p38 activation was observed with NLE treatment (Fig. 6C).
Figure 7. Effect of neem leaf extract (NLE) on the activation of nuclear
fac tor-κB (NF-κB) in lungs of mice. The phosphorylation of NF-κB and
inhibitor of NF-κB (IκB) was detected by western blot analysis. Data are
expressed as the means ± SD. #p<0.01 indicates a statistically signicant
difference f rom the normal control group. *p<0.05 and **p<0.01 indicate
statistically signicant differences compared to the cigarette smoke (CS) and
lipopolysaccharide (LPS) group.
INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 7
NF-κB is a key transcription factor in the inammatory
response, and is activated by numerous extracellular stimuli,
including pro-inflammatory cytokines, such as TNF-α and
IL-6 (10). The NF-κB-dependent production of these cyto-
kines affects the recruitment of inflammatory cells, such
as neutrophils and macrophages to lung tissue, causing lung
injury or emphysema (9,72,73) Therefore, NF-κB signaling is
considered to be an important therapeutic target for pulmonary
inammation induced by CS (74). In this study, NLE treatment
signicantly inhibited the elevated phosphorylation levels of
NF-κB and IκB induced by CS and LPS in lung tissue (Fig. 7).
The neem tree (Azadirachta indica A. Juss.; Meliaceae) is
indigenous to India, and now this tree is cultivated widely in
areas of the world (75). Azadirachta indica A. Juss has been
widely used as neem and has been used in medicine for over
2,0 00 years (76). Various parts of the neem tree have been used
in medicines and food, as well as as insecticides, and many
bioactive constituents, including limonoids (tetra-nortriterpe-
noids) have been isolated and identied (77). NLE has been
reported to possess antibacterial activity (78-80). It has also
been demonstrated that NLE induces apoptosis in the breast
cancer cells (81). Neem leaf fraction has been reported to
possess antioxidant properties (82,83). Recently, it has also
been shown that NLE protects LPS-induced endotoxemia (38).
However, to date, at least to the best of our knowledge, the
protective effects of NLE have not been investigated in CS- and
LPS-induced pulmonary inammation.
In conclusion, the present data demonstrated that NLE
signicantly inhibited the inltration of inammatory cells, such
as neutrophils and macrophages in the lungs of mice with CS-
and LPS-induced pulmonary inammation. NLE also attenuated
the production of inammatory mediators, including ROS, NE,
TNF-α and IL-6 in BALF. Furthermore, NLE decreased the
expression of MCP-1 and iNOS in the lungs of mice with CS-
and LPS-induced pulmonary inammation. NLE also inhibited
the activation of MAPKs (ERK and JNK) and NF-κB in the lungs
of mice. These results thus suggest that NLE may have potential
for use as a valuable therapeutic agent in the treatment of COPD.
Acknowledgements
This study was supported by a grant from the Ministry of
Science, ICT and Future Planning (FGC 1011534), Ministry
for Health and Welfare (HI14C1277) and the KRIBB Research
Initiative Program (KGM 1221713) of the Republic of Korea.
References
1. Lee H, Jung KH, Lee H, Park S, Choi W and Bae H: Casticin,
an active compound isolated from Vitex Fructus, ameliorates the
cigarette smoke-induced acute lung inammatory response in a
murine model. Int Immunopharmacol 28: 1097-1101, 2015.
2. Barnes PJ: Chronic obstructive pulmonary disease * 12: new
treatments for COPD. Thorax 58: 803-808, 2003.
3. Park YC, Jin M, Kim SH, Kim MH, Namgung U and Yeo Y:
Effects of inhalable microparticle of ower of Lonicera japonica
in a mous e mo d e l of CO P D. J Et h nopha r ma c o l 151: 123-1 3 0, 2014.
4. Bak JH, Lee SM and Lim HB: Safety assessment of mainstream
smoke of herbal cigarette. Toxicol Res 31: 41-48, 2015.
5. Shin IS, Shin NR, Park JW, Jeon CM, Hong JM, Kwon OK,
Kim JS, Lee IC, Kim JC, Oh SR, et al: Melatonin attenuates
neutrophil inflammation and mucus secretion in cigarette
smoke-induced chronic obstructive pulmonary diseases via the
suppression of Erk-Sp1 signaling. J Pineal Res 58: 50-60, 2015.
6. Lee JW, Shin NR, Park JW, Park SY, Kwon OK, Lee HS,
Hee Kim J, Lee HJ, Lee J, Zhang ZY, et al: Callicarpa japonica
Thunb. attenuates cigarette smoke-induced neutrophil inam-
mation and mucus secretion. J Ethnopharmacol 175: 1-8, 2015.
7. Kruger P, Saffarzadeh M, Weber AN, Rieber N, Radsak M,
von Bernuth H, Benarafa C, Roos D, Skokowa J and Hartl D:
Neutrophils: between host defence, immune modulation, and
tissue injury. PLoS Pathog 11: e1004651, 2015.
8. Shin IS, Ahn KS, Shin NR, Lee HJ, Ryu HW, Kim JW, Sohn KY,
Kim HJ, Han YH and Oh SR: Protective effect of EC-18, a
synthetic monoacetyldiglyceride on lung inflammation in a
murine model induced by cigarette smoke and lipopolysac-
charide. Int Immunopharmacol 30: 62-68, 2016.
9. Song HH, Shin IS, Woo SY, Lee SU, Sung MH, Ryu HW,
Kim DY, Ahn KS, Lee HK, Lee D, et al: Piscroside C, a novel
iridoid glycoside isolated from Pseudolysimachion rotundum
va r. subinegrum suppresses airway inammation induced by
cigarette smoke. J Ethnopharmacol 170: 20-27, 2015.
10. Rahman I and Adcock IM: Oxidative stress and redox regulation
of lung inammation in COPD. Eur Respir J 28: 219-242, 2006.
11. Shapiro SD, Goldstein NM, Houghton AM, Kobayashi DK,
Kelley D and Belaaouaj A: Neutrophil elastase contributes to
cigarette smoke-induced emphysema in mice. Am J Pathol 163:
2329-2335, 2003.
12. Mukhopadhyay S, Hoidal JR and Mukherjee TK: Role of
TNFalpha in pulmonary pathophysiology. Respir Res 7:
125, 2006.
13. Lundblad LK, Thompson-Figueroa J, Leclair T, Sullivan MJ,
Poynter ME, Irvin CG and Bates JH: Tumor necrosis factor-alpha
overexpression in lung disease: a single cause behind a complex
phenotype. Am J Respir Crit Care Med 171: 1363-1370, 2005.
14. Rincon M and Irvin CG: Role of IL-6 in asthma and other inam-
matory pulmonary diseases. Int J Biol Sci 8: 1281-1290, 2012.
15. Wan MX, Wang Y, Liu Q, Schramm R and Thorlacius H: CC
chemokines induce P-selectin-dependent neutrophil rolling
and recruitment in vivo: intermediary role of mast cells. Br J
Pharmacol 138: 698-706, 2003.
16. Deshmane SL, Kremlev S, Amini S and Sawaya BE: Monocyte
chemoattractant protein-1 (MCP-1): an overview. J Interferon
Cytokine Res 29: 313-326, 2009.
17. Traves SL, Culpitt SV, Russell RE, Barnes PJ and Donnelly LE:
Increased levels of the chemokines GROalpha and MCP-1
in sputum samples from patients with COPD. Thorax 57:
590-595, 2002.
