Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Subst Use Addict Treat ; 160: 209311, 2024 May.
Article in English | MEDLINE | ID: mdl-38336263

ABSTRACT

INTRODUCTION: A growing literature indicates bidirectional associations between pain and tobacco use. Cigarette smokers are at increased risk for chronic pain, and observational and experimental studies indicate that pain increases motivation to smoke. Tobacco use disorder frequently co-occurs with other substance use disorders, which are also associated with chronic pain vulnerability. Despite evidence that pain significantly predicts smoking and relapse, associations between smoking history/trajectory and changes in pain over the course of treatment have not been characterized. The objective of the study was to determine the association between in-treatment smoking trajectory, pack-years (i.e., number of cigarette packs smoked per day multiplied by smoking duration), pain-related interference in daily activities, and pain intensity over the course of residential treatment. METHODS: In this study, 280 adult smokers in a residential SUD treatment center in North Central Florida completed questionnaires assessing cigarette use, pain intensity, and pain interference at treatment entry and discharge (Mean = 80.3 days, SD = 25.6). Most participants were diagnosed with alcohol use disorder (66.1 %). Opioid (27.9 %) and cannabis use disorders (29.6 %) were also common. Participants were grouped by whether their smoking increased (n = 36), decreased (n = 46), or stayed the same (n = 133) from entry to discharge. RESULTS: Analyses indicated a positive association between pack-years and pain intensity at both baseline (r = 0.185, p = 0.018) and discharge (r = 0.184, p = 0.019). Smoking trajectory was associated with pack-years, with those decreasing smoking having greater pack-years than those sustaining or increasing use [F(2,136) = 8.62, p < 0.01, η2p = 0.114]. Mixed general linear models indicated pain intensity [F(1,274) = 44.15, p < 0.0001, η2p = 0.138] and interference in day-to-day activities [F(1,276) = 31.79, p < 0.0001, η2p = 0.103] decreased significantly over time. However, there was no main effect of smoking trajectory on pain intensity [F(2,212) = 2.051, p = 0.131, η2p = 0.019] or of smoking trajectory by time interaction [F(2, 212) = 1.228, p = 0.295, η2p = 0.011]. CONCLUSIONS: Overall, findings provide evidence that smoking behavior influences pain within the context of residential substance use treatment. Given that pain is associated with urge to use substances and risk of return to use, more consistent and rigorous assessment of pain and proactive pain management is likely to enhance substance use treatment outcomes among people who smoke.


Subject(s)
Residential Treatment , Humans , Male , Female , Adult , Smoking/epidemiology , Smoking/adverse effects , Middle Aged , Substance-Related Disorders/epidemiology , Substance-Related Disorders/psychology , Chronic Pain/epidemiology , Chronic Pain/psychology , Opioid-Related Disorders/epidemiology , Opioid-Related Disorders/psychology , Alcoholism/epidemiology , Alcoholism/psychology , Substance Abuse Treatment Centers , Cigarette Smoking/epidemiology , Cigarette Smoking/adverse effects , Cigarette Smoking/therapy , Marijuana Abuse/epidemiology , Marijuana Abuse/psychology , Marijuana Abuse/complications , Pain/epidemiology , Pain/etiology
2.
Nutrients ; 16(3)2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38337717

ABSTRACT

Lung inflammation and alveolar enlargement are the major pathological conditions of chronic obstructive pulmonary disease (COPD) patients. Rice bran oil (RBO), a natural anti-inflammatory and antioxidative agent, has been used for therapeutic purposes in several inflammatory diseases. This study aimed to investigate the anti-inflammatory and antioxidative effect of RBO on a cigarette smoke extract (CSE)-induced emphysema model in mice. The results indicated that CSE significantly induced airspace enlargement in mouse lung. Increased inflammatory cells, macrophage, and TNF-alpha levels in bronchoalveolar lavage fluid (BALF) were noticed in CSE-treated mice. RBO (low and high dose)-supplemented mice showed decreased total BALF inflammatory cell, macrophage, and neutrophil numbers and TNF-alpha levels (p < 0.05). Additionally, the administration of RBO decreased the mean linear alveolar intercept (MLI) in the CSE-treated group. Additionally, RBO treatment significantly increased the total antioxidant capacity in both mouse BALF and serum. However, RBO did not have an effect on the malondialdehyde (MDA) level. These findings suggested that RBO treatment ameliorates lung inflammation in a CSE-induced emphysema mice model through anti-inflammatory and antioxidant pathways. Therefore, the supplementation of RBO could be a new potential therapeutic to relieve the severity of COPD.


Subject(s)
Cigarette Smoking , Emphysema , Pneumonia , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Mice , Animals , Antioxidants/metabolism , Lung/pathology , Rice Bran Oil/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Cigarette Smoking/adverse effects , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Anti-Inflammatory Agents/therapeutic use , Pneumonia/drug therapy , Bronchoalveolar Lavage Fluid , Emphysema/chemically induced , Emphysema/drug therapy , Tobacco Products
3.
Phytomedicine ; 124: 155256, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181527

