51
|
He H, Tang H, Gao L, Wu Y, Feng Z, Lin H, Wu T. Tanshinone IIA attenuates bleomycin-induced pulmonary fibrosis in rats. Mol Med Rep 2015; 11:4190-6. [PMID: 25672255 PMCID: PMC4394983 DOI: 10.3892/mmr.2015.3333] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 12/19/2014] [Indexed: 11/06/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a chronic and progressive fibrotic lung disorder with unknown etiology and a high mortality rate. Tanshinone IIA (Tan IIA) is a lipophilic diterpene extracted from the Chinese herb Salvia miltiorrhiza Bunge with diverse biological functions. The present study was conducted to evaluate the effects of Tan IIA on bleomycin (BLM)-induced pulmonary fibrosis in rats. Rats received an intraperitoneal injection of Tan IIA and normal rats were used as controls. Severe pulmonary edema, inflammation and fibrosis were observed in the BLM-treated rats and the counts of total cells, neutrophils and lymphocytes were significantly increased in the bronchoalveolar lavage fluids of those rats. These pathological changes were markedly attenuated by subsequent treatment with Tan IIA. In addition, BLM-induced increased expression of tumor necrosis factor-α, interleukin (IL)-1β, IL-6, cyclooxygenase-2, prostaglandin E2, malondialdehyde, inducible nitric oxide synthase and nitric oxide in rats, which was also suppressed by Tan IIA injection. The present findings suggest therapeutic potential of Tan IIA for pulmonary fibrosis.
Collapse
Affiliation(s)
- Huanyu He
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Haiying Tang
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lili Gao
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yun Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Zhiqiang Feng
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Hongli Lin
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Taihua Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
52
|
Li X, Ye Y, Zhou X, Huang C, Wu M. Atg7 enhances host defense against infection via downregulation of superoxide but upregulation of nitric oxide. THE JOURNAL OF IMMUNOLOGY 2014; 194:1112-21. [PMID: 25535282 DOI: 10.4049/jimmunol.1401958] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium that can cause serious infection in immunocompromised individuals. Although autophagy may augment immune responses against P. aeruginosa infection in macrophages, the critical components and their role of autophagy in host defense are largely unknown. In this study, we show that P. aeruginosa infection-induced autophagy activates JAK2/STAT1α and increases NO production. Knocking down Atg7 resulted in increased IFN-γ release, excessive reactive oxygen species, and increased Src homology-2 domain-containing phosphatase 2 activity, which led to lowered phosphorylation of JAK2/STAT1α and subdued expression of NO synthase 2 (NOS2). In addition, we demonstrated the physiological relevance of dysregulated NO under Atg7 deficiency as atg7(-/-) mice were more susceptible to P. aeruginosa infection with increased mortality and severe lung injury than wild-type mice. Furthermore, P. aeruginosa-infected atg7(-/-) mice exhibited increased oxidation but decreased bacterial clearance in the lung and other organs compared with wild-type mice. Mechanistically, atg7 deficiency suppressed NOS2 activity by downmodulating JAK2/STAT1α, leading to decreased NO both in vitro and in vivo. Taken together, these findings revealed that the JAK2/STAT1α/NOS2 dysfunction leads to dysregulated immune responses and worsened disease phenotypes.
Collapse
Affiliation(s)
- Xuefeng Li
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yan Ye
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and
| | - Xikun Zhou
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Canhua Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Min Wu
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and
| |
Collapse
|
53
|
Lee MY, Sun KH, Chiang CP, Huang CF, Sun GH, Tsou YC, Liu HY, Tang SJ. Nitric oxide suppresses LPS-induced inflammation in a mouse asthma model by attenuating the interaction of IKK and Hsp90. Exp Biol Med (Maywood) 2014; 240:498-507. [PMID: 25519430 DOI: 10.1177/1535370214554880] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/16/2014] [Indexed: 02/01/2023] Open
Abstract
A feature of allergic airway disease is the observed increase of nitric oxide (NO) in exhaled breath. Gram-negative bacterial infections have also been linked with asthma exacerbations. However, the role of NO in asthma exacerbations with gram-negative bacterial infections is still unclear. In this study, we examined the role of NO in lipopolysaccharide (LPS)-induced inflammation in an ovalbumin (OVA)-challenged mouse asthma model. To determine whether NO affected the LPS-induced response, a NO donor (S-nitroso-N-acetylpenicillamine, SNAP) or a selective inhibitor of NO synthase (1400W) was injected intraperitoneally into the mice before the LPS stimulation. Decreased levels of proinflammatory cytokines were demonstrated in the bronchoalveolar lavage fluid from mice treated with SNAP, whereas increased levels of cytokines were found in the 1400W-treated mice. To further explore the molecular mechanism of NO-mediated inhibition of proinflammatory responses in macrophages, RAW 264.7 cells were treated with 1400W or SNAP before LPS stimulation. LPS-induced inflammation in the cells was attenuated by the presence of NO. The LPS-induced IκB kinase (IKK) activation and the expression of IKK were reduced by NO through attenuation of the interaction between Hsp90 and IKK in the cells. The IKK decrease in the lung immunohistopathology was verified in SNAP-treated asthma mice, whereas IKK increased in the 1400W-treated group. We report for the first time that NO attenuates the interaction between Hsp90 and IKK, decreasing the stability of IKK and causing the down-regulation of the proinflammatory response. Furthermore, the results suggest that NO may repress LPS-stimulated innate immunity to promote pulmonary bacterial infection in asthma patients.
Collapse
Affiliation(s)
- Ming-Yung Lee
- Institute of Bioscience and Biotechnology, Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan, Republic of China Department of Pediatrics, Tri-service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Kuang-Hui Sun
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan, Republic of China Department of Education and Research, Taipei City Hospital, Taipei 100, Taiwan
| | - Chien-Ping Chiang
- Department of Dermatology, Tri-service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Ching-Feng Huang
- Department of Pediatrics, Tri-service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Guang-Huan Sun
- Division of Urology, Department of Surgery, Tri-service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Yu-Chi Tsou
- Institute of Bioscience and Biotechnology, Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan, Republic of China
| | - Huan-Yun Liu
- Division of Urology, Department of Surgery, Taoyuan Armed Forces General Hospital 32551, Taiwan, Republic of China
| | - Shye-Jye Tang
- Institute of Bioscience and Biotechnology, Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan, Republic of China
| |
Collapse
|
54
|
Yao J, Pan D, Zhao Y, Zhao L, Sun J, Wang Y, You QD, Xi T, Guo QL, Lu N. Wogonin prevents lipopolysaccharide-induced acute lung injury and inflammation in mice via peroxisome proliferator-activated receptor gamma-mediated attenuation of the nuclear factor-kappaB pathway. Immunology 2014; 143:241-57. [PMID: 24766487 DOI: 10.1111/imm.12305] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury (ALI) from a variety of clinical disorders, characterized by diffuse inflammation, is a cause of acute respiratory failure that develops in patients of all ages. Previous studies reported that wogonin, a flavonoid-like chemical compound which was found in Scutellaria baicalensis, has anti-inflammatory effects in several inflammation models, but not in ALI. Here, the in vivo protective effect of wogonin in the amelioration of lipopolysaccharide (LPS) -induced lung injury and inflammation was assessed. In addition, the in vitro effects and mechanisms of wogonin were studied in the mouse macrophage cell lines Ana-1 and RAW264.7. In vivo results indicated that wogonin attenuated LPS-induced histological alterations. Peripheral blood leucocytes decreased in the LPS-induced group, which was ameliorated by wogonin. In addition, wogonin inhibited the production of several inflammatory cytokines, including tumour necrosis factor-α, interleukin-1β (IL-1β) and IL-6, in the bronchoalveolar lavage fluid and lung tissues after LPS challenge, while the peroxisome proliferator-activated receptor γ (PPARγ) inhibitor GW9662 reversed these effects. In vitro results indicated that wogonin significantly decreased the secretion of IL-6, IL-1β and tumour necrosis factor-α in Ana-1 and RAW264.7 cells, which was suppressed by transfection of PPARγ small interfering RNA and GW9662 treatment. Moreover, wogonin activated PPARγ, induced PPARγ-mediated attenuation of the nuclear translocation and the DNA-binding activity of nuclear factor-κB in vivo and in vitro. In conclusion, all of these results showed that wogonin may serve as a promising agent for the attenuation of ALI-associated inflammation and pathology by regulating the PPARγ-involved nuclear factor-κB pathway.
Collapse
Affiliation(s)
- Jing Yao
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Abstract
In the previous study, the anti-inflammatory effect of p-cymene had been found. In this study, we investigated anti-inflammatory effects of p-cymene on acute lung injury using lipopolysaccharide (LPS)-induced acute lung injury (ALI) mouse model. The cell counting in the bronchoalveolar lavage fluid (BALF) was measured. The animal lung edema degree was evaluated by wet/dry weight (W/D) ratio. The superoxidase dismutase (SOD) activity and myeloperoxidase (MPO) activity was assayed by SOD and MPO kits, respectively. The levels of inflammatory mediators including tumor necrosis factor alpha (TNF-α), IL-1β, and IL-6 were assayed by enzyme-linked immunosorbent assay method. The pathological changes of the lung tissues were observed by hematoxylin and eosin staining. The inflammatory signal pathway-related protein levels of NF-κB were measured using Western blotting. The data showed that treatment with the p-cymene markedly attenuated inflammatory cell numbers in the BALF, decreased NF-κB protein level in the lungs, improved SOD activity, and inhibited MPO activity. Histological studies demonstrated that p-cymene substantially inhibited LPS-induced neutrophils in the lung tissue compared with the model group. The results indicated that p-cymene had a protective effect on LPS-induced ALI in mice.