18. McNeill E, Crabtree MJ, Sahgal N, Patel J, Chuaiphichai S,
Iqbal AJ, Hale AB, Greaves DR and Channon KM: Regulation
of iNOS function and cellular redox state by macrophage Gch1
reveals specic requirements for tetrahydrobiopterin in NRF2
activation. Free Radic Biol Med 79: 206-216, 2015.
19. Webb JL, Polak JM and Evans TJ: Effect of adhesion on inducible
nitric oxide synthase (iNOS) production in purified human
neutrophils. Clin Exp Immunol 123: 42-48, 2001.
20. Lee JW, Bae CJ, Choi YJ, Kim SI, Kwon YS, Lee HJ, Kim SS and
Chun W: 3,4,5-Trihydroxycinnamic acid inhibits lipopolysac-
charide (LPS)-induced inammation by Nrf2 activation in vitro
and improves survival of mice in LPS-induced endotoxemia
model in vivo. Mol Cell Biochem 390: 143-153, 2014.
21. Marinovic MP, Morandi AC and Otton R: Green tea catechins
alone or in combination alter functional parameters of human
neutrophils via suppressing the activation of TLR-4/NFκB p65
signal pathway. Toxicol In Vitro 29: 1766-1778, 2015.
22. Roh GS, Yi CO, Cho YJ, Jeon BT, Nizamudtinova IT, Kim HJ,
Kim JH, Oh YM, Huh JW, Lee JH, et al: Anti-inammatory
effects of celecoxib in rat lungs with smoke-induced emphysema.
Am J Physiol Lung Cell Mol Physiol 299: L184-L191, 2010.
23. Jiang WT, Liu XS, Xu YJ, Ni W and Chen SX: Expression of
nitric oxide synthase isoenzyme in lung tissue of smokers with
and without chronic obstructive pulmonary disease. Chin Med J
(Engl) 128: 1584-1589, 2015.
24. Singh D: P38 inhibition in COPD; cautious optimism. Thorax 68:
705-706, 2013.
25. Oh YC, Jeong YH, Ha JH, Cho WK and Ma JY: Oryeongsan
inhibits LPS-induced production of inf lammatory mediators
via blockade of the NF-kappaB, MAPK pathways and leads to
HO-1 induction in macrophage cells. BMC Complement Altern
Med 14: 242, 2014.
26. Liang Z, Xie W, Wu R, Geng H, Zhao L, Xie C, Li X, Zhu M,
Zhu W, Zhu J, et al: Inhibition of tobacco smoke-induced bladder
MAPK activation and epithelial-mesenchymal transition in mice
by curcumin. Int J Clin Exp Pathol 8: 4503-4513, 2015.
LEE et al: ANTI-I NFLAMMATORY EFFECTS OF NLE AGAINST CS- AND LPS-INDUCED PULMONARY INFLAMM ATION
8
27. Li L, Sun J, Xu C, Zhang H, Wu J, Liu B and Dong J: Icariin
ameliorates cigarette smoke induced inammatory responses via
suppression of NF-κB and modulation of GR in vivo and in vitro.
PLoS One 9: e102345, 2014.
28. Mahfuzul Hoque MD, Bari ML, Inatsu Y, Juneja VK and
Kawamoto S: Antibacterial act ivity of guava (Psid ium guaj ava L.)
and Neem (Azadirachta indica A. Juss.) extracts against
foodborne pathogens and spoilage bacteria. Foodborne Pathog
Dis 4: 481-488, 2007.
29. Okpanyi SN and Ezeukwu GC: Anti-inammatory and anti-
pyretic activities of Azadirachta indica. Planta Med 41:
34-39, 1981.
30. Rao AD, Devi KN and Thyagaraju K: Isolation of antioxidant
principle from A zadirachta seed kernels: determination of its
role on plant lipoxygenases. J Enzyme Inhib 14: 85-96, 1998.
31. Yanpallewar SU, Sen S, Tapas S, Kumar M, Raju SS and
Acharya SB: Effect of Azadirachta indica on parac etamol-induced
hepatic damage in albino rats. Phytomedicine 10: 391-396, 2003.
32. Almas K: The antimicrobial effects of extracts of Azadi-
rachta indica (Neem) and Salvadora persica (Arak) chewing
sticks. Indian J Dent Res 10: 23-26, 1999.
33. Badam L, Joshi SP and Bedekar SS: ‘In vitro’ antiviral activity of
neem (Azadirachta indica. A. Juss) leaf extract against group B
coxsackieviruses. J Commun Dis 31: 79-90, 1999.
34. Siddiqui BS, Afshan F, Gulzar T and Hanif M: Tetra-
cyclic triterpenoids from the leaves of Azadirachta indica.
Phytochemistry 65: 2363-2367, 2004.
35. Chang YC, Tsai MH, Sheu WH, Hsieh SC and Chiang AN: The
therapeutic potential and mechanisms of action of quercetin in
relation to lipopolysaccharide-induced sepsis in vitro and in vivo.
PLoS One 8: e80744, 2013.
36. Loizou S, Lekakis I, Chrousos GP and Moutsatsou P: Beta-sitosterol
exhibits anti-inammatory activity in human aortic endothelial
cells. Mol Nutr Food Res 54: 551-558, 2010.
37. Pillai NR and Santhakumari G: Anti-arthritic and anti-inam-
matory actions of nimbidin. Planta Med 43: 59-63, 1981.
38. Kim WH, Song HO, Jin CM, Hur JM, Lee HS, Jin HY, Kim SY
and Park H: The methanol extract of Azadirachta indica A. Juss
leaf protects mice against lethal endotoxemia and sepsis. Biomol
Ther (Seoul) 20: 96-103, 2012.
39. Sakuma T, Takahashi K, Ohya N, Usuda K, Handa M and
Abe T: ONO-5046 is a potent inhibitor of neutrophil elastase in
human pleural effusion after lobectomy. Eur J Pharmacol 353:
273-279, 1998.
40. Rytilä P, Rehn T, Ilumets H, Rouhos A, Sovijärvi A,
Myllärniemi M and Kinnula VL: Increased oxidative stress
in asymptomatic current chronic smokers and GOLD stage 0
COPD. Respir Res 7: 69, 2006.
41. Lee JW, Kwon JH, Lim MS, Lee HJ, Kim SS, Lim SY and Chun W:
3,4,5-Trihydroxycinnam ic acid increases heme-oxygenase-1 (HO-1)
and decreases macrophage infiltration in LPS-induced septic
kidney. Mol Cell Biochem 397: 109-116, 2014.
42. Barnes PJ: Chronic obstructive pulmonary disease: a growing
but neglected global epidemic. PLoS Med 4: e112, 2007.
43. Vestbo J, Hurd SS, Agustí AG, Jones PW, Vogelmeier C,
Anzueto A, Barnes PJ, Fabbri LM, Martinez FJ,
Nishimura M, et al: Global strategy for the diagnosis, mana-
gement, and prevention of chronic obstructive pulmonary
disease: GOLD executive summary. Am J Respir Crit Care
Med 187: 347-365, 2013.
44. Yang J, Yu HM, Zhou XD, Huang HP, Han Zh, Kolosov VP
and Perelman JM: Cigarette smoke induces mucin hyperse-
cretion and inammatory response through the p66shc adaptor
protein-mediated mechanism in human bronchial epithelial cells.