ABSTRACT

BACKGROUND: Alveolar macrophages are one of the momentous regulators in pulmonary inflammatory responses, which can secrete extracellular vesicles (EVs) packing miRNAs. Ferroptosis, an iron-dependent cell death, is associated with cigarette smoke-induced lung injury, and EVs have been reported to regulate ferroptosis by transporting intracellular iron. However, the regulatory mechanism of alveolar macrophage-derived EVs has not been clearly illuminated in smoking-related pulmonary ferroptosis. Despite the known anti-ferroptosis effects of naringenin in lung injury, whether naringenin controls EVs-mediated ferroptosis has not yet been explored. PURPOSE: We explore the effects of EVs from cigarette smoke-stimulated alveolar macrophages in lung epithelial ferroptosis, and elucidate the EV miRNA-mediated pharmacological mechanism of naringenin. STUDY DESIGN AND METHODS: Differential and ultracentrifugation were conducted to extract EVs from different alveolar macrophages treatment groups in vitro. Both intratracheal instilled mice and treated epithelial cells were used to investigate the roles of EVs from alveolar macrophages involved in ferroptosis. Small RNA sequencing analysis was performed to distinguish altered miRNAs in EVs. The ferroptotic effects of EV miRNAs were examined by applying dual-Luciferase reporter assay and miRNA inhibitor transfection experiment. RESULTS: Here, we firstly reported that EVs from cigarette smoke extract-induced alveolar macrophages (CSE-EVs) provoked pulmonary epithelial ferroptosis. The ferroptosis inhibitor ferrostatin-1 treatment reversed these changes in vitro. Moreover, EVs from naringenin and CSE co-treated alveolar macrophages (CSE+Naringenin-EVs) markedly attenuated the lung epithelial ferroptosis compared with CSE-EVs. Notably, we identified miR-23a-3p as the most dramatically changed miRNA among Normal-EVs, CSE-EVs, and CSE+Naringenin-EVs. Further experimental investigation showed that ACSL4, a pro-ferroptotic gene leading to lipid peroxidation, was negatively regulated by miR-23a-3p. The inhibition of miR-23a-3p diminished the efficacy of CSE+Naringenin-EVs. CONCLUSION: Our findings firstly provided evidence that naringenin elevated the EV miR-23a-3p level from CSE-induced alveolar macrophages, thereby inhibiting the mouse lung epithelial ferroptosis via targeting ACSL4, and further complemented the mechanism of cigarette-induced lung injury and the protection of naringenin in a paracrine manner. The administration of miR-23a-3p-enriched EVs has the potential to ameliorate pulmonary ferroptosis.


Subject(s)
Cigarette Smoking , Extracellular Vesicles , Ferroptosis , Flavanones , Lung Injury , MicroRNAs , Mice , Animals , Macrophages, Alveolar/metabolism , Cigarette Smoking/adverse effects , Lung/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/metabolism , Iron/metabolism
4.
Int J Chron Obstruct Pulmon Dis ; 18: 1883-1897, 2023.
Article in English | MEDLINE | ID: mdl-37662486

ABSTRACT

Objective: Cigarette smoke exposure is one of the major risk factors for the development of chronic obstructive pulmonary disease (COPD). Ginseng saponin Rb1 (Rb1) is a natural extract from ginseng root with anti-inflammatory and anti-oxidant effects. However, the underlying mechanism of the Rb1 in COPD remains unknown. Therefore, we sought to explore the role of Rb1 in cigarette smoke-induced damage and to reveal the potential mechanism. Methods: The cell viability and lactose dehydrogenase (LDH) activity were analyzed using cell counting kit-8 (CCK-8) and LDH release assays. We further investigated the inflammation, apoptosis and oxidative stress markers and analyzed the nuclear factor-kappa B (NF-κB) and nuclear factor erythroid-2-related factor 2 (Nrf2) pathways in BEAS-2B cells and COPD rat model following cigarette smoke extract (CSE) exposure. Results: Our results showed that CSE promoted inflammation, apoptosis and oxidative stress in BEAS-2B cells. Rb1 suppressed the inflammatory response by inhibiting expression of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-1ß and inhibiting the NF-κB signaling pathway. Rb1 possessed the ability to hinder cell apoptosis induced by CSE. In addition, Rb1 concurrently reduced CSE-induced oxidative reactions and promoted Nrf2 translocation to nucleus. For in vivo study, Rb1 treatment alleviated CSE-induced lung injury, apoptosis, reactive oxygen species (ROS) release and inflammatory reactions. Also, Rb1 treatment activated Nrf2 signaling and inactivated NF-κB signaling in COPD rats. Conclusion: Rb1 attenuates CSE-induced inflammation, apoptosis and oxidative stress by suppressing NF-κB and activating Nrf2 signaling pathways, which provides novel insights into the mechanism underlying CSE-induced COPD.


Subject(s)
Cigarette Smoking , Panax , Pulmonary Disease, Chronic Obstructive , Animals , Rats , NF-kappa B , NF-E2-Related Factor 2 , Cigarette Smoking/adverse effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/etiology , Signal Transduction , Oxidative Stress , Inflammation/drug therapy , Inflammation/prevention & control , Apoptosis
5.
Brain Behav Immun ; 109: 292-307, 2023 03.
Article in English | MEDLINE | ID: mdl-36775074

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a major, incurable respiratory condition that is primarily caused by cigarette smoking (CS). Neurocognitive disorders including cognitive dysfunction, anxiety and depression are highly prevalent in people with COPD. It is understood that increased lung inflammation and oxidative stress from CS exposure may 'spill over' into the systemic circulation to promote the onset of these extra-pulmonary comorbidities, and thus impacts the quality of life of people with COPD. The precise role of the 'spill-over' of inflammation and oxidative stress in the onset of COPD-related neurocognitive disorders are unclear. The present study investigated the impact of chronic CS exposure on anxiety-like behaviors and social recognition memory, with a particular focus on the role of the 'spill-over' of inflammation and oxidative stress from the lungs. Adult male BALB/c mice were exposed to either room air (sham) or CS (9 cigarettes per day, 5 days a week) for 24 weeks and were either daily co-administered with the NOX2 inhibitor, apocynin (5 mg/kg, in 0.01 % DMSO diluted in saline, i.p.) or vehicle (0.01 % DMSO in saline) one hour before the initial CS exposure of the day. After 23 weeks, mice underwent behavioral testing and physiological diurnal rhythms were assessed by monitoring diurnal regulation profiles. Lungs were collected and assessed for hallmark features of COPD. Consistent with its anti-inflammatory and oxidative stress properties, apocynin treatment partially lessened lung inflammation and lung function decline in CS mice. CS-exposed mice displayed marked anxiety-like behavior and impairments in social recognition memory compared to sham mice, which was prevented by apocynin treatment. Apocynin was unable to restore the decreased Bmal1-positive cells, key in cells in diurnal regulation, in the suprachiasmatic nucleus of the hypothalamus to that of sham levels. CS-exposed mice treated with apocynin was associated with a restoration of microglial area per cell and basal serum corticosterone. This data suggests that we were able to model the CS-induced social recognition memory impairments seen in humans with COPD. The preventative effects of apocynin on memory impairments may be via a microglial dependent mechanism.