Collapse
|
56
|
Wu F, Szczepaniak WS, Shiva S, Liu H, Wang Y, Wang L, Wang Y, Kelley EE, Chen AF, Gladwin MT, McVerry BJ. Nox2-dependent glutathionylation of endothelial NOS leads to uncoupled superoxide production and endothelial barrier dysfunction in acute lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 307:L987-97. [PMID: 25326583 DOI: 10.1152/ajplung.00063.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microvascular barrier integrity is dependent on bioavailable nitric oxide (NO) produced locally by endothelial NO synthase (eNOS). Under conditions of limited substrate or cofactor availability or by enzymatic modification, eNOS may become uncoupled, producing superoxide in lieu of NO. This study was designed to investigate how eNOS-dependent superoxide production contributes to endothelial barrier dysfunction in inflammatory lung injury and its regulation. C57BL/6J mice were challenged with intratracheal LPS. Bronchoalveolar lavage fluid was analyzed for protein accumulation, and lung tissue homogenate was assayed for endothelial NOS content and function. Human lung microvascular endothelial cell (HLMVEC) monolayers were exposed to LPS in vitro, and barrier integrity and superoxide production were measured. Biopterin species were quantified, and coimmunoprecipitation (Co-IP) assays were performed to identify protein interactions with eNOS that putatively drive uncoupling. Mice exposed to LPS demonstrated eNOS-dependent increased alveolar permeability without evidence for altered canonical NO signaling. LPS-induced superoxide production and permeability in HLMVEC were inhibited by the NOS inhibitor nitro-l-arginine methyl ester, eNOS-targeted siRNA, the eNOS cofactor tetrahydrobiopterin, and superoxide dismutase. Co-IP indicated that LPS stimulated the association of eNOS with NADPH oxidase 2 (Nox2), which correlated with augmented eNOS S-glutathionylation both in vitro and in vivo. In vitro, Nox2-specific inhibition prevented LPS-induced eNOS modification and increases in both superoxide production and permeability. These data indicate that eNOS uncoupling contributes to superoxide production and barrier dysfunction in the lung microvasculature after exposure to LPS. Furthermore, the results implicate Nox2-mediated eNOS-S-glutathionylation as a mechanism underlying LPS-induced eNOS uncoupling in the lung microvasculature.
Collapse
Affiliation(s)
- Feng Wu
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - William S Szczepaniak
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Pharmacology, Pittsburgh, Pennsylvania
| | - Huanbo Liu
- University of Pittsburgh School of Medicine Department of Surgery, Pittsburgh, Pennsylvania
| | - Yinna Wang
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Ling Wang
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Ying Wang
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - Eric E Kelley
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Anesthesiology, Pittsburgh, Pennsylvania
| | - Alex F Chen
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Surgery, Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania; University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Bryan J McVerry
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania; University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania;
| |
Collapse
|
57
|
Kanter M, Sahin SH, Basaran UN, Ayvaz S, Aksu B, Erboga M, Colak A. The effect of methylene blue treatment on aspiration pneumonia. J Surg Res 2014; 193:909-19. [PMID: 25260956 DOI: 10.1016/j.jss.2014.08.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 08/15/2014] [Accepted: 08/22/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND The study aimed to examine whether methylene blue (MB) prevents different pulmonary aspiration materials-induced lung injury in rats. METHODS The experiments were designed in 60 Sprague-Dawley rats, ranging in weight from 250-300 g, randomly allotted into one of six groups (n = 10): saline control, Biosorb Energy Plus (BIO), hydrochloric acid (HCl), saline + MB treated, BIO + MB treated, and HCl + MB treated. Saline, BIO, and HCl were injected into the lungs in a volume of 2 mL/kg. After surgical procedure, MB was administered intraperitoneally for 7 days at a daily dose of 2 mg/kg per day. Seven days later, rats were killed, and both lungs in all groups were examined biochemically and histopathologically. RESULTS Our findings show that MB inhibits the inflammatory response reducing significantly (P < 0.05) peribronchial inflammatory cell infiltration, alveolar septal infiltration, alveolar edema, alveolar exudate, alveolar histiocytes, interstitial fibrosis, granuloma, and necrosis formation in different pulmonary aspiration models. Pulmonary aspiration significantly increased the tissue hydroxyproline content, malondialdehyde levels, and decreased (P < 0.05) the antioxidant enzyme (superoxide dismutase and glutathione peroxidase) activities. MB treatment significantly (P < 0.05) decreased the elevated tissue hydroxyproline content and malondialdehyde levels and prevented the inhibition of superoxide dismutase and glutathione peroxidase (P < 0.05) enzymes in the tissues. Furthermore, there is a significant reduction in the activity of inducible nitric oxide synthase (iNOS), terminal deoxynucleotidyl transferase dUTP nick end labeling, and arise in the expression of surfactant protein D in lung tissue of different pulmonary aspiration models with MB therapy. CONCLUSIONS MB treatment might be beneficial in lung injury and therefore shows potential for clinical use.
Collapse
Affiliation(s)
- Mehmet Kanter
- Department of Histology and Embryology, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey.
| | - Sevtap Hekimoglu Sahin
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Umit Nusret Basaran
- Department of Pediatric Surgery, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Suleyman Ayvaz
- Department of Pediatric Surgery, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Burhan Aksu
- Department of Pediatric Surgery, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Mustafa Erboga
- Department of Histology and Embryology, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - Alkin Colak
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
58
|
Keles S, Ates O, Kartal B, Alp HH, Ekinci M, Ceylan E, Ondas O, Arpali E, Dogan S, Yildirim K, Keles MS. Evaluation of cardiovascular biomarkers in patients with age-related wet macular degeneration. Clin Ophthalmol 2014; 8:1573-8. [PMID: 25210424 PMCID: PMC4154890 DOI: 10.2147/opth.s66160] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
AIM To evaluate levels of homocysteine, asymmetric dimethylarginine (ADMA), and nitric oxide (NO), as well as activity of endothelial NO synthase (eNOS), in patients with age-related macular degeneration (AMD). METHODS The levels of homocysteine, ADMA, and NO and activity of eNOS in patients who were diagnosed with wet AMD by fundus fluorescein angiography (n=30) were compared to a control group with no retinal pathology (n=30). RESULTS Levels of homocysteine and ADMA were found to be significantly higher in the wet AMD group than in the control group (P<0.001), whereas NO levels and eNOS activity were higher in the control group (P<0.001). In the wet AMD group, we detected a 2.64- and 0.33-fold increase in the levels of ADMA and homocysteine, respectively, and a 0.49- and 2.41-fold decrease in the eNOS activity and NO level, respectively. CONCLUSION Elevated levels of homocysteine and ADMA were observed in patients with wet AMD. Increased ADMA may be responsible for the diminished eNOS activity found in these patients, which in turn contributes to the decrease in NO levels, which likely plays a role in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Sadullah Keles
- Department of Ophthalmology, School of Medicine, Ataturk University, Erzurum, Turkey
| | - Orhan Ates
- Department of Ophthalmology, School of Medicine, Ataturk University, Erzurum, Turkey
| | - Baki Kartal
- Department of Ophthalmology, Regional Training and Research Hospital, Erzurum, Turkey
| | - Hamit Hakan Alp
- Department of Biochemistry, School of Medicine, Yuzuncu Yil University, Van, Turkey
| | - Metin Ekinci
- Department of Ophthalmology, School of Medicine, Kafkas University, Kars, Turkey
| | - Erdinc Ceylan
- Department of Ophthalmology, Regional Training and Research Hospital, Erzurum, Turkey
| | - Osman Ondas
- Department of Ophthalmology, Erbaa Government Hospital, Tokat, Turkey
| | - Eren Arpali
- Department of Ophthalmology, Regional Training and Research Hospital, Erzurum, Turkey
| | - Semih Dogan
- Department of Ophthalmology, Kolan Hospital, Istanbul, Turkey
| | - Kenan Yildirim
- Department of Ophthalmology, Igdır Government Hospital, Igdır, Turkey
| | - Mevlut Sait Keles
- Department of Biochemistry, School of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
59
|
Liu T, Schneider RA, Lee NY, Hoyt DG. Peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1) regulates pulmonary effects of endotoxin and tumor necrosis factor-α in mice. Biochem Biophys Res Commun 2014; 452:468-72. [PMID: 25159840 DOI: 10.1016/j.bbrc.2014.08.089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 08/19/2014] [Indexed: 01/13/2023]
Abstract
Peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1) modulates phospho-signaling by catalyzing rotation of the bond between a phosphorylated serine or threonine before proline in proteins. As depletion of PIN1 increased inflammatory protein expression in cultured endothelial cells treated with bacterial endotoxin (lipopolysaccharide, LPS) and interferon-γ, we hypothesized that PIN1 knockout would increase sensitivity to LPS-induced lung inflammation in mice. Mortality due to a high dose of LPS (30mg/kg) was greater in knockout than wildtype mice. Lung myeloperoxidase activity, reflecting neutrophils, was increased to a 35% higher level in PIN1 knockout mouse lung, as compared with wildtype, after treatment with a sublethal dose of 3mgLPS/kg, ip. Unexpectedly, plasma tumor necrosis factor-α (TNF) was approximately 50% less than in wildtype mice. Knockout mice, however, were more sensitive than wildtype to TNF-induced neutrophil accumulation. The neutrophil adhesion molecule, E-selectin, was also elevated in lungs of knockout mice treated with TNF, suggesting that PIN1 depletion increases endothelial sensitivity to TNF. Indeed, TNF induced more reactive oxygen species in cultured endothelial cells depleted of PIN1 with short hairpin RNA than in control cells. Collectively, the results indicate that while PIN1 normally facilitates TNF production in LPS-treated mice, it suppresses pulmonary and endothelial reactions to the cytokine. Tissue or cell-specific effects of PIN1 may affect the overall inflammatory response to LPS and other stimuli.