Mol Immunol 69: 86-98, 2016.
45. Shao MX, Nakanaga T and Nadel JA: Cigarette smoke
induces MUC5AC mucin overproduction via tumor necrosis
factor-alpha-converting enzyme in human airway epithelial
(NCI-H292) cells. Am J Physiol Lung Cell Mol Physiol 287:
L420-L 427, 2004.
46. Chung KF: Inflammatory mediators in chronic obstructive
pulmonary disease. Curr Drug Targets Inf lamm Allergy 4:
619-625, 2005.
47. Thorley AJ and Tetley TD: Pulmonary epithelium, cigarette
smoke, and chronic obstructive pulmonary disease. Int J Chron
Obstruct Pulmon Dis 2: 409-428, 2007.
48. Kwak HG and Lim HB: Inhibitory effects of Cnidium monnieri
fruit extract on pulmonary inammation in mice induced by
cigarette smoke condensate and lipopolysaccharide. Chin J Nat
Med 12: 641-647, 2014.
49. Baraldo S, Turato G, Badin C, Bazzan E, Beghé B, Zuin R,
Calabrese F, Casoni G, Maestrelli P, Papi A, et al: Neutrophilic
inltration within the airway smooth muscle in patients with
COPD. Thorax 59: 308-312, 2004.
50. Li H, Yang T, Ning Q, Li F, Chen T, Yao Y and Sun Z: Cigarette
smoke extract-treated mast cells promote alveolar macrophage
inltration and polarization in experimental chronic obstructive
pulmonary disease. Inhal Toxicol 27: 822-831, 2015.
51. Neofytou E, Tzortzaki EG, Chatziantoniou A and Siafakas NM:
DNA damage due to oxidative stress in chronic obstructive
pulmonary disease (COPD). Int J Mol Sci 13: 16853-16864, 2012.
52. Chan KH, Chan SC, Yeung SC, Man RY, Ip MS and Mak JC:
Inhibitory effect of Chinese green tea on cigarette smoke-induced
up-regulation of airway neutrophil elastase and matrix metal-
loproteinase-12 via antioxidant activity. Free Radic Res 46:
1123-1129, 2012.
53. Iizuka T, Ishii Y, Itoh K, Kiwamoto T, Kimura T, Matsuno Y,
Morishima Y, Hegab AE, Homma S, Nomura A, et al:
Nrf2-deficient mice are highly susceptible to cigarette
smoke-induced emphysema. Genes Cells 10: 1113-1125, 2005.
54. Bhowmik A, Chahal K, Austin G and Chakravorty I: Improving
mucociliar y clearance in chronic obstructive pulmonary disease.
Respir Med 103: 496-502, 2009.
55. van E eden SF and Sin DD: Oxidative stress in chronic obstructive
pulmonary disease: a lung and systemic process. Can Respir
J 20: 27-29, 2013.
56. Hwang JH, Lee BJ, Jung HJ, Kim KI, Choi JY, Joo M and
Jung SK: Effects of Chung-pae inhalation therapy on a mouse
model of chronic obstructive pulmonary disease. Evid Based
Complement Alternat Med 2015: 461295, 2015.
57. Lee E, Yun N, Jang YP and Kim J: Lilium lancifolium Thunb.
extract attenuates pulmonary inflammation and air space
enlargement in a cigarette smoke-exposed mouse model. J
Ethnopharmacol 149: 148-156, 2013.
58. Pedroza M, Schneider DJ, Karmouty-Quintana H, Coote J,
Shaw S, Corrigan R, Molina JG, Alcorn JL, Galas D,
Gelinas R, et al: Interleukin-6 contributes to inammation and
remodeling in a model of adenosine mediated lung injury. PLoS
One 6: e22667, 2011.
59. Fernandez-Real JM, Broch M, Vendrell J and Ricart W: Smoking,
fat mass and activation of the tumor necrosis factor-alpha
pathway. Int J Obes Relat Metab Disord 27: 1552-1556, 2003.
60. Xu GH, Shen J, Sun P, Yang ML, Zhao PW, Niu Y, Lu JK,
Wang ZQ, Gao C, Han X, et al: Anti-inammatory effects of
potato extract on a rat model of cigarette smoke-induced chronic
obstructive pulmonary disease. Food Nutr Res 59: 28879, 2015.
61. Chen X, Guan XJ, Peng XH, Cui ZL, Luan CY and Guo XJ:
Acetylation of lysine 9 on histone H3 is associated with increased
pro-inammatory cytokine release in a cigarette smoke-induced
rat model through HDAC1 depression. Inflamm Res 64:
513-526, 2015.
62. Zhao Y, Cui A, Wang F, Wang XJ, Chen X, Jin ML and
Huang KW: Characteristics of pulmonary inflammation in
combined pulmonary fibrosis and emphysema. Chin Med J
(Engl) 125: 3015-3021, 2012.
63. Hesslinger C, Strub A, Boer R, Ulrich WR, Lehner MD and
Braun C: Inhibition of inducible nitric oxide synthase in respi-
ratory diseases. Biochem Soc Trans 37: 886-891, 2009.
64. Li D, Xu D, Wang T, Shen Y, Guo S, Zhang X, Guo L, Li X, Liu L
and Wen F: Silymarin attenuates airway inammation induced
by cigarette smoke in mice. Inammation 38: 871-878, 2015.
65. Ng DS, Liao W, Tan WS, Chan TK, Loh XY and Wong WS:
Anti-malarial drug artesunate protects against cigarette
smoke-induced lung injury in mice. Phytomedicine 21:
1638 -16 4 4, 2014.
66. Ma WJ, Sun YH, Jiang JX, Dong XW, Zhou JY and Xie QM:
Epoxyeicosatrienoic acids attenuate cigarette smoke
extract-induced interleukin-8 production in bronchial epithelial
cells. Prostaglandins Leukot Essent Fatty Acids 94: 13-19, 2015.
67. Coskun M, Olsen J, Seidelin JB and Nielsen OH: MAP kinases in
inam matory bowel disease. Cli n Ch im Acta 412: 513-520, 2011.
68. Hoshino S, Yoshida M, Inoue K, Yano Y, Yanagita M, Mawatar i H,
Yamane H, Kijima T, Kumagai T, Osaki T, et al: Cigarette smoke
extract induces endothelial cell injury via JNK pathway. Biochem
Biophys Res Commun 329: 58-63, 2005.
69. Xu X, Balsiger R, Tyrrell J, Boyaka PN, Tarran R and
Cormet-Boyaka E: Cigarette smoke exposure reveals a novel role
for the MEK/ERK1/2 MAPK pathway in regulation of CFTR.
Biochim Biophys Acta 1850: 1224-1232, 2015.
INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 9
70. Marumo S, Hoshino Y, Kiyokawa H, Tanabe N, Sato A, Ogawa E,
Muro S, Hirai T and Mishima M: p38 mitogen-activated protein
kinase determines the susceptibility to cigarette smoke-induced
emphysema in mice. BMC Pulm Med 14: 79, 2014.