Subject(s)
Cigarette Smoking , Pneumonia , Pulmonary Disease, Chronic Obstructive , Humans , Adult , Male , Mice , Animals , Cigarette Smoking/adverse effects , Microglia , Dimethyl Sulfoxide/pharmacology , Quality of Life , Lung , Pneumonia/complications , Suprachiasmatic Nucleus , Hypothalamus , Inflammation/complications , Mice, Inbred C57BL
6.
Fertil Steril ; 119(5): 838-846, 2023 05.
Article in English | MEDLINE | ID: mdl-36716812

ABSTRACT

OBJECTIVE: To investigate cannabis smoking and tobacco cigarette smoking in relation to adenomyosis risk. DESIGN: We used data from a case-control study of adenomyosis conducted among enrollees ages 18-59 years of an integrated health care system in Washington State. The case-control study used 2 control groups given the challenge of selecting noncases when cases are diagnosed by hysterectomy. SUBJECTS: Cases (n = 386) were enrollees with incident, pathology-confirmed adenomyosis diagnosed between April 1, 2001, and March 31, 2006. The 2 control groups comprised hysterectomy controls (n = 233) with pathology-confirmed absence of adenomyosis and population controls (n = 323) with an intact uterus selected randomly from the health care system population and frequency matched to cases on age. EXPOSURE: Detailed data on cannabis and tobacco cigarette smoking history were ascertained through in-person structured interviews, allowing estimation of joint-years of cannabis smoking and pack-years of tobacco cigarette smoking. MAIN OUTCOME MEASURES: Odds ratios (ORs) and 95% confidence intervals (CIs) for the associations between cannabis smoking, tobacco cigarette smoking, and adenomyosis were estimated using multivariable unconditional logistic regression. Analyses were adjusted for age, reference year, menarche age, education, and pack-years of cigarette smoking (or joint-years of cannabis smoking). RESULTS: No association was observed between cannabis smoking history and adenomyosis risk. However, we did observe the suggestion of an association between ever tobacco cigarette smoking and adenomyosis risk, comparing cases to hysterectomy controls (OR, 1.3; 95% CI, 0.9-1.9) and population controls (OR, 1.2; 95% CI, 0.8-1.8). Our data suggested a 50% increased odds of adenomyosis with >15 pack-years of smoking (vs. never smoking), comparing cases to hysterectomy controls (OR, 1.5; 95% CI, 0.9-2.6; Ptrend=.135). The suggestion of a 40% increased adenomyosis odds was observed with smoking >5-15 pack-years (vs. never smoking), comparing cases to population controls (OR, 1.4; 95% CI, 0.8-2.4; Ptrend=0.136). CONCLUSION: In the first study of cannabis smoking and adenomyosis risk, no association was observed. However, our data suggested an increased odds of adenomyosis with history of tobacco cigarette smoking. Further research is warranted to replicate our results given the substantial morbidity with adenomyosis and frequency of cigarette smoking and recreational and medical cannabis use.


Subject(s)
Adenomyosis , Cannabis , Cigarette Smoking , Marijuana Smoking , Female , Humans , Adolescent , Young Adult , Adult , Middle Aged , Marijuana Smoking/adverse effects , Marijuana Smoking/epidemiology , Cigarette Smoking/adverse effects , Cigarette Smoking/epidemiology , Nicotiana , Case-Control Studies , Adenomyosis/diagnosis , Adenomyosis/epidemiology
7.
J Ethnopharmacol ; 301: 115758, 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36167232

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: As a traditional medicine, seeds of Ginkgo biloba L. (Gbs) have been used to treat cough or asthma for a long time. It is commonly used in clinic for lung diseases. However, its mechanism of lung protection is not completely clear. AIMS OF THE STUDY: This research was designed to explore the protective effects of Gbs on antioxidant and inflammation during the chronic obstructive pulmonary disease (COPD) pathological process provoked by cigarette smoking (CS) in rats. MATERIALS AND METHODS: Six random groups including control group, CS model group, Gbs intervention groups (25 mg/kg, 50 mg/kg, and 100 mg/kg) and aminophylline group were composed of forty-eight rats. Smoking and intratracheal instillation of lipopolysaccharide (LPS) were used to establish the COPD rat model. Glutathione peroxidase (GSH-PX), malondialdehyde (MDA), superoxide dismutase (SOD), and enzyme-linked immunosorbent assay (ELISA) was used for quantifying the inflammatory factors such as IL-8, IL-6, IL-10, IL-17 and TNF-α. Western blotting were used for detecting the protein expressions of Nrf2, Keap1 and HO-1 in the lung tissues. RESULTS: Gbs inhibits lung histological changes and decreased the inflammatory factors in both bronchoalveolar lavage fluid (BALF) and serum of CS-exposed rats, including IL-10, IL-17, IL-6, IL-8 and TNF-α. Gbs also inhibited the MDA level, increased SOD and GSH-PX activity in serum and changed expressions of Nrf2, Keap1 and HO-1 in the lung tissues. CONCLUSION: Gbs inhibit oxidative stress and inflammation induced by cigarette smoke in COPD rats through the Nrf2 Pathway.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Animals , Rats , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/metabolism , Cigarette Smoking/adverse effects , Ginkgo biloba , Inflammation/pathology , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Lung , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Pulmonary Disease, Chronic Obstructive/metabolism , Seeds/metabolism , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Int J Chron Obstruct Pulmon Dis ; 17: 2589-2602, 2022.
Article in English | MEDLINE | ID: mdl-36267325