Collapse
Affiliation(s)
- Tongzheng Liu
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Ryan A Schneider
- College of Pharmacy, The University of Findlay, Findlay, OH 45840, USA.
| | - Nam Y Lee
- Division of Pharmacology, The Ohio State University College of Pharmacy and The Dorothy M. Davis Heart and Lung Research Institute, Columbus, OH 43210, USA.
| | - Dale G Hoyt
- Division of Pharmacology, The Ohio State University College of Pharmacy and The Dorothy M. Davis Heart and Lung Research Institute, Columbus, OH 43210, USA.
| |
Collapse
|
60
|
Cohen MD, Vaughan JM, Garrett B, Prophete C, Horton L, Sisco M, Kodavanti UP, Ward WO, Peltier RE, Zelikoff J, Chen LC. Acute high-level exposure to WTC particles alters expression of genes associated with oxidative stress and immune function in the lung. J Immunotoxicol 2014; 12:140-53. [PMID: 24911330 DOI: 10.3109/1547691x.2014.914609] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
First responders (FR) present at Ground Zero in the first 72 h after the World Trade Center (WTC) collapsed have progressively exhibited significant respiratory injuries. The few toxicology studies performed to date evaluated effects from just fine (< 2.5 µm) WTC dusts; none examined health effects/toxicities from atmospheres bearing larger particle sizes, despite the fact the majority (> 96%) of dusts were > 10 µm and most FR likely entrained dusts by mouth breathing. Using a system that generated/delivered supercoarse (10-53 µm) WTC dusts to F344 rats (in a manner that mimicked FR exposures), this study sought to examine potential toxicities in the lungs. In this exploratory study, rats were exposed for 2 h to 100 mg WTC dust/m(3) (while under isoflurane [ISO] anesthesia) or an air/ISO mixture; this dose conservatively modeled likely exposures by mouth-breathing FR facing ≈750-1000 mg WTC dust/m(3). Lungs were harvested 2 h post-exposure and total RNA extracted for subsequent global gene expression analysis. Among the > 1000 genes affected by WTC dust (under ISO) or ISO alone, 166 were unique to the dust exposure. In many instances, genes maximally-induced by the WTC dust exposure (relative to in naïve rats) were unchanged/inhibited by ISO only; similarly, several genes maximally inhibited in WTC dust rats were largely induced/unchanged in rats that received ISO only. These outcomes reflect likely contrasting effects of ISO and the WTC dust on lung gene expression. Overall, the data show that lungs of rats exposed to WTC dust (under ISO) - after accounting for any impact from ISO alone - displayed increased expression of genes related to lung inflammation, oxidative stress, and cell cycle control, while several involved in anti-oxidant function were inhibited. These changes suggested acute inflammogenic effects and oxidative stress in the lungs of WTC dust-exposed rats. This study, thus, concludes that a single very high exposure to WTC dusts could potentially have adversely affected the respiratory system - in terms of early inflammatory and oxidative stress processes. As these changes were not compared with other types of dusts, the uniqueness of these WTC-mediated effects remains to be confirmed. It also still remains to be determined if these effects might have any relevance to chronic lung pathologies that became evident among FR who encountered the highest dust levels on September 11, 2001 and the 2 days thereafter. Ongoing studies using longer-range post-exposure analyses (up to 1-year or more) will help to determine if effects seen here on genes were acute, reversible, or persistent, and associated with corresponding histopathologic/biochemical changes in situ.
Collapse
Affiliation(s)
- Mitchell D Cohen
- Department of Environmental Medicine, New York University School of Medicine , NY , USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Tianzhu Z, Shihai Y, Juan D. The Effects of Morin on Lipopolysaccharide-Induced Acute Lung Injury by Suppressing the Lung NLRP3 Inflammasome. Inflammation 2014; 37:1976-83. [DOI: 10.1007/s10753-014-9930-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
62
|
Messias-Reason IJT, van Tong H, Velavan TP. Analysis of polymorphic sites in the promoter of the nitric oxide synthase 2 gene in Brazilian patients with leprosy. Int J Immunogenet 2014; 41:231-5. [PMID: 24495190 DOI: 10.1111/iji.12108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 11/26/2013] [Accepted: 12/19/2013] [Indexed: 12/01/2022]
Abstract
Leprosy is one of the most neglected infectious tropical diseases of the skin and the nerves caused by the intracellular pathogen Mycobacterium leprae. The inducible NOS isoform encoded by NOS2A plays a vital role in host defence against bacterial infections. The functional promoter polymorphisms in NOS2A are associated with various autoimmune and infectious diseases. We investigated the association of NOS2A variants with progression of leprosy in a Brazilian cohort including 221 clinically classified patients and 103 unrelated healthy controls. We observed a novel variant ss528838018A/G in the promoter region at position -6558. The other functional variants were observed with low frequency of minor allele (<0.005). NOS2A promoter variant (-954G/C) was not observed in Brazilian populations, and the new observed promoter variant (ss528838018A/G) as well as other promoter variants were not associated with any clinical forms of leprosy in the Brazilian populations.
Collapse
Affiliation(s)
- I J T Messias-Reason
- Laboratório de Imunopatologia Molecular, Departamento de Patologia Médica, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, Brazil
| | | | | |
Collapse
|
63
|
Abstract
Pulmonary hypertension in human patients can result from increased pulmonary vascular tone, pressure transferred from the systemic circulation, dropout of small pulmonary vessels, occlusion of vessels with thrombi or intimal lesions, or some combination of all of these. Different animal models have been designed to reflect these different mechanistic origins of disease. Pulmonary hypertension models may be roughly grouped into tone-related models, inflammation-related models, and genetic models with unusual or mixed mechanism. Models of tone generally use hypoxia as a base, and then modify this with either genetic modifications (SOD, NOS, and caveolin) or with drugs (Sugen), although some genetic modifications of tone-related pathways can result in spontaneous pulmonary hypertension (Hph-1). Inflammation-related models can use either toxic chemicals (monocrotaline, bleomycin), live pathogens (stachybotrys, schistosomiasis), or genetic modifications (IL-6, VIP). Additional genetic models rely on alterations in metabolism (adiponectin), cell migration (S100A4), the serotonin pathway, or the BMP pathway. While each of these shares molecular and pathologic symptoms with different classes of human pulmonary hypertension, in most cases the molecular etiology of human pulmonary hypertension is unknown, and so the relationship between any model and human disease is unclear. There is thus no best animal model of pulmonary hypertension; instead, investigators must select the model most related to the specific pathology they are studying.
Collapse
Affiliation(s)
- James West
- Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | | |
Collapse
|
64
|
Kar S, Kavdia M. Endothelial NO and O₂·⁻ production rates differentially regulate oxidative, nitroxidative, and nitrosative stress in the microcirculation. Free Radic Biol Med 2013; 63:161-74. [PMID: 23639567 PMCID: PMC4051226 DOI: 10.1016/j.freeradbiomed.2013.04.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 04/04/2013] [Accepted: 04/13/2013] [Indexed: 02/07/2023]
Abstract
Endothelial dysfunction causes an imbalance in endothelial NO and O₂·⁻ production rates and increased peroxynitrite formation. Peroxynitrite and its decomposition products cause multiple deleterious effects including tyrosine nitration of proteins, superoxide dismutase (SOD) inactivation, and tissue damage. Studies have shown that peroxynitrite formation during endothelial dysfunction is strongly dependent on the NO and O₂·⁻ production rates. Previous experimental and modeling studies examining the role of NO and O₂·⁻ production imbalance on peroxynitrite formation showed different results in biological and synthetic systems. However, there is a lack of quantitative information about the formation and biological relevance of peroxynitrite under oxidative, nitroxidative, and nitrosative stress conditions in the microcirculation. We developed a computational biotransport model to examine the role of endothelial NO and O₂·⁻ production on the complex biochemical NO and O₂·⁻ interactions in the microcirculation. We also modeled the effect of variability in SOD expression and activity during oxidative stress. The results showed that peroxynitrite concentration increased with increase in either O₂·⁻ to NO or NO to O₂·⁻ production rate ratio (QO₂·⁻/QNO or QNO/QO₂·⁻, respectively). The peroxynitrite concentrations were similar for both production rate ratios, indicating that peroxynitrite-related nitroxidative and nitrosative stresses may be similar in endothelial dysfunction or inducible NO synthase (iNOS)-induced NO production. The endothelial peroxynitrite concentration increased with increase in both QO₂·⁻/QNO and QNO/QO₂·⁻ ratios at SOD concentrations of 0.1-100 μM. The absence of SOD may not mitigate the extent of peroxynitrite-mediated toxicity, as we predicted an insignificant increase in peroxynitrite levels beyond QO₂·⁻/QNO and QNO/QO₂·⁻ ratios of 1. The results support the experimental observations of biological systems and show that peroxynitrite formation increases with increase in either NO or O₂·⁻ production, and excess NO production from iNOS or from NO donors during oxidative stress conditions does not reduce the extent of peroxynitrite mediated toxicity.
Collapse
Affiliation(s)
- Saptarshi Kar
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA.
| | | |
Collapse
|
65
|
Zhong WT, Wu YC, Xie XX, Zhou X, Wei MM, Soromou LW, Ci XX, Wang DC. Phillyrin attenuates LPS-induced pulmonary inflammation via suppression of MAPK and NF-κB activation in acute lung injury mice. Fitoterapia 2013; 90:132-9. [DOI: 10.1016/j.fitote.2013.06.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/29/2013] [Accepted: 06/02/2013] [Indexed: 12/17/2022]
|
66
|
Badiei A, Rivers-Auty J, Ang AD, Bhatia M. Inhibition of hydrogen sulfide production by gene silencing attenuates inflammatory activity of LPS-activated RAW264.7 cells. Appl Microbiol Biotechnol 2013; 97:7845-7852. [PMID: 23838794 DOI: 10.1007/s00253-013-5080-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 12/11/2022]
Abstract
Hydrogen sulfide is an inflammatory mediator and is produced by the activity of the enzyme cystathionine γ-lyase (CSE) in macrophages. Previously, pharmacological inhibition of CSE has been reported to have conflicting results, and this may be due to the lack of specificity of the pharmacological agents. Therefore, this study used a very specific approach of small interfering RNA (siRNA) to inhibit the production of the CSE in an in vitro setting. We found that the activation of macrophages by lipopolysaccharide (LPS) resulted in higher levels of CSE mRNA and protein as well as the increased production of proinflammatory cytokines and nitric oxide (NO). We successfully used siRNA to specifically reduce the levels of CSE mRNA and protein in activated macrophages. Furthermore, the levels of proinflammatory cytokines in LPS-activated macrophages were significantly lower in siRNA-transfected cells compared to those in untransfected controls. However, the production levels of NO by the transfected cells were higher, suggesting that CSE activity has an inhibitory effect on NO production. These findings suggest that the CSE enzyme has a crucial role in the activation of macrophages, and its activity has an inhibitory effect on NO production by these cells.