71. Shen N, Gong T, Wang JD, Meng FL, Qiao L, Yang RL, Xue B,
Pan FY, Zhou XJ, Chen HQ, et al: Cigarette smoke-induced
pulmonary inflammatory responses are mediated by
EG R-1/G GPPS/ MA PK sig na l i n g. Am J Pa t h o l 178: 110-118, 2 011.
72. Metcalfe HJ, Lea S, Hughes D, Khalaf R, Abbott-Banner K and
Singh D: Effects of cigarette smoke on toll-like receptor (TLR)
activation of chronic obstructive pulmonary disease (COPD)
macrophages. Clin Exp Immunol 176: 461-472, 2014.
73. Zhao Y, Xu Y, Li Y, Xu W, Luo F, Wang B, Pang Y, Xiang Q,
Zhou J, Wang X, et al: NF-κB-mediated inammation leading to
EMT via miR-200c is involved in cell transformation induced by
cigarette smoke extract. Toxicol Sci 135: 265-276, 2013.
74. Edwards MR, Bartlett NW, Clarke D, Birrell M, Belvisi M and
Johnston SL: Targeting the NF-kappaB pathway in asthma and
chronic obstructive pulmonary disease. Pharmacol Ther 121:
1-13, 2009.
75. Akihisa T, Noto T, Takahashi A, Fujita Y, Banno N, Tokuda H,
Koike K, Suzuki T, Yasukawa K and Kimura Y: Melanogenesis
inhibitory, anti-inammatory, and chemopreventive effects of
limonoids from the seeds of Azadirachta indicia A. Juss. (neem).
J Oleo Sci 58: 581-594, 2009.
76. Faccin-Galhardi LC, Yamamoto KA, Ray S, Ray B,
Carvalho Linhares RE and Nozawa C: The in vitro antiviral
property of Azadirachta indica polysaccharides for poliovirus. J
Ethnopharmacol 142: 86-90, 2012.
77. Akihisa T, Takahashi A, Kikuchi T, Takagi M, Watanabe K,
Fukatsu M, Fujita Y, Banno N, Tokuda H and Yasukawa K: The
melanogenesis-inhibitory, anti-inammatory, and chemopreventive
effects of limonoids in n-hexane extract of Azadirachta indica A.
Juss. (neem) seeds. J Oleo Sci 60: 53-59, 2011.
78. Agyare C, Spiegler V, Sarkodie H, Asase A, Liebau E and
Hensel A: An ethnopharmacological survey and in vitro conr-
mation of the ethnopharmacological use of medicinal plants as
anthelmintic remedies in the Ashanti region, in the central part
of Ghana. J Ethnopharmacol 158: 255-263, 2014.
79. Quelemes PV, Perfeito ML, Guimarães MA, dos Santos RC,
Lima DF, Nascimento C, Silva MP, Soares MJ, Ropke CD,
Eaton P, et al: Effect of neem (Azadirachta indica A. Juss) leaf
extract on resistant Staphylococcus aureus biolm formation
and Schistosoma mansoni worms. J Ethnopharmacol 175:
287-294, 2015.
80. Sharma J, Gairola S, Sharma YP and Gaur RD: Ethnomedicinal
plants used to treat skin diseases by Tharu community of district
Udham Singh Nagar, Uttarakhand, India. J Ethnopharmacol 158:
140 -206, 2014.
81. Othman F, Motalleb G, Lam Tsuey Peng S, Rahmat A, Basri R
and Pei Pei C: Effect of neem leaf extract (Azadirachta indica)
on c-Myc oncogene expression in 4T1 breast cancer cells of
BALB/c mice. Cell J 14: 53-60, 2012.
82. Manikandan P, Anandan R and Nagini S: Evaluation of Azadi-
rachta indica leaf fractions for in vitro antioxidant potential
and protective effects against H2O2-induced oxidative damage
to pBR322 DNA and red blood cells. J Agric Food Chem 57:
6990-6996, 2009.
83. Sithisarn P, Supabphol R and Gritsanapan W: Comparison of
free radical scavenging activity of Siamese neem tree (Azadi-
rachta indica A. Juss var. siamensis Valeton) leaf extracts
prepared by different methods of extraction. Med Princ Pract 15:
219-222, 2006.
... Nimbolide is a compound found in Azadiractha indica, a plant that has been linked with in vivo and in vitro antiinflammatory effects [41][42][43][44][45]. Earlier, nimbolide was shown to reduce the expression of pro-inflammatory cytokines IL-6, IL-8, IL-12 and TNFα in LPS-stimulated RAW 264.7 and IL-10 −/− peritoneal macrophages [39]. ...
Article
Full-text available
Nimbolide, a limonoid compound found in the neem plant, was investigated for effects on neuroinflammation in BV-2 microglia activated with lipopolysaccharide (LPS). Cultured BV-2 cells were treated with nimbolide (125, 250 and 500 nM) followed by stimulation with LPS (100 ng/ml). Results showed that nimbolide caused a significant reduction in the levels of TNFα, IL-6, IFNγ, NO/iNOS and PGE2/COX-2 in LPS-activated BV-2 cells. Further experiments revealed that LPS-induced increased expression of phospho-p65 and phospho-IκBα proteins were reduced in the presence of nimbolide. Also, LPS-induced NF-κB acetylation, increased binding to consensus sites and transactivation, as well as phosphorylation of p38 and JNK MAPKs were reduced by nimbolide. Reduction of cellular ROS generation by nimbolide was accompanied by a reduction in gp91phox protein levels, while antioxidant effects were also observed through elevation in protein levels of HO-1 and NQO-1. It was observed that treatment of BV-2 microglia with nimbolide resulted in reduced levels of cytoplasmic Nrf2, which was accompanied by increased levels in the nucleus. Furthermore, treatment with this compound resulted in increased binding of Nrf2 to antioxidant responsive element (ARE) consensus sites accompanied by enhanced ARE luciferase activity. Knockdown experiments revealed a loss of anti-inflammatory activity by nimbolide in cells transfected with Nrf2 siRNA. Treatment with nimbolide resulted in nuclear accumulation of SIRT-1, while siRNA knockdown of SIRT-1 resulted in the reversal of anti-inflammatory activity of nimbolide. It is proposed that nimbolide reduces neuroinflammation in BV-2 microglia through mechanisms resulting in dual inhibition of NF-κB and MAPK pathways. It is also proposed that activation of Nrf2 antioxidant mechanisms may be contributing to its anti-inflammatory activity. Graphical Abstract
... All these parameters indicated the development of COPD. Thus, it can be corroborated that compared to pre-treatment with LPS, post-treatment with it aggravated the disease progression [39][40][41]. Another study used a 3-week exposure of MCS (2R4F reference cigarette, aerosol-0.61 ...
Article
Full-text available
Introduction and objective Chronic obstructive pulmonary disease (COPD) is a progressive disorder that makes breathing difficult, characterized by chronic bronchitis, mucus hypersecretion, airway remodelling, and emphysema. Although caused by various factors, the leading cause is active or passive cigarette smoking (CS) is. The treatment available provides symptomatic relief and is associated with serious adverse effects, such as cardiovascular events and pneumonia; hence, there is an unmet need for research on drug treatment for COPD. This literature review provides an update on the various animal models of COPD that have been developed by the exposure of CS alone, or in combination with other inducing agents. Abbreviated description of the state of knowledge The combination of SCS/MCS with LPS should be preferred to a single smoke component to induce COPD. It was observed that mouse models are extensively used, C57B1/6 and BALB/c females are more vulnerable to COPD, whereas, in rat models, male SD rats are mostly used. Guinea pigs, due to their anatomical similarity, are found to be a better model that can be used to develop COPD. Conclusions Suitable animal models and validated apparatus are crucial for successful COPD animal model development. Conventionally authenticated research-grade cigarettes should be used for effortless distribution of a specific concentration of total suspended particles (TSP) or total particulate matter (TPM), including nicotine and carbon monoxide. There is also a need to focus on the various types of apparatus to be used for COPD induction in murine models considering optimum exposure, reliability as well as the sturdiness of the apparatus which would provide better execution of the protocol with minimum harm to the experimenter.