ABSTRACT

Background: It has been reported that a disintegrin and metalloproteinase 9 (ADAM9) is involved in the pathogenesis of cigarette smoke (CS)-associated chronic obstructive pulmonary disease (COPD). But how CS exposure leads to upregulation of ADAM9 remains unknown. Methods: Patients who underwent lobectomy for a solitary pulmonary nodule were enrolled and divided into three groups: non-smokers with normal lung function, smokers without COPD and smoker patients with COPD. Immunoreactivity of interleukin (IL)-17A and ADAM9 in small airways and alveolar walls was measured by immunohistochemistry. Wild-type and Il17a -/- C57BL/6 mice were exposed to CS for six months, and ADAM9 expression in the airway epithelia was measured by immunoreactivity. In addition, the protein and mRNA expression levels of IL-17A and ADAM9 were assessed in CS extract (CSE) and/or IL-17A-treated human bronchial epithelial (HBE) cells. Results: The immunoreactivity of ADAM9 was increased in the airway epithelia and alveolar walls of patients with COPD compared to that of the controls. The expression of IL-17A was also upregulated in airway epithelial cells of patients with COPD and correlated positively with the level of ADAM9. The results from the animal model showed that Il17a -/- mice were protected from emphysema induced by CS exposure, together with a reduced level of ADAM9 expression in the airway epithelia, suggesting a possible link between ADAM9 and IL-17A. Consistently, our in vitro cell model showed that CSE stimulated the expression of ADAM9 and IL-17A in HBE cells in a dose- and time-dependent manner. Recombinant IL-17A induced ADAM9 upregulation in HBE cells and had a synergistic effect with CSE, whereas blocking IL-17A inhibited CSE-induced ADAM9 expression. Further analysis revealed that IL-17A induced c-Jun N-terminal kinase (JNK) phosphorylation, thereby increasing ADAM9 expression. Conclusion: Our results revealed a novel role of IL-17A in CS-related COPD, where IL-17A contributes to ADAM9 expression by activating JNK signaling.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Humans , Mice , Animals , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Interleukin-17/genetics , Disintegrins/metabolism , Cigarette Smoking/adverse effects , Mice, Inbred C57BL , Nicotiana , Epithelial Cells/metabolism , RNA, Messenger/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Plant Extracts , Membrane Proteins/genetics , Membrane Proteins/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism
9.
Eur Respir Rev ; 31(164)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35705209

ABSTRACT

COPD is predicted to become the third leading cause of morbidity and mortality worldwide by 2030. Cigarette smoking (active or passive) is one of its chief causes, with about 20% of cigarette smokers developing COPD from cigarette smoke (CS)-induced irreversible damage and sustained inflammation of the airway epithelium. Inflammasome activation leads to the cleavage of pro-interleukin (IL)-1ß and pro-IL-18, along with the release of pro-inflammatory cytokines via gasdermin D N-terminal fragment membrane pores, which further triggers acute phase pro-inflammatory responses and concurrent pyroptosis. There is currently intense interest in the role of nucleotide-binding oligomerisation domain-like receptor family, pyrin domain containing protein-3 inflammasomes in chronic inflammatory lung diseases such as COPD and their potential for therapeutic targeting. Phytochemicals including polyphenols and flavonoids have phyto-medicinal benefits in CS-COPD. Here, we review published articles from the last decade regarding the known associations between inflammasome-mediated responses and ameliorations in pre-clinical manifestations of CS-COPD via polyphenol and flavonoid treatment, with a focus on the underlying mechanistic insights. This article will potentially assist the development of drugs for the prevention and therapy of COPD, particularly in cigarette smokers.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Cigarette Smoking/adverse effects , Flavonoids/therapeutic use , Humans , Inflammasomes , Inflammation , Polyphenols , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/etiology
10.
Can Respir J ; 2022: 8509396, 2022.
Article in English | MEDLINE | ID: mdl-35465190

ABSTRACT

Berberine (Ber) is an isoquinoline alkaloid that has shown therapeutic potential in mice with chronic obstructive pulmonary disease (COPD). However, the therapeutic efficiency of Ber is restricted by its low aqueous solubility and bioavailability. Chitosan and solid lipid nanoparticles (SLNs) have demonstrated great abilities as delivery systems in enhancing the bioavailability of therapeutic compounds. The present study aimed to get together the biological features of SLNs with the advantages of chitosan to formulate an efficient nano-carrier platform for the oral delivery of Ber and evaluate the therapeutic effect of the prepared Ber-encapsulated nanoparticles on airway inflammation in cigarette smoke (CS)-induced COPD rats. The Ber-encapsulated SLE-chitosan formulation was manufactured using a modified solvent-injection method followed by a homogenization process. Physicochemical properties, encapsulation efficiency, in vitro stability and Ber release, and pharmacokinetics of the manufactured formulation were evaluated. The COPD rat model was developed by exposing animals to CS. To study the therapeutic efficiency of Ber-encapsulated SLE-chitosan nanoparticles and pure berberine, the histopathological changes of the lung tissues, levels of inflammatory cells and cytokines, and activities of myeloperoxidase (MPO) and superoxide dismutase (SOD) enzymes were evaluated in bronchoalveolar lavage fluid (BALF). Ber-encapsulated SLE-chitosan showed the particle size in nano-range with high stability and controlled slow-release profile in vitro in simulated gastric (pH 1.5) and intestinal (pH 6.8) fluids. Administration of Ber-loaded SLE-chitosan nanoparticles could significantly ameliorate inflammation scores in lung tissues and reduce levels of inflammatory cells (neutrophils and macrophages) and inflammatory cytokines (IL-1ß, Il-6, Il-17, and TNFα) in BALF when compared with the pure Ber. SLE-chitosan-based nanoparticles can strongly improve the therapeutic anti-inflammatory impact of Ber against CS-induced airway inflammation in COPD rats, suggesting the promising application of Ber-encapsulated SLN-chitosan nanoparticles for treating COPD and other inflammation-mediated diseases.