Collapse
Affiliation(s)
- Alireza Badiei
- Department of Pathology, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand
| | | | | | | |
Collapse
|
67
|
Lucas R, Czikora I, Sridhar S, Zemskov EA, Oseghale A, Circo S, Cederbaum SD, Chakraborty T, Fulton DJ, Caldwell RW, Romero MJ. Arginase 1: an unexpected mediator of pulmonary capillary barrier dysfunction in models of acute lung injury. Front Immunol 2013; 4:228. [PMID: 23966993 PMCID: PMC3736115 DOI: 10.3389/fimmu.2013.00228] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 07/19/2013] [Indexed: 12/31/2022] Open
Abstract
The integrity of epithelial and endothelial barriers in the lower airspaces of the lungs has to be tightly regulated, in order to prevent leakage and to assure efficient gas exchange between the alveoli and capillaries. Both G− and G+ bacterial toxins, such as lipopolysaccharide and pneumolysin, respectively, can be released in high concentrations within the pulmonary compartments upon antibiotic treatment of patients suffering from acute respiratory distress syndrome (ARDS) or severe pneumonia. These toxins are able to impair endothelial barrier function, either directly, or indirectly, by induction of pro-inflammatory mediators and neutrophil sequestration. Toxin-induced endothelial hyperpermeability can involve myosin light chain phosphorylation and/or microtubule rearrangement. Endothelial nitric oxide synthase (eNOS) was proposed to be a guardian of basal barrier function, since eNOS knock-out mice display an impaired expression of inter-endothelial junction proteins and as such an increased vascular permeability, as compared to wild type mice. The enzyme arginase, the activity of which can be regulated by the redox status of the cell, exists in two isoforms – arginase 1 (cytosolic) and arginase 2 (mitochondrial) – both of which can be expressed in lung microvascular endothelial cells. Upon activation, arginase competes with eNOS for the substrate l-arginine, as such impairing eNOS-dependent NO generation and promoting reactive oxygen species generation by the enzyme. This mini-review will discuss recent findings regarding the interaction between bacterial toxins and arginase during acute lung injury and will as such address the role of arginase in bacterial toxin-induced pulmonary endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA ; Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA ; Division of Pulmonary Medicine, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Mini-review: novel therapeutic strategies to blunt actions of pneumolysin in the lungs. Toxins (Basel) 2013; 5:1244-60. [PMID: 23860351 PMCID: PMC3737495 DOI: 10.3390/toxins5071244] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 06/25/2013] [Accepted: 06/27/2013] [Indexed: 01/11/2023] Open
Abstract
Severe pneumonia is the main single cause of death worldwide in children under five years of age. The main etiological agent of pneumonia is the G+ bacterium Streptococcus pneumoniae, which accounts for up to 45% of all cases. Intriguingly, patients can still die days after commencing antibiotic treatment due to the development of permeability edema, although the pathogen was successfully cleared from their lungs. This condition is characterized by a dramatically impaired alveolar epithelial-capillary barrier function and a dysfunction of the sodium transporters required for edema reabsorption, including the apically expressed epithelial sodium channel (ENaC) and the basolaterally expressed sodium potassium pump (Na+-K+-ATPase). The main agent inducing this edema formation is the virulence factor pneumolysin, a cholesterol-binding pore-forming toxin, released in the alveolar compartment of the lungs when pneumococci are being lysed by antibiotic treatment or upon autolysis. Sub-lytic concentrations of pneumolysin can cause endothelial barrier dysfunction and can impair ENaC-mediated sodium uptake in type II alveolar epithelial cells. These events significantly contribute to the formation of permeability edema, for which currently no standard therapy is available. This review focuses on discussing some recent developments in the search for the novel therapeutic agents able to improve lung function despite the presence of pore-forming toxins. Such treatments could reduce the potentially lethal complications occurring after antibiotic treatment of patients with severe pneumonia.
Collapse
|
69
|
Zhong WT, Jiang LX, Wei JY, Qiao AN, Wei MM, Soromou LW, Xie XX, Zhou X, Ci XX, Wang DC. Protective effect of esculentoside A on lipopolysaccharide-induced acute lung injury in mice. J Surg Res 2013; 185:364-72. [PMID: 23764313 DOI: 10.1016/j.jss.2013.05.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 04/25/2013] [Accepted: 05/03/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Esculentoside A (EsA) is a saponin isolated from the Chinese herb Phytolacca esculenta. In our study, we sought to investigate the protective effects of EsA on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. MATERIALS AND METHODS To determine the effects of EsA on the reduction of histopathologic changes in mice with ALI, inflammatory cell count in bronchoalveolar lavage fluid (BALF) and lung wet-to-dry weight ratio were measured in LPS-challenged mice, and lung histopathologic changes observed via paraffin section were assessed. Next, cytokine production induced by LPS in BALF was measured by enzyme-linked immunosorbent assay. To further study the mechanism of EsA protective effects on ALI, IκBa, p38, and extracellular signal receptor-activated kinase pathways were investigated in lung tissue of mice with ALI. RESULTS In the present investigation, EsA showed marked effects by reducing inflammatory infiltration, thickening of the alveolar wall, and pulmonary congestion. Levels of tumor necrosis factor α and interleukin 6 elevated by LPS were significantly decreased in BALF in EsA-pretreated ALI model. Furthermore, EsA significantly suppressed phosphorylation of IκBa, p38, and extracellular signal receptor-activated kinase. CONCLUSIONS Taken together, our results suggest that EsA suppressed inflammatory responses in LPS-induced ALI through inhibition of the nuclear factor kappa B and mitogen activated protein kinase signaling pathways. EsA may be a promising potential preventive agent for ALI treatment.
Collapse
Affiliation(s)
- Wei-ting Zhong
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Abstract
Genetically modified mouse models have unparalleled power to determine the mechanisms behind different processes involved in the molecular and physiologic etiology of various classes of human pulmonary hypertension (PH). Processes known to be involved in PH for which there are extensive mouse models available include the following: (1) Regulation of vascular tone through secreted vasoactive factors; (2) regulation of vascular tone through potassium and calcium channels; (3) regulation of vascular remodeling through alteration in metabolic processes, either through alteration in substrate usage or through circulating factors; (4) spontaneous vascular remodeling either before or after development of elevated pulmonary pressures; and (5) models in which changes in tone and remodeling are primarily driven by inflammation. PH development in mice is of necessity faster and with different physiologic ramifications than found in human disease, and so mice make poor models of natural history of PH. However, transgenic mouse models are a perfect tool for studying the processes involved in pulmonary vascular function and disease, and can effectively be used to test interventions designed against particular molecular pathways and processes involved in disease.
Collapse
Affiliation(s)
- Mita Das
- Department of Internal Medicine, University of Arkansas Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | |
Collapse
|
71
|
Gao M, Chen L, Yu H, Sun Q, Kou J, Yu B. Diosgenin down-regulates NF-κB p65/p50 and p38MAPK pathways and attenuates acute lung injury induced by lipopolysaccharide in mice. Int Immunopharmacol 2012; 15:240-5. [PMID: 23246979 DOI: 10.1016/j.intimp.2012.11.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 11/25/2012] [Accepted: 11/30/2012] [Indexed: 01/16/2023]
Abstract
Diosgenin (Dio), a major active component of steroidal sapogenin of the traditional Chinese herb Dioscorea zingiberensis C.H.Wright, shows various activities including anti-inflammatory, anti-thrombotic activities, anti-cancer properties etc. In the present study, we found that diosgenin significantly suppressed the phosphorylation of lung NF-κB p50/p65 and MAPK/p38 in lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice, when given orally at doses of 0.1, 1.0 and 10mg/kg 1h prior to LPS challenge (30 mg/kg, intravenous injection). Moreover, diosgenin attenuated the lung histopathological changes such as pulmonary edema, coagulation and infiltration of inflammatory cells. In addition, diosgenin significantly decreased the lung wet to dry weight (W/D) ratio and nitrite/nitrate content at three doses, and also markedly inhibited LPS-induced body temperature decrease and nitrite/nitrate elevation in plasma. Besides, diosgenin could significantly suppress activation of NF-κB p65/p50, p38 and expression of inducible nitric oxide synthase (iNOS) in LPS-induced THP-1 cells. Our findings indicate the potential application of diosgenin for ALI treatment.
Collapse
Affiliation(s)
- Mengyu Gao
- Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, PR China
| | | | | | | | | | | |
Collapse
|
72
|
Percário S, Moreira DR, Gomes BAQ, Ferreira MES, Gonçalves ACM, Laurindo PSOC, Vilhena TC, Dolabela MF, Green MD. Oxidative stress in malaria. Int J Mol Sci 2012; 13:16346-72. [PMID: 23208374 PMCID: PMC3546694 DOI: 10.3390/ijms131216346] [Citation(s) in RCA: 191] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 11/08/2012] [Accepted: 11/23/2012] [Indexed: 12/16/2022] Open
Abstract
Malaria is a significant public health problem in more than 100 countries and causes an estimated 200 million new infections every year. Despite the significant effort to eradicate this dangerous disease, lack of complete knowledge of its physiopathology compromises the success in this enterprise. In this paper we review oxidative stress mechanisms involved in the disease and discuss the potential benefits of antioxidant supplementation as an adjuvant antimalarial strategy.