... It is said to have health-promoting properties since it is a rich source of both nonisoprenoids and isoprenoids (especially azadirachtin) (12). Earlier investigators have confirmed their role as antibacterial (13), antifungal (14,15), antimalarial (16), anti-inflammatory (17,18), antiparasitic activities (19). ...
Article
Full-text available
One of the most crucial approaches for treating human diseases, particularly parasite infections, is nanomedicine. One of the most significant protozoan diseases that impact farm and domestic animals is coccidiosis. While, amprolium is one of the traditional anticoccidial medication, the advent of drug-resistant strains of Eimeria necessitates the development of novel treatments. The goal of the current investigation was to determine whether biosynthesized selenium nanoparticles (Bio-SeNPs) using Azadirachta indica leaves extract might treat mice with Eimeria papillata infection in the jejunal tissue. Five groups of seven mice each were used, as follows: Group 1 : Non-infected-non-treated (negative control). Group 2 : Non-infected treated group with Bio-SeNPs (0.5 mg/kg of body weight). Groups 3-5 were orally inoculated with 1×10 ³ sporulated oocysts of E. papillata . Group 3 : Infected-non-treated (positive control). Group 4: Infected and treated group with Bio-SeNPs (0.5 mg/kg). Group 5 : Infected and treated group with the Amprolium. Groups 4 and 5 daily received oral administration (for 5 days) of Bio-SeNPs and anticoccidial medication, respectively, after infection. Bio-SeNPs caused a considerable reduction in oocyst output in mice feces (97.21%). This was also accompanied by a significant reduction in the number of developmental parasitic stages in the jejunal tissues. Glutathione reduced (GSH), glutathione peroxidase (GPx), and superoxide dismutase (SOD) levels were dramatically reduced by the Eimeria parasite, whereas, nitric oxide (NO) and malonaldehyde (MDA) levels were markedly elevated. The amount of goblet cells and MUC2 gene expression were used as apoptotic indicators, and both were considerably downregulated by infection. However, infection markedly increased the expression of inflammatory cytokines ( IL-6 and TNF-α ) and the apoptotic genes ( Caspase-3 and BCL2 ). Bio-SeNPs were administrated to mice to drastically lower body weight, oxidative stress, and inflammatory and apoptotic indicators in the jejunal tissue. Our research thus showed the involvement of Bio-SeNPs in protecting mice with E. papillata infections against jejunal damage.
... Nimbolide is a compound found in Azadiractha indica, a plant that has been linked with in vivo and in vitro anti-in ammatory effects [25][26][27][28][29]. Earlier, nimbolide was shown to reduce the expression of proin ammatory cytokines IL-6, IL-8, IL-12, and TNFα in LPS-stimulated RAW 264.7 and IL10 −/− peritoneal macrophages [23]. ...
Preprint
Full-text available
Nimbolide, a limonoid compound found in the neem plant, was investigated for effects on neuroinflammation in BV2 microglia activated with LPS. Cultured BV2 cells were stimulated with LPS (100 ng/ml) followed by treatment with nimbolide (125, 250 and 500 nM). Results showed that nimbolide caused significant reduction in the levels of TNFα, IL-6, IFNγ, NO/iNOS, PGE 2 /COX-2 in LPS-activated BV2 cells. Further experiments revealed that LPS-induced increased expression of phospho-p65 and phospho-IκBα proteins were reduced in the presence of nimbolide. Also, LPS-induced NF-κB acetylation, increased binding to consensus sites, and transactivation, as well as phosphorylation of p38 and JNK MAPKs were reduced by nimbolide. Reduction of cellular ROS generation by nimbolide was accompanied by reduction in gp91phox protein levels, while antioxidant effects were observed through elevation in protein levels of HO-1 and NQO-1. It was observed that treatment of BV2 microglia with nimbolide resulted in reduced levels of cytoplasmic Nrf2 which was accompanied by increased levels in the nucleus. Furthermore, treatment with this compound resulted in increased binding of Nrf2 to ARE consensus sites accompanied by enhanced ARE luciferase activity. Knockdown experiments revealed a loss of anti-inflammatory activity by nimbolide in cells transfected with Nrf2 siRNA. Treatment with nimbolide resulted in nuclear accumulation of SIRT-1, while siRNA knockdown of SIRT-1 resulted in reversal of anti-inflammatory activity of nimbolide. It is proposed that nimbolide reduces neuroinflammation in BV2 microglia through mechanisms resulting in dual inhibition of NF-κB and MAPK pathways. It is also proposed that activation of Nrf2 antioxidant as well as SIRT-1 deacetylation mechanisms may be contributing to its anti-inflammatory activity.
... Increasing evidence demonstrated that cigarette smoke promotes increased infiltration of immune cells including macrophages and neutrophils hence, modulating pulmonary inflammation (Harrison et al. 2008;Lee et al. 2017). In addition, our study demonstrated that ATA treatment decreased the CSE-induced immune cellular influx and reduced the expression of TNF-a in the alveolar and bronchial epithelium. ...
Article
Full-text available
Cigarette smoke (CS) induced emphysema and chronic pulmonary inflammation are major comorbidities of chronic obstructive pulmonary disease (COPD), a major cause of morbidity and mortality worldwide. CS exposure exacerbates pulmonary inflammation and compromises immunity to various infections. Aurintricarboxylic acid (ATA) is a polyanionic aromatic compound especially recognized for its anti-inflammatory, nucleic acid, and protein interaction inhibition properties. The study was designed to investigate the anti-inflammatory role of ATA against cigarette smoke extract (CSE) induced pulmonary inflammation. Nicotine concentration was quantified in CSE by UPLC/MS technique. In vitro, fluorescence microscopy, and flow cytometry was performed in CSE stimulated alveolar epithelial cells to determine the effect of ATA on oxidative stress-mediated cellular apoptosis. In vivo, pulmonary inflammation was induced in male Wistar rats via a modified non-invasive intratracheal instillation of cigarette smoke extract (100 µl/animal) twice a week for 8 weeks and post-treated with ATA (10 mg/kg) intraperitoneally for 15 days. Lung homogenates were assessed for MDA and GSH. Lung tissues were subjected to western blotting and histopathological analysis. As result, ATA reduced CSE-induced chromatin condensation, fragmentation, cellular apoptosis in alveolar epithelial cells, and apoptotic biomarkers expression including BAX and Caspase-3 in the lungs. ATA reduced inflammation by normalizing redox balance reflected by MDA/GSH levels. ATA obviated airspace enlargement, fiber deposition, and immune cell infiltration. Reduced inflammation was accompanied by inhibition of inflammatory biomarkers TNF-α, TNFR1, TWEAK, and NF-ҡB/p65 activation and nuclear translocation. ATA efficaciously diminished the oxidative stress and pulmonary inflammation associated with lung pathogenesis through TNF-α/TNFR1/NF-ҡB/p65 signaling pathway. • HIGHLIGHTS • ATA treatment attenuates CSE-stimulated chromatin condensation, fragmentation, and cellular apoptosis in alveolar epithelial cells. • ATA treatment inhibits CSE stimulated activation and nuclear translocation of NF-ҡB/p65. • ATA treatment diminishes CSE-induced oxidant injury, apoptosis, and emphysema-like phenotypic changes in the lungs. • ATA inhibits lung inflammation via suppression of the NF-ҡB/p65 signaling pathway.