Subject(s)
Berberine , Chitosan , Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Animals , Humans , Mice , Rats , Berberine/pharmacology , Berberine/therapeutic use , Chitosan/chemistry , Chitosan/therapeutic use , Cigarette Smoking/adverse effects , Cytokines , Inflammation/drug therapy , Lipids/chemistry , Lipids/therapeutic use , Liposomes , Nanoparticles , Pulmonary Disease, Chronic Obstructive/drug therapy
11.
Respir Res ; 23(1): 40, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236342

ABSTRACT

BACKGROUND: In chronic obstructive pulmonary disease (COPD), exacerbations cause acute inflammatory flare-ups and increase the risk for hospitalization and mortality. Exacerbations are common in all disease stages and are often caused by bacterial infections e.g., non-typeable Heamophilus influenzae (NTHi). Accumulating evidence also associates vitamin D deficiency with the severity of COPD and exacerbation frequency. However, it is still unclear whether vitamin D deficiency when combined with cigarette smoking would worsen and prolong exacerbations caused by repeated infections with the same bacterial strain. METHODS: Vitamin D sufficient (VDS) and deficient (VDD) mice were exposed to nose-only cigarette smoke (CS) for 14 weeks and oropharyngeally instilled with NTHi at week 6, 10 and 14. Three days after the last instillation, mice were assessed for lung function, tissue remodeling, inflammation and immunity. The impact of VDD and CS on inflammatory cells and immunoglobulin (Ig) production was also assessed in non-infected animals while serum Ig production against NTHi and dsDNA was measured in COPD patients before and 1 year after supplementation with Vitamin D3. RESULTS: VDD enhanced NTHi eradication, independently of CS and complete eradication was reflected by decreased anti-NTHi Ig's within the lung. In addition, VDD led to an increase in total lung capacity (TLC), lung compliance (Cchord), MMP12/TIMP1 ratio with a rise in serum Ig titers and anti-dsDNA Ig's. Interestingly, in non-infected animals, VDD exacerbated the CS-induced anti-NTHi Ig's, anti-dsDNA Ig's and inflammatory cells within the lung. In COPD patients, serum Ig production was not affected by vitamin D status but anti-NTHi IgG increased after vitamin D3 supplementation in patients who were Vitamin D insufficient before treatment. CONCLUSION: During repeated infections, VDD facilitated NTHi eradication and resolution of local lung inflammation through production of anti-NTHi Ig, independently of CS whilst it also promoted autoantibodies. In COPD patients, vitamin D supplementation could be protective against NTHi infections in vitamin D insufficient patients. Future research is needed to decipher the determinants of dual effects of VDD on adaptive immunity. TRAIL REGISTRATION: ClinicalTrials, NCT00666367. Registered 23 April 2008, https://www.clinicaltrials.gov/ct2/show/study/NCT00666367 .


Subject(s)
Cigarette Smoking/adverse effects , Haemophilus Infections/complications , Haemophilus influenzae/immunology , Lung/microbiology , Pneumonia/complications , Vitamin D Deficiency/metabolism , Animals , Disease Models, Animal , Haemophilus Infections/metabolism , Haemophilus Infections/microbiology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pneumonia/metabolism
12.
Asian Pac J Cancer Prev ; 22(10): 3385-3391, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34711016

ABSTRACT

BACKGROUND: Bladder cancer disproportionally affects the communities. While it is the ninth most common cancer in the world, in some parts of Iran including Kerman province it is the most common cancer among men. This study aimed to determine potential risk factors of bladder cancer in Kerman province, Iran. METHODS: During February to July 2020, in this matched hospital-based case-control study, 100 patients with bladder cancer and 200 healthy individuals (matched in age and sex) were recruited. Socio-demographics status, occupational exposures, common diet, history of drug use and family history of cancer, were collected using a structured questionnaire. Bivariable and multivariable logistic regression were applied and crude and adjusted odds ratios (AOR) along with their 95% confidence intervals (95%CI) were calculated. Data were analyzed using Stata version 14 software. RESULTS: Opium consumption, cigarette smoking and low level of income were associated with increased chance of bladder cancer. Compared to never use, use of opium up to 18000 Gram -year was associated with increased chance of bladder cancer (AOR: 6; 95% CI =2.3, 15.5). The chance was higher among those who used opium more than 18,000 Gram - year (AOR: 11.3; 95% CI =2.3, 15.5). In comparison with never smokers, the chance of bladder cancer increased among those who smoked up to 20 pack-year cigarette) (AOR: 3.4; 95%CI= 1.3, 8.9) and those who smoke ≥ 20 pack-year (AOR: 15.8; 95% CI= 5.9, 42.4). CONCLUSIONS: The observed strong dose-response association between opium consumption, cigarette smoking and bladder cancer highlights the need for extension of harm reduction programs especially in regions with high burden of disease.


Subject(s)
Cigarette Smoking/adverse effects , Narcotics/adverse effects , Opium/adverse effects , Urinary Bladder Neoplasms/etiology , Adult , Aged , Case-Control Studies , Confidence Intervals , Dose-Response Relationship, Drug , Environmental Exposure/adverse effects , Female , Humans , Income , Iran , Logistic Models , Male , Middle Aged , Narcotics/administration & dosage , Non-Smokers , Odds Ratio , Opium/administration & dosage , Risk Factors
13.
Microbiol Spectr ; 9(2): e0013821, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34523990