Collapse
Affiliation(s)
- Sandro Percário
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Para (LAPEO/ICB/UFPA) Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mails: (D.R.M.); (B.A.Q.G.); (M.E.S.F.); (A.C.M.G.); (P.S.O.C.L.); (T.C.V.)
| | - Danilo R. Moreira
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Para (LAPEO/ICB/UFPA) Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mails: (D.R.M.); (B.A.Q.G.); (M.E.S.F.); (A.C.M.G.); (P.S.O.C.L.); (T.C.V.)
| | - Bruno A. Q. Gomes
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Para (LAPEO/ICB/UFPA) Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mails: (D.R.M.); (B.A.Q.G.); (M.E.S.F.); (A.C.M.G.); (P.S.O.C.L.); (T.C.V.)
| | - Michelli E. S. Ferreira
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Para (LAPEO/ICB/UFPA) Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mails: (D.R.M.); (B.A.Q.G.); (M.E.S.F.); (A.C.M.G.); (P.S.O.C.L.); (T.C.V.)
| | - Ana Carolina M. Gonçalves
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Para (LAPEO/ICB/UFPA) Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mails: (D.R.M.); (B.A.Q.G.); (M.E.S.F.); (A.C.M.G.); (P.S.O.C.L.); (T.C.V.)
| | - Paula S. O. C. Laurindo
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Para (LAPEO/ICB/UFPA) Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mails: (D.R.M.); (B.A.Q.G.); (M.E.S.F.); (A.C.M.G.); (P.S.O.C.L.); (T.C.V.)
| | - Thyago C. Vilhena
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Para (LAPEO/ICB/UFPA) Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mails: (D.R.M.); (B.A.Q.G.); (M.E.S.F.); (A.C.M.G.); (P.S.O.C.L.); (T.C.V.)
| | - Maria F. Dolabela
- Pharmacy Faculty, Institute of Health Sciences, Federal University of Para. Av. Augusto Correa, 1, Guama, Belem, Para 66075-110, Brazil; E-Mail:
| | - Michael D. Green
- US Centers for Disease Control and Prevention, 1600 Clifton Road NE, mailstop G49, Atlanta, GA 30329, USA; E-Mail:
| |
Collapse
|
73
|
Tseng TL, Chen MF, Tsai MJ, Hsu YH, Chen CP, Lee TJF. Oroxylin-A rescues LPS-induced acute lung injury via regulation of NF-κB signaling pathway in rodents. PLoS One 2012; 7:e47403. [PMID: 23071799 PMCID: PMC3468516 DOI: 10.1371/journal.pone.0047403] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 09/14/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Successful drug treatment for sepsis-related acute lung injury (ALI) remains a major clinical problem. This study was designed to assess the beneficial effects of post-treatment of oroxylin A (OroA), a flavonoid, in ameliorating lipopolysaccharides (LPS)-induced lung inflammation and fatality. EXPERIMENTAL APPROACH Rats were injected with LPS (10 mg/kg, iv) to induce ALI, and OroA was given (15 mg/kg, iv) 1 hr or 6 hrs after LPS challenge. Twenty four hrs after LPS challenge, biochemical changes in the blood and lung tissues, and morphological/histological alterations in the lung associated with inflammation and injury were examined. Therapeutic effect of OroA was assessed by measuring the survival rate in endotoxemic mice. KEY RESULTS LPS (10 mg/kg, iv) significantly altered WBC counts, elevated plasma tumor necrosis factor (TNF)-α and nitric oxide (NO), increased pulmonary edema, thickened alveolar septa, and decreased survival rate. These changes were ameliorated by OroA (15 mg/kg, iv) administered 1 hr or 6 hrs after LPS challenge. This post-treatment also significantly attenuated LPS-induced activation of nuclear factor-κB (NF-κB) and the release of high mobility group box 1 (HMGB1) in lung tissues. Furthermore, post-treatment with OroA (60 mg/kg, ip) administered 1 hr or 6 hrs after LPS challenge in mice significantly increased survival rate. CONCLUSION AND IMPLICATION OroA administered after induction of ALI by LPS significantly prevent and revere lung tissues injuries with increased survival rate. Positive post-treatment effects of OroA suggest that OroA is a potentially useful candidate for managing lung inflammation in LPS-induced endotoxemia and septic shock.
Collapse
Affiliation(s)
- Tzu-Ling Tseng
- Institute of Medical Sciences, Tzu-Chi University, Hualien, Taiwan
- Center of Vascular Medicine, College of Life Science, Tzu-Chi University, Hualien, Taiwan
| | - Mei-Fang Chen
- Center of Vascular Medicine, College of Life Science, Tzu-Chi University, Hualien, Taiwan
- Department of Research, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan
- Tzu-Chi College of Technology, Hualien, Taiwan
| | - Ming-Jen Tsai
- Institute of Pharmacology and Toxicology, Tzu-Chi University, Hualien, Taiwan
- Department of Emergency Medicine, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan
| | - Yung-Hsiang Hsu
- Institute of Medical Sciences, Tzu-Chi University, Hualien, Taiwan
- Department of Pathology, College of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Chin-Piao Chen
- Department of Chemistry, National Dong Hwa University, Hualien, Taiwan
| | - Tony J. F. Lee
- Institute of Pharmacology and Toxicology, Tzu-Chi University, Hualien, Taiwan
- Department of Life Science, Tzu-Chi University, Hualien, Taiwan
- Center of Vascular Medicine, College of Life Science, Tzu-Chi University, Hualien, Taiwan
- Department of Research, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- * E-mail:
| |
Collapse
|
74
|
Li J, Long C, Cui W, Wang H. Iptakalim ameliorates monocrotaline-induced pulmonary arterial hypertension in rats. J Cardiovasc Pharmacol Ther 2012; 18:60-9. [PMID: 22947433 DOI: 10.1177/1074248412458154] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES We sought to investigate the experimental therapeutic effects and mechanisms of iptakalim, a new adenosine triphosphate (ATP)-sensitive potassium channel (K(ATP)) opener, on monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) and right heart ventricle remodeling in rats. METHODS Rats were injected with a single dose (50 mg/kg, ip) of MCT and given iptakalim (1, 3, and 9 mg/kg·per d, orally [po]) or saline for 28 days. The hemodynamic and morphometric parameters were assessed. Tissue and plasma samples were collected for histological and molecular analysis. RESULTS Treatment with iptakalim at daily oral doses of 1, 3, and 9 mg/kg from the day of MCT injection attenuated the high right ventricle systolic pressure (RVSP) and the increased weight ratio of right ventricle (RV) to left ventricle (LV) plus septum (S) (RV/(LV+S)), decreased heart rate (HR) and decreased mean arterial pressure (MAP), inhibited the RV myocardial tissue cell apoptosis, and the RV myocardial cell B-type natriuretic peptide (BNP) protein expression. Iptakalim also decreased the serum levels of nitric oxide (NO), endothelin 1 (ET-1), BNP, and the levels of NO, ET-1, and tumor necrosis factor-alpha (TNF-α) in the lung tissue. CONCLUSION These results indicate that iptakalim prevents MCT-induced PAH and RV remodeling and its mechanisms are related to inhibiting the pathological increases in NO, ET-1, BNP, and TNF-α, and Iptakalim may be a promising candidate for the treatment of PAH.
Collapse
Affiliation(s)
- Junshan Li
- Cardiovascular Drug Research Center, Institute of Health and Environmental Medicine, Academy of Military Medical Sciences, Beijing, China
| | | | | | | |
Collapse
|
75
|
Rentea RM, Liedel JL, Fredrich K, Welak SR, Pritchard KA, Oldham KT, Simpson PM, Gourlay DM. Intestinal alkaline phosphatase administration in newborns decreases systemic inflammatory cytokine expression in a neonatal necrotizing enterocolitis rat model. J Surg Res 2012; 177:228-34. [PMID: 22687880 DOI: 10.1016/j.jss.2012.05.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 05/05/2012] [Accepted: 05/10/2012] [Indexed: 11/24/2022]
Abstract
BACKGROUND Supplementation of intestinal alkaline phosphatase (IAP), an endogenous protein expressed in the intestines, decreases the severity of necrotizing enterocolitis (NEC)-associated intestinal injury and permeability. We hypothesized that IAP administration is protective in a dose-dependent manner of the inflammatory response in a neonatal rat model. MATERIALS AND METHODS Pre- and full-term newborn Sprague-Dawley rat pups were sacrificed on day of life 3. Control pups were vaginally delivered and dam fed. Preterm pups were delivered via cesarean section and exposed to intermittent hypoxia and formula feeds containing lipopolysaccharide (NEC) with and without IAP. Three different standardized doses were administered to a group of pups treated with 40, 4, and 0.4U/kg of bovine IAP (NEC+IAP40, IAP4, or IAP0.4U). Reverse transcription-real-time polymerase chain reaction (RT-PCR) for inducible nitric oxide synthase (iNOS) and tumor necrosis factor (TNF)-α on liver and lung tissues and serum cytokine analysis for interleukin (IL)-1β, IL-6, IL-10, and TNF-α were performed. Data were analyzed by Kruskal-Wallis and Mann-Whitney tests, expressed as mean±standard error of the mean and P≤0.05 considered significant. RESULTS Levels of cytokines IL-1β, IL-6, and TNF-α increased significantly in NEC versus control, returning to control levels with increasing doses of supplemental enteral IAP. Hepatic and pulmonary TNF-α and iNOS messenger ribonucleic acid expressions increased in NEC, and the remaining elevated despite IAP supplementation. CONCLUSIONS Proinflammatory cytokine expression is increased systemically with intestinal NEC injury. Administration of IAP significantly reduces systemic proinflammatory cytokine expression in a dose-dependent manner. Early supplemental enteral IAP may reduce NEC-related injury and be useful for reducing effects caused by a proinflammatory cascade.