... In our present study, we have further elucidated that LPS induced oxidative stress and inflammatory responses, as well as altered mitochondrial bioenergetics and redox homeostasis in Rin-5F cells, and were protected by AZD treatment. Studies have suggested the strong inhibitory action of AZD on pro-inflammatory cell signaling, cytokines, and NO activation via inhibition of nuclear translocation of NF-κB (Schumacher et al., 2011;Amyot et al., 2012;Lee et al., 2017). Our present study also demonstrates strong anti-inflammatory action by AZD via inhibition of oxidative stress, cytokine release as well as NF-κB-dependent cell signaling controlling the GSHredox homeostasis. ...
Article
Full-text available
Inflammation and redox imbalance are hallmarks of cancer, diabetes, and other degenerative disorders. Pathophysiological response to these disorders leads to oxidative stress and mitochondrial dysfunction by alterations and reprogramming in cellular signaling and metabolism. Pancreatic beta cells are very sensitive to the inflammatory and altered nutrient signals and hence play a crucial role in diabetes and cancer. In this study, we treated insulin-secreting pancreatic beta cells, Rin-5F, with the bacterial endotoxin, LPS (1 μg/ml) to induce an inflammatory response in vitro and then treated the cells with a known anti-inflammatory, anticancer and antioxidant phytochemical, azadirachtin (AZD, 25 µM for 24 h). Our results demonstrated lipid peroxidation and nitric oxide production causing increased nitro/oxidative stress and alterations in the activities of anti-oxidant enzymes, superoxide dismutase and catalase after LPS treatment. Pro-inflammatory responses caused by translocation of nuclear factor kappa B and release of inflammatory cytokines were also observed. These changes were accompanied by GSH-dependent redox imbalance and alterations in mitochondrial membrane potential and respiratory complexes enzyme activities leading to mitochondrial respiratory dysfunction, reduced ATP synthesis, and intrinsic caspase-9 mediated apoptosis. Caspase-9 was activated due to alterations in Bcl-2 and Bax proteins and release of cytochrome c into the cytosol. The activities of oxidative stress-sensitive mitochondrial matrix enzymes, aconitase, and glutamate dehydrogenase were also inhibited. Treatment with AZD showed beneficial effects on the recovery of antioxidant enzymes, inflammatory responses, and mitochondrial functions. GSH-dependent redox homeostasis also recovered after the treatment with AZD. This study may help in better understanding the etiology and pathogenesis of inflammation-induced disorders in pancreatic beta cells to better manage therapeutic strategies.
... Consistent with this, a recent clinical trial suggests that subject's prophylactically taking neem capsules had a reduced risk of infection resulting in COVID-19 (Nesari et al., 2021). Moreover, neem leaf extract has been shown to inhibit inflammation associated with endotoxin-induced lung injury in mice (Lee et al., 2017), indicating that NBE may have a comparable effect in ameliorating the impact of SARS-CoV-2 on the respiratory system. ...
Article
Emerging mutations in the SARS-CoV-2 genome pose a challenge for vaccine development and antiviral therapy. The antiviral efficacy of Azadirachta indica bark extract (NBE) was assessed against SARS-CoV-2 and m-CoV-RSA59 infection. Effects of in vivo intranasal or oral NBE administration on viral load, inflammatory response, and histopathological changes were assessed in m-CoV-RSA59-infection. NBE administered inhibits SARS-CoV-2 and m-CoV-RSA59 infection and replication in vitro, reducing Envelope and Nucleocapsid gene expression. NBE ameliorates neuroinflammation and hepatitis in vivo by restricting viral replication and spread. Isolated fractions of NBE enriched in Nimbin isomers shows potent inhibition of m-CoV-RSA59 infection in vitro. In silico studies revealed that NBE could target Spike and RdRp of m-CoV and SARS-CoV-2 with high affinity. NBE has a triterpenoids origin that may allow them to competitively target panoply of viral proteins to inhibit mouse and different strains of human coronavirus infections, suggesting its potential as an antiviral against pan-β-Coronaviruses.
Article
Ethnopharmacological relevance: Azadirachta indica A. Juss (Meliaceae), popularly known as "neem", is used for the treatment of rheumatism, cancer, ulcers, diabetes, respiratory problems, among others. This species is present on six continents and contains more than 400 bioactive compounds. Practically all parts of the plant are used in the treatment of diseases. Although it is widely used, no study has evaluated the safety of this species throughout the gestational period in Wistar rats. Aim of the study: To evaluate the genotoxicity and the effect of treatment with dried extract of leaves of Azadirachta indica on maternal toxicity and fetal development. Materials and methods: The dried extract of leaves of A. indica was obtained by spray drying after percolation of the plant material in 30% ethanol (w/w). The total flavonoids and rutin contents of the extract were determined by spectrophotometric method and HPLC-DAD, respectively. Pregnant Wistar rats (n = 40) were divided into four groups (n = 10/group): one control and three groups treated with dried extract of leaves of A. indica at doses of 300, 600 or 1200 mg/kg. Treatments were carried out from gestational day (GD) 0-20. During gestation, clinical signs of toxicity, weight gain, feed and water consumption of the dams were evaluated. On GD 21, rats were euthanized and cardiac blood was collected. Liver, kidneys, lung, heart, uterus, ovaries and bone marrow were collected. Reproductive performance parameters, histopathological analysis, biochemistry and genotoxicity were evaluated. Fetuses were evaluated for external morphology, skeletal and visceral changes. Results: The total flavonoid content of the extract ranged from 2.64 to 3.01%, and the rutin content was 1.07%. There was no change in body mass gain, food and water consumption between the evaluated groups. There was also no difference between the groups in terms of biochemical parameters, reproductive performance, histopathological analysis of the mother's organs and genotoxicity. Supernumerary ossification sites of the sternum were observed, and other skeletal and visceral alterations were not significant. Conclusions: The treatment did not induce maternal toxicity, it was neither embryotoxic nor fetotoxic. The extract was not potentially genotoxic, and at a dose of 1200 mg/kg, it caused changes in the ossification of the sternum.