ABSTRACT

Cigarettes and opium contain chemicals and particulate matter that may modify the oral microbiota. This study aimed to investigate the association between cigarette and opium use with the oral microbiota. A total of 558 participants were recruited from Iran between 2011 and 2015. Individuals were categorized as never cigarette nor opium users, ever cigarette-only smokers, ever opium-only users, and ever both cigarette and opium users. Participants provided saliva samples for 16S rRNA gene sequencing. Logistic regression, microbiome regression-based kernel association test (MiRKAT), and zero-inflated beta regression models were calculated. For every increase in 10 observed amplicon sequence variants (ASVs), the odds for being a cigarette-only smoker, opium-only user, and both user compared to never users decreased by 9% (odds ratio [OR] = 0.91; 95% confidence interval [95% CI] = 0.86 to 0.97), 13% (OR = 0.87; 95% CI = 0.75 to 1.01), and 12% (OR = 0.88; 95% CI = 0.80 to 0.96), respectively. The microbial communities differed by cigarette and opium use as indicated by MiRKAT models testing the three beta-diversity matrices (P < 0.05 for all). Three genera were less likely and one genus was more likely to be detected in cigarette-only smokers or opium-only users than in never users. The relative abundance of the phylum Actinobacteria (never, 14.78%; both, 21.20%) was higher and the phyla Bacteroidetes (never, 17.63%; both, 11.62%) and Proteobacteria (never, 9.06%; both, 3.70%) were lower in users of both cigarettes and opium, while the phylum Firmicutes (never, 54.29%; opium, 65.49%) was higher in opium-only users. Cigarette and opium use was associated with lower alpha-diversity, overall oral microbiota community composition, and both the presence and relative abundance of multiple taxa. IMPORTANCE Cigarette smoking and opium use are associated with periodontal disease caused by specific bacteria such as Porphyromonas gingivalis, which suggests a link between cigarette smoking and opium use and the oral microbiota. Alterations of the oral microbiota in cigarette smokers compared to nonsmokers have been reported, but this has not been studied across diverse populations. Additionally, the association of opium use with the oral microbiota has not been investigated to date. We conducted this study to investigate differences in the oral microbiota between ever users of cigarettes only, opium only, and both cigarettes and opium and never users of cigarettes and opium in Iran. Lower alpha-diversity, distinct overall oral microbial communities, and the presence and relative abundance of multiple taxa have been found for users of cigarettes and/or opium.


Subject(s)
Bacteria/classification , Cigarette Smoking/adverse effects , Microbiota/drug effects , Mouth/microbiology , Opium Dependence/epidemiology , Aged , Aged, 80 and over , Bacteria/genetics , Bacteria/isolation & purification , Biodiversity , Female , Humans , Iran/epidemiology , Male , Middle Aged , Opium/adverse effects , Periodontal Diseases/microbiology
14.
Int Immunopharmacol ; 96: 107593, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33819731

ABSTRACT

BACKGROUND: Antioxidant and anti-inflammatory effects are two main pharmacological mechanisms of pirfenidone (PFD) besides the anti-fibrotic effect. This study aims to investigate whether PFD could mediate cigarette smoke extract (CSE) induced inflammation and oxidative stress in vitro and in vivo. METHODS: BALB/C mice and alveolar epithelial (A549) cells treated with CSE were established as disease models in vivo and in vitro. Effects of PFD treatment on disease models were further measured. Hematoxylin and eosin (HE) staining was used to evaluate the pathological changes in lung tissues of mice. CCK-8 assay kit was applied to measure the viability of A549 cells treated by different concentrations of PFD. Inflammation cytokine expression in cell supernatants was measured with ELISA kits. The mRNA and protein levels of inflammation and oxidative stress-related factors were determined by real-time quantitative polymerase chain reaction analysis (RT-qPCR) and Western blotting. Furthermore, myeloperoxidase (MPO), malondialdehyde (MDA), and total antioxidant capacity (T-AOC) were measured to detect the antioxidative activity of lung tissues. Moreover, an assay kit with fluorescent probe 2',7'-dichlorofluorescin diacetate (DCFH-DA) was used to evaluate the intracellular reactive oxygen species (ROS) generation. RESULTS: In vitro and in vivo, PFD significantly reversed TNF-α, IL-6, CCL2, SOD1, and CAT mRNA level changes led by CSE; in addition, PFD significantly decreased the ratios of p-p65 to p65, p-ikBα to ikBα and increased Nrf-2 protein level compared with CSE group. In mice, high-dose (100 mg/kg/d) PFD significantly reversed MPO and MDA increases induced by CSE. However, PFD didn't significantly reverse T-AOC decrease induced by CSE. In A549 cell supernatant, PFD dramatically reversed the elevated levels of TNF-α and IL-1ß induced by CSE. Furthermore, PFD could significantly reverse the increased level of ROS induced by CSE in A549 cells. CONCLUSION: Our study reveals the potential role of PFD in regulating inflammatory response and oxidative stress induced by CSE.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cigarette Smoking/adverse effects , Inflammation/drug therapy , Lung/pathology , Pyridones/therapeutic use , Respiratory Mucosa/drug effects , A549 Cells , Animals , Cytokines/metabolism , Humans , Inflammation/chemically induced , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred BALB C , Oxidative Stress , Plant Extracts/administration & dosage , Reactive Oxygen Species/metabolism , Respiratory Mucosa/physiology , Signal Transduction
15.
Biochem Biophys Res Commun ; 555: 1-6, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33812052

ABSTRACT

Cigarette smoke (CS) contains many toxins that collectively harm nearly every organ in the body, and smoking is a key risk factor for many chronic diseases. Aside from its toxic actions, CS may alter expression of the drug- and steroid-binding pregnane X receptor (PXR), which when activated upregulates expression of cytochrome P450 (CYP) enzymes, glutathione transferases (GSTs), and multidrug resistance protein 1 (MDR1), an adaptive metabolic array that mediates clearance of CS component toxins. We sought to identify new PXR agonists that may be useful for restoring PXR activity in conditions wherein it is suppressed, and their mechanisms of PXR binding and activation. PXR has a uniquely larger, hydrophobic, and highly flexible ligand-binding domain (LBD) vs. other nuclear receptors, enabling it to interact with structurally diverse molecules. We tested certain calcium channel blockers (CCBs) as a pharmacological subset of potential PXR ligands, analyzing by molecular docking methods, and identified a putative active site in the PXR LBD, along with the relevant bonds and bonding energies. We analyzed felodipine binding and agonist activity in detail, as it showed the lowest binding energy among CCBs tested. We found felodipine was a potent PXR agonist as measured by luciferase reporter assay, whereas CCBs with higher binding energies were less potent (amlodipine) or nearly inactive (manidipine), and it induced CYP3A4 expression in HepG2 cells, a known target of PXR agonism. Felodipine also both induced PXR mRNA in HepG2 hepatocytes and reduced CS extract-induced diminution of PXR levels, indicating it modulates PXR expression. The results illuminate mechanisms of ligand-induced PXR activation and identify felodipine as a novel PXR agonist.