Collapse
Affiliation(s)
- Rebecca M Rentea
- Department of Surgery Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Kvietys PR, Granger DN. Role of reactive oxygen and nitrogen species in the vascular responses to inflammation. Free Radic Biol Med 2012; 52:556-592. [PMID: 22154653 PMCID: PMC3348846 DOI: 10.1016/j.freeradbiomed.2011.11.002] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/04/2011] [Accepted: 11/04/2011] [Indexed: 12/23/2022]
Abstract
Inflammation is a complex and potentially life-threatening condition that involves the participation of a variety of chemical mediators, signaling pathways, and cell types. The microcirculation, which is critical for the initiation and perpetuation of an inflammatory response, exhibits several characteristic functional and structural changes in response to inflammation. These include vasomotor dysfunction (impaired vessel dilation and constriction), the adhesion and transendothelial migration of leukocytes, endothelial barrier dysfunction (increased vascular permeability), blood vessel proliferation (angiogenesis), and enhanced thrombus formation. These diverse responses of the microvasculature largely reflect the endothelial cell dysfunction that accompanies inflammation and the central role of these cells in modulating processes as varied as blood flow regulation, angiogenesis, and thrombogenesis. The importance of endothelial cells in inflammation-induced vascular dysfunction is also predicated on the ability of these cells to produce and respond to reactive oxygen and nitrogen species. Inflammation seems to upset the balance between nitric oxide and superoxide within (and surrounding) endothelial cells, which is necessary for normal vessel function. This review is focused on defining the molecular targets in the vessel wall that interact with reactive oxygen species and nitric oxide to produce the characteristic functional and structural changes that occur in response to inflammation. This analysis of the literature is consistent with the view that reactive oxygen and nitrogen species contribute significantly to the diverse vascular responses in inflammation and supports efforts that are directed at targeting these highly reactive species to maintain normal vascular health in pathological conditions that are associated with acute or chronic inflammation.
Collapse
Affiliation(s)
- Peter R Kvietys
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA.
| |
Collapse
|
77
|
The Effects of the Combination of Sodium Ferulate and Oxymatrine on Lipopolysaccharide-Induced Acute Lung Injury in Mice. Inflammation 2012; 35:1161-8. [DOI: 10.1007/s10753-011-9424-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
78
|
Luteolin Suppresses Inflammatory Mediator Expression by Blocking the Akt/NFκB Pathway in Acute Lung Injury Induced by Lipopolysaccharide in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2012:383608. [PMID: 22203870 PMCID: PMC3235668 DOI: 10.1155/2012/383608] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 09/09/2011] [Accepted: 09/09/2011] [Indexed: 11/25/2022]
Abstract
Acute lung injury (ALI), instilled by lipopolysaccharide (LPS), is a severe illness with excessive mortality and has no specific treatment strategy. Luteolin is an anti-inflammatory flavonoid and widely distributed in the plants. Pretreatment with luteolin inhibited LPS-induced histological changes of ALI and lung tissue edema. In addition, LPS-induced inflammatory responses, including increased vascular permeability, tumor necrosis factor (TNF)-α and interleukin (IL)-6 production, and expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), were also reduced by luteolin in a concentration-dependent manner. Furthermore, luteolin suppressed activation of NFκB and its upstream molecular factor, Akt. These results suggest that the protection mechanism of luteolin is by inhibition of NFκB activation possibly via Akt.
Collapse
|
79
|
Huang SW, Lee YP, Hung YT, Lin CH, Chuang JI, Lei HY, Su IJ, Yu CK. Exogenous interleukin-6, interleukin-13, and interferon-γ provoke pulmonary abnormality with mild edema in enterovirus 71-infected mice. Respir Res 2011; 12:147. [PMID: 22054060 PMCID: PMC3223501 DOI: 10.1186/1465-9921-12-147] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 11/06/2011] [Indexed: 11/10/2022] Open
Abstract
Background Neonatal mice developed neurological disease and pulmonary dysfunction after an infection with a mouse-adapted human Enterovirus 71 (EV71) strain MP4. However, the hallmark of severe human EV71 infection, pulmonary edema (PE), was not evident. Methods To test whether EV71-induced PE required a proinflammatory cytokine response, exogenous pro-inflammatory cytokines were administered to EV71-infected mice during the late stage of infection. Results After intracranial infection of EV71/MP4, 7-day-old mice developed hind-limb paralysis, pulmonary dysfunction, and emphysema. A transient increase was observed in serum IL-6, IL-10, IL-13, and IFN-γ, but not noradrenaline. At day 3 post infection, treatment with IL-6, IL-13, and IFN-γ provoked mild PE and severe emphysema that were accompanied by pulmonary dysfunction in EV71-infected, but not herpes simplex virus-1 (HSV-1)-infected control mice. Adult mice did not develop PE after an intracerebral microinjection of EV71 into the nucleus tractus solitarii (NTS). While viral antigen accumulated in the ventral medulla and the NTS of intracerebrally injected mice, neuronal loss was observed in the ventral medulla only. Conclusions Exogenous IL-6, IL-13, and IFN-γ treatment could induce mild PE and exacerbate pulmonary abnormality of EV71-infected mice. However, other factors such as over-activation of the sympathetic nervous system may also be required for the development of classic PE symptoms.
Collapse
Affiliation(s)
- Szu-Wei Huang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Mu E, Ding R, An X, Li X, Chen S, Ma X. Heparin attenuates lipopolysaccharide-induced acute lung injury by inhibiting nitric oxide synthase and TGF-β/Smad signaling pathway. Thromb Res 2011; 129:479-85. [PMID: 22035631 DOI: 10.1016/j.thromres.2011.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 09/13/2011] [Accepted: 10/04/2011] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Heparin, a potent blood anticoagulant, has been shown to exert a variety of pharmacological activities. The purpose of this study was to investigate whether heparin has a beneficial effect on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in rats and to further explore the possible underlying mechanisms. MATERIALS AND METHODS Adult Sprague-Dawley rats were randomly assigned into the control, heparin, LPS, and LPS plus heparin groups. ALI was induced by intratracheal instillation of LPS at a dose of 1 mg/kg. Rats in the LPS plus heparin group were intravenously received 50 U/ kg heparin every 1 h after the induction of ALI. RESULTS We found that heparin significantly improved LPS-induced lung pathological changes, inhibited myeloperoxidase (MPO) activity, and reduced malondialdehyde (MDA) level and lung wet/dry weight ratio. Heparin also inhibited the release of tumor necrosis factor (TNF)-α and interleukin (IL)-6, and markedly decreased the expression of inducible nitric oxide synthase (iNOS) in lung tissues and thus prevented nitric oxide (NO) release in response to LPS challenge. Additionally, heparin decreased the expression of transforming growth factor-β1 (TGF-β1), p-Smad 2, and p-Smad 3, which are all important molecules of the TGF-β1/Smad signaling pathway. CONCLUSIONS Heparin significantly ameliorated the lung injury induced by LPS in rats via the inhibition of nitric oxide synthase expression and the TGF-β/Smad pathway. Heparin may be a potential therapeutic reagent for treating ALI in the future.
Collapse
Affiliation(s)
- En Mu
- Department of Intensive Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | | | | | | | | | | |
Collapse
|
81
|
Liu Y, Wu H, Nie YC, Chen JL, Su WW, Li PB. Naringin attenuates acute lung injury in LPS-treated mice by inhibiting NF-κB pathway. Int Immunopharmacol 2011; 11:1606-12. [DOI: 10.1016/j.intimp.2011.05.022] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 05/19/2011] [Accepted: 05/19/2011] [Indexed: 01/08/2023]
|
82
|
Kumagai Y, Shinkai Y, Miura T, Cho AK. The chemical biology of naphthoquinones and its environmental implications. Annu Rev Pharmacol Toxicol 2011; 52:221-47. [PMID: 21942631 DOI: 10.1146/annurev-pharmtox-010611-134517] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Quinones are a group of highly reactive organic chemical species that interact with biological systems to promote inflammatory, anti-inflammatory, and anticancer actions and to induce toxicities. This review describes the chemistry, biochemistry, and cellular effects of 1,2- and 1,4-naphthoquinones and their derivatives. The naphthoquinones are of particular interest because of their prevalence as natural products and as environmental chemicals, present in the atmosphere as products of fuel and tobacco combustion. 1,2- and 1,4-naphthoquinones are also toxic metabolites of naphthalene, the major polynuclear aromatic hydrocarbon present in ambient air. Quinones exert their actions through two reactions: as prooxidants, reducing oxygen to reactive oxygen species; and as electrophiles, forming covalent bonds with tissue nucleophiles. The targets for these reactions include regulatory proteins such as protein tyrosine phosphatases; Kelch-like ECH-associated protein 1, the regulatory protein for NF-E2-related factor 2; and the glycolysis enzyme glyceraldehyde-3-phosphate dehydrogenase. Through their actions on regulatory proteins, quinones affect various cell signaling pathways that promote and protect against inflammatory responses and cell damage. These actions vary with the specific quinone and its concentration. Effects of exposure to naphthoquinones as environmental chemicals can vary with the physical state, i.e., whether the quinone is particle bound or is in the vapor state. The exacerbation of pulmonary diseases by air pollutants can, in part, be attributed to quinone action.