Article
Full-text available
Background Seeking cures for chronic inflammation-associated diseases and infectious diseases caused by critical human pathogens is challenging and time-consuming. Even as the research community searches for novel bioactive agents, consuming a healthy diet with functional ability might be an effective way to delay and prevent the progression of severe health conditions. Many plant ingredients in Thai food are considered medicinal, and these vegetables, herbs, and spices collectively possess multiple biological and pharmacological activities, such as anti-inflammatory, antimicrobial, antidiabetic, antipyretic, anticancer, hepatoprotective, and cardioprotective effects. Methodology In this review, the selected edible plants are unspecific to Thai food, but our unique blend of recipes and preparation techniques make traditional Thai food healthy and functional. We searched three electronic databases: PUBMED, Science Direct, and Google Scholar, using the specific keywords “Plant name” followed by “Anti-inflammatory” or “Antibacterial” or “Antiviral” and focusing on articles published between 2017 and 2021. Results Our selection of 69 edible and medicinal plant species (33 families) is the most comprehensive compilation of Thai food sources demonstrating biological activities to date. Focusing on articles published between 2017 and 2021, we identified a total of 245 scientific articles that have reported main compounds, traditional uses, and pharmacological and biological activities from plant parts of the selected species. Conclusions Evidence indicates that the selected plants contain bioactive compounds responsible for anti-inflammatory, antibacterial, and antiviral properties, suggesting these plants as potential sources for bioactive agents and suitable for consumption for health benefits.
Article
Full-text available
Oxidative stress and inflammation are hypothesised as the main contributor for Chronic Obstructive Pulmonary Disease (COPD). Cigarette smoke (CS), a major cause of COPD leads to inflammation resulting in recruitment of neutrophils and macrophages which are rich sources of oxidants. Activation of these cells produces excess oxidants and depletes antioxidants resulting in stress. Presently, effective drug for COPD is limited; therefore, novel compounds from natural sources, including plants are under exploration. The present study aims to investigate the protective effect of Ocimum sanctum leaf extract (OLE) in CS − induced model of COPD. Exposure to CS was performed thrice a week for 8 weeks and OLE (200 mg/kg and 400 mg/kg) was administered an hour before CS exposure. Control group (negative control) were exposed to ambient air while COPD group was exposed to CS (positive control). Administration of OLE doses reduced inflammation, decreased oxidant concentration and increased antioxidant concentration (p < 0.01). Molecular docking studies between the major phytocompounds of OLE (Eugenol, Cyclohexane and Caryophyllene) and antioxidant enzymes Superoxide dismutase (SOD), Catalase, Glutathione peroxidase (GPx), Glutathione reductase (GR) and Glutathione S Transferase (GST) showed strong binding interaction in terms of binding energy. In vivo and in silico findings for the first time indicates that OLE extract significantly alleviates oxidative stress by its potent free radical scavenging property and strong interaction with antioxidant enzymes. OLE extract may prove to be a therapeutic option for COPD prevention and treatment.
Article
Full-text available
Chung-pae (CP) inhalation therapy is a method frequently used in Korea to treat lung disease, especially chronic obstructive pulmonary disease (COPD). This study investigated the effects of CP inhalation on a COPD animal model. C57BL/6 mice received porcine pancreatic elastase (PPE) and lipopolysaccharide (LPS) alternately three times for 3 weeks to induce COPD. Then, CP (5 or 20 mg/kg) was administered every 2 h after the final LPS administration. The effect of CP was evaluated by bronchoalveolar lavage (BAL) fluid analysis, histological analysis of lung tissue, and reverse transcription polymerase chain reaction analysis of mRNA of interleukin- (IL-) 1β, tumor necrosis factor- (TNF-) α, IL-6, and tumor growth factor- (TGF-) β. Intratracheal CP administration reduced the number of leukocytes and neutrophils in BAL fluid, inhibited the histological appearance of lung damage, and decreased the mRNA levels of the proinflammatory cytokines IL-1β, TNF-α, IL-6, and TGF-β. Intratracheal CP administration effectively decreased the chronic inflammation and pathological changes in a PPE- and LPS-induced COPD mouse model. Therefore, we suggest that CP is a promising strategy for COPD.
Article
Full-text available
Objective: This study aimed to evaluate the therapeutic effects of potato extract (PE) on cigarette smoke (CS)-induced chronic obstructive pulmonary disease (COPD). Methods: PE was first prepared by frozen centrifugation, and its amino acid composition was detected. Toxicity of PE was analyzed by changes in morphology, behavior, routine blood indexes, and biochemical criteria of mice. Then, the COPD rat model was established by CS exposure, and PE, doxofylline, and prednisolone acetate were used to treat these rats. After 45 days of treatment, the morphology and behavior of rats were recorded. In addition, the histopathology of lung tissue was evaluated by chest x-ray and hematoxylin and eosin staining. The expression of interleukine-10 (IL-10), tumor necrosis factor-α (TNF-α), and granulocyte colony-stimulating factor (G-CSF) was detected in serum and lung tissue by enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry, respectively. Results: Various amino acids were identified in PE, and no toxicity was exhibited in mice. The CS-induced COPD rat model was successfully established, which exhibited significant thickened and disordered lung markings on 90% of the rats. After administering doxofylline and prednisolone acetate, inflammation symptoms were improved. However, side effects such as emaciation, weakness, and loosening of teeth appeared. In the PE group, obviously improved histopathology was observed in lung tissues. Meanwhile, it was revealed that PE could increase the expression of IL-10 and reduce the expression of TNF-α and G-CSF in COPD rats, and doxofylline and prednisolone acetate also elicited similar results. Conclusion: Our study suggests PE might be effective in the treatment of CS-induced COPD by inhibiting inflammation.
Article
Objective: Cigarette smoking is the main cause of chronic obstructive pulmonary disease (COPD) and may modulate the immune response of exposed individuals. Mast cell function can be altered by cigarette smoking, but the role of smoking in COPD remains poorly understood. The current study aimed to explore the role of cigarette smoke extract (CSE)-treated mast cells in COPD pathogenesis. Methods: Cytokine and chemokine expression as well as degranulation of bone marrow-derived mast cells (BMMCs) were detected in cells exposed to immunoglobulin E (IgE) and various doses of CSE. Adoptive transfer of CSE-treated BMMCs into C57BL/6J mice was performed, and macrophage infiltration and polarization were evaluated by fluorescence-activated cell sorting (FACS). Furthermore, a coculture system of BMMCs and macrophages was established to examine macrophage phenotype transition. The role of protease serine member S31 (Prss31) was also investigated in the co-culture system and in COPD mice. Results: CSE exposure suppressed cytokine expression and degranulation in BMMCs, but promoted the expressions of chemokines and Prss31. Adoptive transfer of CSE-treated BMMCs induced macrophage infiltration and M2 polarization in the mouse lung. Moreover, CSE-treated BMMCs triggered macrophage M2 polarization via Prss31 secretion. Recombinant Prss31 was shown to activate interleukin (IL)-13/IL-13Rα/Signal transducers and activators of transcription (Stat) 6 signaling in macrophages. Additionally, a positive correlation was found between Prss31 expression and the number of M2 macrophages in COPD mice. Conclusion: In conclusion, CSE-treated mast cells may induce macrophage infiltration and M2 polarization via Prss31 expression, and potentially contribute to COPD progression.