Subject(s)
Calcium Channel Blockers/pharmacology , Cigarette Smoking/adverse effects , Felodipine/pharmacology , Pregnane X Receptor/agonists , Pregnane X Receptor/metabolism , Binding Sites , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/metabolism , Computer Simulation , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inducers/pharmacology , Down-Regulation/drug effects , Drug Evaluation, Preclinical/methods , Felodipine/chemistry , Felodipine/metabolism , Hep G2 Cells , Humans , Ligands , Molecular Docking Simulation , Pregnane X Receptor/chemistry
16.
Int J Mol Sci ; 22(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33916310

ABSTRACT

Epidemiological evidence shows that smoking causes a thrombophilic milieu that may play a role in the pathophysiology of chronic obstructive pulmonary disease (COPD) as well as pulmonary thromboembolism. The increased nicotine level induces a prothrombotic status and abnormal blood coagulation in smokers. Since several anticoagulants increase bleeding risk, alternative therapies need to be identified to protect against thrombosis without affecting hemostasis. Astragalin is a flavonoid present in persimmon leaves and green tea seeds and exhibits diverse activities of antioxidant and anti-inflammation. The current study investigated that astragalin attenuated smoking-induced pulmonary thrombosis and alveolar inflammation. In addition, it was explored that molecular links between thrombosis and inflammation entailed protease-activated receptor (PAR) activation and oxidative stress-responsive mitogen-activated protein kinase (MAPK)-signaling. BALB/c mice were orally administrated with 10-20 mg/kg astragalin and exposed to cigarette smoke for 8 weeks. For the in vitro study, 10 U/mL thrombin was added to alveolar epithelial A549 cells in the presence of 1-20 µM astragalin. The cigarette smoking-induced the expression of PAR-1 and PAR-2 in lung tissues, which was attenuated by the administration of ≥10 mg/kg astragalin. The oral supplementation of ≥10 mg/kg astragalin to cigarette smoke-challenged mice attenuated the protein induction of urokinase plasminogen activator, plasminogen activator inhibitor-1and tissue factor, and instead enhanced the induction of tissue plasminogen activator in lung tissues. The astragalin treatment alleviated cigarette smoke-induced lung emphysema and pulmonary thrombosis. Astragalin caused lymphocytosis and neutrophilia in bronchoalveolar lavage fluid due to cigarette smoke but curtailed infiltration of neutrophils and macrophages in airways. Furthermore, this compound retarded thrombin-induced activation of PAR proteins and expression of inflammatory mediators in alveolar cells. Treating astragalin interrupted PAR proteins-activated reactive oxygen species production and MAPK signaling leading to alveolar inflammation. Accordingly, astragalin may interrupt the smoking-induced oxidative stress-MAPK signaling-inflammation axis via disconnection between alveolar PAR activation and pulmonary thromboembolism.


Subject(s)
Kaempferols/therapeutic use , MAP Kinase Signaling System/drug effects , Pulmonary Embolism/prevention & control , Pulmonary Emphysema/prevention & control , Receptors, Proteinase-Activated/antagonists & inhibitors , Animals , Cigarette Smoking/adverse effects , Drug Evaluation, Preclinical , Kaempferols/pharmacology , Male , Mice, Inbred BALB C , Oxidative Stress , Pulmonary Embolism/etiology
17.
Inflammation ; 44(4): 1643-1661, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33730343

ABSTRACT

The present study was aimed to investigate the phototherapy effect with low-level laser on human bronchial epithelial cells activated by cigarette smoke extract (CSE). Phototherapy has been reported to actuate positively for controlling the generation/release of anti-inflammatory and pro-inflammatory mediators from different cellular type activated by distinct stimuli. It is not known whether the IL-8 and IL-10 release from CSE-stimulated human bronchial epithelium (BEAS) cells can be influenced by phototherapy. Human bronchial epithelial cell (BEAS) line was cultured in a medium with CSE and irradiated (660 nm) at 9 J. Apoptosis index was standardized with Annexin V and the cellular viability was evaluated by MTT. IL-8, IL-10, cAMP, and NF-κB were measured by ELISA as well as the Sp1, JNK, ERK1/2, and p38MAPK. Phototherapy effect was studied in the presence of mithramycin or the inhibitors of JNK or ERK. The IL-8, cAMP, NF-κB, JNK, p38, and ERK1/2 were downregulated by phototherapy. Both the JNK and the ERK inhibitors potentiated the phototherapy effect on IL-8 as well as on cAMP secretion from BEAS. On the contrary, IL-10 and Sp1 were upregulated by phototherapy. The mithramycin blocked the phototherapy effect on IL-10. The results suggest that phototherapy has a dual effect on BEAS cells because it downregulates the IL-8 secretion by interfering with CSE-mediated signaling pathways, and oppositely upregulates the IL-10 secretion through of Sp1 transcription factor. The manuscript provides evidence that the phototherapy can interfere with MAPK signaling via cAMP in order to attenuate the IL-8 secretion from CSE-stimulated BEAS. In addition, the present study showed that phototherapy effect is driven to downregulation of the both the IL-8 and the ROS secretion and at the same time the upregulation of IL-10 secretion. Besides it, the increase of Sp-1 transcription factor was crucial for laser effect in upregulating the IL-10 secretion. The dexamethasone corticoid produces a significant inhibitory effect on IL-8 as well as ROS secretion, but on the other hand, the corticoid blocked the IL-10 secretion. Taking it into consideration, it is reasonable to suggest that the beneficial effect of laser therapy on lung diseases involves its action on unbalance between pro-inflammatory and anti-inflammatory mediators secreted by human bronchial epithelial cells through different signaling pathway.