Collapse
Affiliation(s)
- Yoshito Kumagai
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | |
Collapse
|
83
|
Hawkes M, Opoka RO, Namasopo S, Miller C, Conroy AL, Serghides L, Kim H, Thampi N, Liles WC, John CC, Kain KC. Nitric oxide for the adjunctive treatment of severe malaria: hypothesis and rationale. Med Hypotheses 2011; 77:437-44. [PMID: 21745716 PMCID: PMC3162048 DOI: 10.1016/j.mehy.2011.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 06/07/2011] [Indexed: 11/24/2022]
Abstract
We hypothesize that supplemental inhaled nitric oxide (iNO) will improve outcomes in children with severe malaria receiving standard antimalarial therapy. The rationale for the hypothesized efficacy of iNO rests on: (1) biological plausibility, based on known actions of NO in modulating endothelial activation; (2) pre-clinical efficacy data from animal models of experimental cerebral malaria; and (3) a human trial of the NO precursor l-arginine, which improved endothelial function in adults with severe malaria. iNO is an attractive new candidate for the adjunctive treatment of severe malaria, given its proven therapeutic efficacy in animal studies, track record of safety in clinical practice and numerous clinical trials, inexpensive manufacturing costs, and ease of administration in settings with limited healthcare infrastructure. We plan to test this hypothesis in a randomized controlled trial (ClinicalTrials.gov Identifier: NCT01255215).
Collapse
Affiliation(s)
- Michael Hawkes
- Sandra A. Rotman Laboratories, McLaughlin-Rotman Centre for Global Health, Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Kelleher ZT, Potts EN, Brahmajothi MV, Foster MW, Auten RL, Foster WM, Marshall HE. NOS2 regulation of LPS-induced airway inflammation via S-nitrosylation of NF-{kappa}B p65. Am J Physiol Lung Cell Mol Physiol 2011; 301:L327-33. [PMID: 21724860 DOI: 10.1152/ajplung.00463.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inducible nitric oxide synthase (NOS2) expression is increased in the airway epithelium in acute inflammatory disorders although the physiological impact remains unclear. We have previously shown that NOS2 inhibits NF-κB (p50-p65) activation in respiratory epithelial cells by inducing S-nitrosylation of the p65 monomer (SNO-p65). In addition, we have demonstrated that mouse lung SNO-p65 levels are acutely depleted in a lipopolysaccharide (LPS) model of lung injury and that augmenting SNO-p65 levels before LPS treatment results in decreased airway epithelial NF-κB activation, airway inflammation, and lung injury. We now show that aerosolized LPS induces NOS2 expression in the respiratory epithelium concomitant with an increase in lung SNO-p65 levels and a decrease in airway NF-κB activity. Genetic deletion of NOS2 results in an absence of SNO-p65 formation, persistent NF-κB activity in the respiratory epithelium, and prolonged airway inflammation. These results indicate that a primary function of LPS-induced NOS2 expression in the respiratory epithelium is to modulate the inflammatory response through deactivation of NF-κB via S-nitrosylation of p65, thereby counteracting the initial stimulus-coupled denitrosylation.
Collapse
Affiliation(s)
- Zachary T Kelleher
- Division of Pulmonary Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
85
|
Endogenous MCP-1 promotes lung inflammation induced by LPS and LTA. Mol Immunol 2011; 48:1468-76. [DOI: 10.1016/j.molimm.2011.04.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 03/28/2011] [Accepted: 04/01/2011] [Indexed: 02/07/2023]
|
86
|
The effect of hyperbaric oxygen treatment on aspiration pneumonia. J Mol Histol 2011; 42:301-10. [PMID: 21656021 DOI: 10.1007/s10735-011-9334-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 05/24/2011] [Indexed: 12/26/2022]
Abstract
We have studied whether hyperbaric oxygen (HBO) prevents different pulmonary aspiration materials-induced lung injury in rats. The experiments were designed in 60 Sprague-Dawley rats, ranging in weight from 250 to 300 g, randomly allotted into one of six groups (n = 10): saline control, Biosorb Energy Plus (BIO), hydrochloric acid (HCl), saline + HBO treated, BIO + HBO treated, and HCl + HBO treated. Saline, BIO, HCl were injected into the lungs in a volume of 2 ml/kg. A total of seven HBO sessions were performed at 2,4 atm 100% oxygen for 90 min at 6-h intervals. Seven days later, rats were sacrificed, and both lungs in all groups were examined biochemically and histopathologically. Our findings show that HBO inhibits the inflammatory response reducing significantly (P < 0.05) peribronchial inflammatory cell infiltration, alveolar septal infiltration, alveolar edema, alveolar exudate, alveolar histiocytes, interstitial fibrosis, granuloma, and necrosis formation in different pulmonary aspiration models. Pulmonar aspiration significantly increased the tissue HP content, malondialdehyde (MDA) levels and decreased (P < 0.05) the antioxidant enzyme (SOD, GSH-Px) activities. HBO treatment significantly (P < 0.05) decreased the elevated tissue HP content, and MDA levels and prevented inhibition of SOD, and GSH-Px (P < 0.05) enzymes in the tissues. Furthermore, there is a significant reduction in the activity of inducible nitric oxide synthase, TUNEL and arise in the expression of surfactant protein D in lung tissue of different pulmonary aspiration models with HBO therapy. It was concluded that HBO treatment might be beneficial in lung injury, therefore, shows potential for clinical use.
Collapse
|
87
|
Sumi D, Akimori M, Inoue KI, Takano H, Kumagai Y. 1,2-Naphthoquinone suppresses lipopolysaccharide-dependent activation of IKKβ/NF-κB/NO signaling: an alternative mechanism for the disturbance of inducible NO synthase-catalyzed NO formation. J Toxicol Sci 2011; 35:891-8. [PMID: 21139339 DOI: 10.2131/jts.35.891] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
1,2-Naphthoquinone (1,2-NQ) is an uncoupling agent for constitutive nitric oxide (NO) synthase (NOS), thereby inhibiting its catalytic activity. However, little information on whether this quinone can affect inducible NOS (iNOS) is available. To address this issue, we examined the effect of 1,2-NQ on lipopolysaccharide (LPS)-mediated induction of iNOS. Exposure of LPS-challenged RAW264.7 cells to 1,2-NQ resulted in decreased NO formation through a reduction in iNOS production. Under these conditions, LPS-induced activation of nuclear transcription factor-κB (NF-κB) coupled to phosphorylation of inhibitory κBα (IκBα) declined. Similar effects of 1,2-NQ were observed in the lungs of mice exposed to LPS. Using IκB kinase β (IKKβ)-transfected RAW264.7 cells and recombinant IKKβ protein, we found that 1,2-NQ diminished the phosphorylation of IκB by IKKβ enzymatic activity. Taken together, these results suggest that 1,2-NQ reduces iNOS-catalyzed NO production through 1) an uncoupling reaction, as reported previously, and/or 2) disruption of IKKβ/NF-κB signaling.
Collapse
Affiliation(s)
- Daigo Sumi
- Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | | | | | | |
Collapse
|
88
|
Abstract
Chemokines are a large group of small cytokines known for their chemotactic ability to regulate the recruitment of leukocytes to sites of inflammation. This occurs through the binding of chemokines to their receptors located on the leukocyte that results in cellular changes such as actin rearrangement and cell shape, which allow for the migration of the leukocyte. In addition to regulating leukocyte function, it is now becoming apparent that other nonhematopoetic cells, such as smooth muscle cells and endothelial cells, can also be regulated by chemokines. Studies within the past 10 years has demonstrated the presence of various chemokine receptors on endothelial cells as well as the ability of chemokines to activate these receptors resulting in various cellular responses including migration, proliferation, and cellular activation. The purpose of this review is to highlight the research that has been done to date demonstrating the important role for chemokines in regulating endothelial function during various inflammatory conditions associated with angiogenesis, homeostasis, and leukocyte transmigration. This review will focus specifically on the role of the endothelium in mediating chemokine effects associated with wound healing, atherosclerosis, and autoimmune diseases, conditions where leukocyte recruitment and angiogenesis play a major role. Recent progress in the development and implementation of therapeutics agents against these small molecules, or their receptors, will also be addressed.
Collapse
Affiliation(s)
- Cecilia L Speyer
- Department of Surgery, Wayne State University School of Medicine, Detroit, MI 48201, United States.
| | | |
Collapse
|
89
|
Kobayashi Y. The regulatory role of nitric oxide in proinflammatory cytokine expression during the induction and resolution of inflammation. J Leukoc Biol 2010; 88:1157-62. [DOI: 10.1189/jlb.0310149] [Citation(s) in RCA: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
90
|
Zhang X, Huang H, Yang T, Ye Y, Shan J, Yin Z, Luo L. Chlorogenic acid protects mice against lipopolysaccharide-induced acute lung injury. Injury 2010; 41:746-52. [PMID: 20227691 DOI: 10.1016/j.injury.2010.02.029] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 12/15/2009] [Accepted: 02/15/2010] [Indexed: 02/02/2023]
Abstract
Chlorogenic acid (CGA) is one of the most abundant polyphenol compounds in human diet. Our previous in vitro study demonstrates that CGA presents anti-inflammatory activities in RAW 264.7 cells. Here we show that CGA protects mice against lipopolysaccharide (LPS)-induced acute lung injury (ALI). We treated mice with CGA (5, 20 and 50 mg/kg body weight) 30 min or 3 h after intratracheal administration of LPS. The histological results showed that CGA, at dose of 50 mg/kg, protected mice from LPS-induced ALI which displayed by edema, haemorrhage, blood vessel and alveolar structural damage. CGA inhibited LPS-increased pulmonary MPO activity and migration of polymorphonuclear neutrophils (PMNs) into bronchoalveolar lavage fluid (BALF). Furthermore, CGA markedly decreased the activity of inducible nitric oxide synthase (iNOS) in lung tissues and thus prevented nitric oxide (NO) release in response to LPS challenge. In conclusion, these results indicated that CGA was greatly effective in inhibiting ALI and might act as a potential therapeutic reagent for treating ALI in the future.