Article
The antler of Sika deer (Cervus nippon Temminck) has been used a natural medicine in Korea, China and Japan, and a monoacetyldiaglyceride (1-palmitoyl-2-linoleoyl-3-acetylglycerol, PLAG) was found in the antler of Sika deer as a constituent for immunomodulation. In this study, we investigated protective effects of EC-18 (a synthetic copy of PLAG) on inflammatory responses using a cigarette smoke with lipopolysaccharide (LPS)-induced airway inflammation model. Mice were exposed to cigarette smoke for 1 h per day for 3 days. Ten micrograms of LPS dissolved in 50 μL of PBS was administered intra nasally 1 h after the final cigarette smoke exposure. EC-18 was administered by oral gavage at doses of 30 and 60 mg/kg for 3 days. EC-18 significantly reduced the number of neutrophils, reactive oxygen species production, cytokines and elastase activity in bronchoalveolar lavage fluid (BALF) compared with the cigarette smoke and LPS induced mice. Histologically, EC-18 attenuated airway inflammation with a reduction in myeloperoxidase expression in lung tissue. Additionally, EC-18 inhibited the phosphorylation of NF-κB and IκB induced by cigarette smoke and LPS exposure. Our results show that EC-18 effectively suppresses neutrophilic inflammation induced by cigarette smoke and LPS exposure. In conclusion, this study suggests that EC-18 has therapeutic potential for the treatment of chronic obstructive pulmonary disease.
Article
The p66Shc adaptor protein is a newly recognized mediator of mitochondrial dysfunction and might play a role in cigarette smoke (CS)-induced airway epithelial cell injury. CS can induce an excessive amount of reactive oxygen species (ROS) generation, which can cause mitochondrial depolarization and injury through the oxidative stress-mediated Serine36 phosphorylation of p66Shc. The excessive production of ROS can trigger an inflammatory response and mucin hypersecretion by enhancing the transcriptional activity of pro-inflammatory cytokines and mucin genes. Therefore, we speculate that p66Shc plays an essential role in airway epithelial cell injury and the process of mucin generation in CS-induced chronic inflammatory airway diseases. Our present study focuses on the role of p66Shc in ROS generation, and on the resulting mitochondrial dysfunction, inflammatory response and mucus hypersecretion in CS-stimulated human bronchial epithelial cells (16HBE). We found that CS disturbed the mitochondrial function by increasing the level of phosphorylated p66Shc in these cells and that the effects were significantly reduced by silencing p66Shc. Conversely, the ectopic overexpression of wild-type p66Shc enhanced these effects. We also found that high levels of ROS inhibited FOXO3a transcriptional activity, which led to NF-κB activation. Subsequently, activated NF-κB promoted pro-inflammatory cytokine production and mucin hypersecretion. Thus, manipulating p66Shc might offer a new therapeutic modality with which to treat chronic inflammatory airway diseases.
Article
Ethnopharmacological relevance: There are ethnopharmacological reports supporting the use of neem (Azadirachta indica A. Juss) leaf against bacterial and worm infections. However there is a lack of studies about its effect on bacterial biofilm formation and Schistosoma mansoni worms. This study reports the in vitro effects of neem leaf ethanolic extract (Neem EE) on Methicillin-resistant Staphylococcus aureus (MRSA) biofilm and planktonic aggregation formation, and against S. mansoni worms. Materials and methods: Quantification of the Azadirachtin (AZA), thought to be one of their main compounds related to biological effects, was performed. The effect of sub-inhibitory concentrations of Neem EE on biofilm formation and planktonic aggregates of S. aureus was tested using the crystal violet dye method and atomic force microscopy (AFM) analysis, respectively. Changes in S. mansoni motor activity and death of worms were analyzed in vitro after exposition to the extract. Treated schistosomes were also examined using confocal laser scanning microscopy. Results: It was observed the presence of AZA in the extract (0.14±0.02mg/L). Testing Neem EE sub-inhibitory concentrations, a significant biofilm adherence inhibition from 62.5µg/mL for a sensitive S. aureus and 125µg/mL for two MRSA strains was observed. AFM images revealed that as the Neem EE concentration increases (from 250 to 1000µg/mL) decreased ability of a chosen MRSA strain to form large aggregates. In relation of anti-schistosoma assay, the extract caused 100% mortality of female worms at a concentration of 50µg/mL at 72h of incubation, while 300µg/mL at 24hours of incubation was required to achieve 100% mortality of male worms. The extract also caused significant motor activity reduction in S. mansoni. For instance, at 96h of incubation with 100µg/mL, 80% of the worms presented significant motor activity reduction. By the confocal microscopy analysis, the dorsal surface of the tegument of worms exposed to 300µg/mL (male) and 100µg/mL (female) of the extract showed severe morphological changes after 24h of treatment. Conclusions: Neem leaf ethanolic extract presented inhibitory effect on MRSA biofilm and planktonic aggregation formation, and anthelmintic activity against S. mansoni worms.
Article
Callicarpa japonica Thunb. (CJT) is traditionally used as an herbal remedy for the treatment of inflammatory diseases in Korea, China, and Japan. In this study, we evaluated the effects of Callicarpa japonica Thunb. (CJT) on the development of COPD using a Cigarette smoke (CS)-induced murine model and cigarette smoke condensate (CSC)-stimulated H292 cells, human pulmonary mucoepidermoid cell line. Callicarpa japonica Thunb. was isolated from the leaves and stem of C. japonica. The methanol extract profile was obtained by UPLC Q-TOF-MS analysis. In in vivo experiment, the mice received 1h of cigarette smoke for 10 days. Callicarpa japonica Thunb. was administered to mice by oral gavage 1h before cigarette smoke exposure for 10 days. In in vitro experiment, we evaluated the effect of Callicarpa japonica Thunb. on the expression of MUC5AC and proinflammatory cytokines in H292 cells stimulated with CSC. CJT treatment effectively suppressed the infiltration of neutrophils, and decreased the production of ROS and the activity of neutrophil elastase in the bronchoalveolar lavage fluid (BALF) induced by CS. CJT also significantly attenuated production of proinflammatory cytokines such as IL-6 and TNF-α in the BALF, and reduced the infiltration of inflammatory cells and the production of mucus in lung tissue induced by CS. In in vitro experiments, CJT decreased the expression of MUC5AC and proinflammatory cytokines in CSC-stimulated H292 cells. Furthermore, CJT attenuated the phosphorylation of ERK induced by CSC in H292 cells. Taken together, CJT effectively reduced the neutrophil airway inflammation and mucus secretion induced by CS in murine model, and inhibited the expression of MUC5AC in CSC-stimulated H292 human lung cell line. These findings suggest that CJT has a therapeutic potential for the treatment of COPD. Copyright © 2015. Published by Elsevier Ireland Ltd.
Article
Tobacco smoke (TS) has been shown to cause bladder cancer. Epithelial-mesenchymal transition (EMT) is a crucial pathophysiological process in cancer development. MAPK pathways play central roles in tumorigenesis including EMT process. Curcumin is a promising chemopreventive agent for several types of cancers. In the present study we investigated the effects of TS on MAPK pathway activation and EMT alterations in the bladder of mice, and the preventive effect of curcumin was further examined. Results showed that exposure of mice to TS for 12 weeks resulted in activation of ERK1/2, JNK, p38 and ERK5 MAPK pathways as well as AP-1 proteins in bladder. TS reduced mRNA and protein expression levels of epithelial markers E-cadherin and ZO-1, while mRNA and protein expression levels of the mesenchymal markers vimentin and N-cadherin were increased. Curcumin treatment effectively attenuated TS-triggered activation of ERK1/2, JNK and p38 MAPK pathways, AP-1 proteins and EMT alterations in bladder tissue. These results suggest the protective effects of curcumin in TS-induced MAPK activation and EMT, thus providing new insights into the chemoprevention of TS-associated bladder cancer.