Subject(s)
Cytokines/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Nicotiana/adverse effects , Phototherapy/methods , Respiratory Mucosa/metabolism , Smoke/adverse effects , Sp1 Transcription Factor/metabolism , Bronchi/drug effects , Bronchi/metabolism , Cell Line , Cigarette Smoking/adverse effects , Cigarette Smoking/therapy , Humans , Respiratory Mucosa/drug effects
18.
Sci Rep ; 11(1): 7008, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33772084

ABSTRACT

Dietary fiber functions as a prebiotic to determine the gut microbe composition. The gut microbiota influences the metabolic functions and immune responses in human health. The gut microbiota and metabolites produced by various dietary components not only modulate immunity but also impact various organs. Although recent findings have suggested that microbial dysbiosis is associated with several respiratory diseases, including asthma, cystic fibrosis, and allergy, the role of microbiota and metabolites produced by dietary nutrients with respect to pulmonary disease remains unclear. Therefore, we explored whether the gut microbiota and metabolites produced by dietary fiber components could influence a cigarette smoking (CS)-exposed emphysema model. In this study, it was demonstrated that a high-fiber diet including non-fermentable cellulose and fermentable pectin attenuated the pathological changes associated with emphysema progression and the inflammatory response in CS-exposed emphysema mice. Moreover, we observed that different types of dietary fiber could modulate the diversity of gut microbiota and differentially impacted anabolism including the generation of short-chain fatty acids, bile acids, and sphingolipids. Overall, the results of this study indicate that high-fiber diets play a beneficial role in the gut microbiota-metabolite modulation and substantially affect CS-exposed emphysema mice. Furthermore, this study suggests the therapeutic potential of gut microbiota and metabolites from a high-fiber diet in emphysema via local and systemic inflammation inhibition, which may be useful in the development of a new COPD treatment plan.


Subject(s)
Dietary Fiber/pharmacology , Emphysema/diet therapy , Emphysema/prevention & control , Gastrointestinal Microbiome/physiology , Prebiotics/administration & dosage , Animals , Bile Acids and Salts/biosynthesis , Cellulose/pharmacology , Cigarette Smoking/adverse effects , Diet , Dysbiosis/prevention & control , Fatty Acids, Volatile/biosynthesis , Female , Inflammation/diet therapy , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Pectins/pharmacology , Sphingolipids/biosynthesis
19.
Hum Exp Toxicol ; 40(9): 1445-1462, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33686898

ABSTRACT

Smoking is one of the most important leading death cause worldwide. From a toxicological perspective, cigarette smoke serves hazards especially for the human being exposed to passive smoke. Over the last decades, the effects of natural compounds on smoking-mediated respiratory diseases such as COPD, asthma, and lung cancer have been under investigation, as well as the mechanistic aspects of disease progression. In the present study, the protective mechanism of eucalyptol (EUC), curcumin (CUR), and their combination on BEAS-2B cells were investigated in vitro to understand their impact on cell death, oxidative cell injury, and inflammatory response induced by 3R4F reference cigarette extract (CSE). According to the present findings, EUC, CUR, and their combination improved cell viability, attenuated CSE-induced apoptosis, and LC3B expression. Further, CSE-induced oxidative damage and inflammatory response in human bronchial epithelial cells were remarkably reduced by the combination treatment through modification of enzymatic antioxidant activity, GSH, MDA, and intracellular ROS levels as well as nitrite and IL-6 levels. In addition, nuclear translocation of Nrf2, a regulatory protein involved in the indirect antioxidant response, was remarkably up-regulated with the combination pre-treatment. In conclusion, EUC and CUR in combination might be a potential therapeutic against smoking-induced lung diseases through antioxidant and inflammatory pathways and results represent valuable background for future in vivo pulmonary toxicity studies.


Subject(s)
Bronchi/drug effects , Cigarette Smoking/adverse effects , Curcumin/therapeutic use , Epithelial Cells/drug effects , Eucalyptol/therapeutic use , Lung Diseases/chemically induced , Lung Diseases/drug therapy , Plant Extracts/toxicity , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Survival/drug effects , Cells, Cultured/drug effects , Humans , Plant Extracts/chemistry , Protective Agents/therapeutic use , Nicotiana/chemistry
20.
J Nutr Biochem ; 91: 108596, 2021 05.
Article in English | MEDLINE | ID: mdl-33548472

ABSTRACT

Cigarette smoke (CS) is an independent risk factor in development of nonalcoholic steatohepatitis (NASH) and fibrosis. Lycopene, a carotenoid naturally occurring in tomatoes, has been shown to be a protective agent against tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced NASH. In the present study using a ferret model we investigated whether CS promotes NASH and whether dietary lycopene can inhibit CS-promoted NASH development, and if so, what potential mechanisms were involved. Ferrets were divided into 4 groups (n=12-16/group): control, NNK/CS exposed, NNK/CS plus low-dose lycopene (2.2 mg/kg BW/day), and NNK/CS plus high-dose lycopene (6.6 mg/kg BW/day) groups, for 26 weeks. Results showed that hepatic steatosis, infiltrates of inflammatory cells, and the number and size of inflammatory foci in liver, together with key genes involved in hepatic fibrogenesis were higher in the NNK/CS group compared to the control group; a lycopene diet reversed these changes to the levels of the control group. Interestingly, a major lycopene cleavage enzyme, beta-carotene 9',10'-oxygenase (BCO2), which recently has been recognized to play metabolic roles beyond cleavage function, was down-regulated by NNK/CS exposure, but this decrease was prevented by lycopene feeding. NNK/CS exposure also downregulated liver expression of antioxidant enzymes and upregulated oxidative stress marker, which were all prevented by lycopene. In conclusion, our results suggest that CS can promote development of NASH and liver fibrosis in ferrets, which is associated with downregulation of BCO2 and impairment of antioxidant system in liver; dietary lycopene may inhibit CS-promoted NASH by preventing suppression of BCO2 and decline in antioxidant network.


Subject(s)
Antioxidants/therapeutic use , Cigarette Smoking/adverse effects , Dietary Supplements , Lycopene/therapeutic use , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/therapy , Animals , Ferrets , Male , Non-alcoholic Fatty Liver Disease/enzymology , Oxidative Stress
SELECTION OF CITATIONS
SEARCH DETAIL