Collapse
Affiliation(s)
- Xu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Rd, Nanjing, Jiangsu 210093, PR China
| | | | | | | | | | | | | |
Collapse
|
91
|
Sun L, Gao H, Sarma VJ, Guo RF, Ward PA. Adenovirus-mediated in vivo silencing of anaphylatoxin receptor C5aR. J Biomed Biotechnol 2010; 2006:28945. [PMID: 17057363 PMCID: PMC1510939 DOI: 10.1155/jbb/2006/28945] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
C5a, one of the most potent inflammatory peptides, induces its
inflammatory functions by interacting with C5a receptor (C5aR)
that belongs to the rhodopsin family of seven-transmembrane G
protein-coupled receptors. C5a/C5aR signaling has been implicated
in the pathogenesis of many inflammatory and immunological
diseases such as sepsis and acute lung injury. Widespread
upregulation of C5aR has been seen at both the protein level and
transcriptional level under pathological conditions. Here, we show
that C5aR gene expression can be specifically suppressed by siRNA,
both in vitro and in vivo. A panel of chemically siRNA
oligonucleotides was first synthesized to identify the functional
siRNA sequences. The short hairpin RNAs (shRNAs) were also
designed, cloned, and tested for the silencing effects in C5aR
transfected cells. The effective shRNA expression cassettes were
then transferred to an adenovirus DNA vector. ShRNA-expressing
adenoviruses were intratracheally administered into mouse lung,
and a significant in vivo silencing of C5aR was obtained four days
after administration. Thus, C5aR shRNA-expressing adenoviruses
appear to be an alternative strategy for the treatment of
complement-induced disorders.
Collapse
Affiliation(s)
- Lei Sun
- Department of Pathology, University of Michigan Medical School,
1301 Catherine Road, Ann Arbor, MI 48109-0602, USA
| | - Hongwei Gao
- Department of Pathology, University of Michigan Medical School,
1301 Catherine Road, Ann Arbor, MI 48109-0602, USA
| | - Vidya J. Sarma
- Department of Pathology, University of Michigan Medical School,
1301 Catherine Road, Ann Arbor, MI 48109-0602, USA
| | - Ren-feng Guo
- Department of Pathology, University of Michigan Medical School,
1301 Catherine Road, Ann Arbor, MI 48109-0602, USA
- *Ren-feng Guo:
| | - Peter A. Ward
- Department of Pathology, University of Michigan Medical School,
1301 Catherine Road, Ann Arbor, MI 48109-0602, USA
| |
Collapse
|
92
|
Ramos DS, Olivo CR, Quirino Santos Lopes FDT, Toledo AC, Martins MA, Lazo Osório RA, Dolhnikoff M, Ribeiro W, Vieira RDP. Low-intensity swimming training partially inhibits lipopolysaccharide-induced acute lung injury. Med Sci Sports Exerc 2010; 42:113-119. [PMID: 20010123 DOI: 10.1249/mss.0b013e3181ad1c72] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Aerobic exercise decreases pulmonary inflammation and remodeling in experimental models of allergic asthma. However, the effects of aerobic exercise on pulmonary inflammation of nonallergic origin, such as in experimental models of acute lung injury induced by lipopolysaccharide (LPS), have not been evaluated. OBJECTIVE The present study evaluated the effects of aerobic exercise in a model of LPS-induced acute lung injury. METHODS BALB/c mice were divided into four groups: Control, Aerobic Exercise, LPS, and Aerobic Exercise + LPS. Swimming tests were conducted at baseline and at 3 and 6 wk. Low-intensity swimming training was performed for 6 wk, four times per week, 60 min per session. Intranasal LPS (1 mg x kg(-1) (60 microg per mouse)) was instilled 24 h after the last swimming physical test in the LPS and Aerobic Exercise + LPS mice, and the animals were studied 24 h after LPS instillation. Exhaled nitric oxide, respiratory mechanics, total and differential cell counts in bronchoalveolar lavage, and lung parenchymal inflammation and remodeling were evaluated. RESULTS LPS instillation resulted in increased levels of exhaled nitric oxide (P < 0.001), higher numbers of neutrophils in the bronchoalveolar lavage (P < 0.001) and in the lung parenchyma (P < 0.001), and decreased lung tissue resistance (P < 0.05) and volume proportion of elastic fibers (P < 0.01) compared with the Control group. Swim training in LPS-instilled animals resulted in significantly lower exhaled nitric oxide levels (P < 0.001) and fewer neutrophils in the bronchoalveolar lavage (P < 0.001) and the lung parenchyma (P < 0.01) compared with the LPS group. CONCLUSIONS These results suggest that low-intensity swimming training inhibits lung neutrophilic inflammation, but not remodeling and impaired lung mechanics, in a model of LPS-induced acute lung injury.
Collapse
Affiliation(s)
- Daniel Souza Ramos
- Laboratory of Physiology and Pharmacodynamics, Institute of Research and Development, University of Vale do Paraíba, Săo Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
The Effect of Epigallocatechin Gallate on Lipopolysaccharide-Induced Acute Lung Injury in a Murine Model. Inflammation 2009; 33:82-91. [DOI: 10.1007/s10753-009-9161-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
94
|
von Toerne C, Schmidt C, Adams J, Kiss E, Bedke J, Porubsky S, Gretz N, Lindenmeyer MT, Cohen CD, Gröne HJ, Nelson PJ. Wnt pathway regulation in chronic renal allograft damage. Am J Transplant 2009; 9:2223-39. [PMID: 19681821 DOI: 10.1111/j.1600-6143.2009.02762.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The Wnt signaling pathway, linked to development, has been proposed to be recapitulated during the progressive damage associated with chronic organ failure. Chronic allograft damage following kidney transplantation is characterized by progressive fibrosis and a smoldering inflammatory infiltrate. A modified, Fischer 344 (RT1(lvl)) to Lewis (RT1(l)) rat renal allograft model that reiterates many of the major pathophysiologic processes seen in patients with chronic allograft failure was used to study the progressive disease phenotype and specific gene product expression by immunohistochemistry and transcriptomic profiling. Central components of the Tgfb, canonical Wnt and Wnt-Ca2+ signaling pathways were significantly altered with the development of chronic damage. In the canonical Wnt pathway, Wnt3, Lef1 and Tcf1 showed differential regulation. Target genes Fn1, Cd44, Mmp7 and Nos2 were upregulated and associated with the progression of renal damage. Changes in the Wnt-Ca2+ pathway were evidenced by increased expression of Wnt6, Wnt7a, protein kinase C, Cam Kinase II and Nfat transcription factors and the target gene vimentin. No evidence for alterations in the Wnt planar cell polarity (PCP) pathway was detected. Overall results suggest cross talk between the Wnt and Tgfb signaling pathways during allograft inflammatory damage and present potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- C von Toerne
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Meng X, Kawahara KI, Matsushita K, Nawa Y, Shrestha B, Kikuchi K, Sameshima H, Hashiguchi T, Maruyama I. Attenuation of LPS-induced iNOS expression by 1,5-anhydro-d-fructose. Biochem Biophys Res Commun 2009; 387:42-6. [DOI: 10.1016/j.bbrc.2009.06.108] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 11/30/2022]
|
96
|
Kantrow SP, Shen Z, Jagneaux T, Zhang P, Nelson S. Neutrophil-mediated lung permeability and host defense proteins. Am J Physiol Lung Cell Mol Physiol 2009; 297:L738-45. [PMID: 19648288 DOI: 10.1152/ajplung.00045.2009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neutrophil recruitment to the alveolar space is associated with increased epithelial permeability. The present study investigated in mice whether neutrophil recruitment to the lung leads to accumulation of plasma-derived host defense proteins in the alveolar space and whether respiratory burst contributes to this increase in permeability. Albumin, complement C1q, and IgM were increased in bronchoalveolar lavage (BAL) fluid 6 h after intratracheal LPS challenge. Neutrophil depletion before LPS treatment completely prevented this increase in BAL fluid protein concentration. Respiratory burst was not detected in neutrophils isolated from BAL fluid, and BAL proteins were increased in mice deficient in a key subunit of the respiratory burst apparatus, gp91(phox), similar to wild-type mice. Neutrophil recruitment elicited by intratracheal instillation of the chemokines macrophage inflammatory protein-2 and keratinocyte-derived chemokine was also accompanied by accumulation of albumin, C1q, and IgM. During neutrophil recruitment to the alveolar space, epithelial permeability facilitates delivery of host defense proteins. The observed increase in epithelial permeability requires recruitment of neutrophils, but not activation of the respiratory burst, and occurs with chemokine-induced neutrophil migration independent of LPS exposure.
Collapse
Affiliation(s)
- Stephen P Kantrow
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
| | | | | | | | | |
Collapse
|
97
|
Pasula R, Britigan BE, Turner J, Martin WJ. Airway delivery of silica increases susceptibility to mycobacterial infection in mice: potential role of repopulating macrophages. THE JOURNAL OF IMMUNOLOGY 2009; 182:7102-9. [PMID: 19454707 DOI: 10.4049/jimmunol.0803642] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Silica exposure results in an increased lifelong risk of developing mycobacterial pulmonary infections. To date, there are no animal models that replicate this finding to permit assessment of the mechanisms underlying susceptibility to mycobacterial infection. To test the hypothesis that prior silica exposure increases risk of mycobacterial infection, we intratracheally (I.T.) administered silica, a control dust (Al(2)O(3)) or saline into mechanically ventilated C57BL/6 mice. Later, the mice received Mycobacterium avium or Mycobacterium tuberculosis I.T. Mice were sacrificed at defined time points and mycobacteria in lung homogenates were quantified. M. avium or M. tuberculosis infection was markedly increased in silica-exposed mice compared with mice exposed to either Al(2)O(3) or saline beginning 3 wk after silica exposure. Similarly, lung sections from silica-exposed mice had many more acid fast bacilli(+) (AFB(+)) organisms than from control mice. Alveolar macrophages (AMs) from bronchoalveolar lavage of silica-exposed mice also revealed a higher number of mycobacteria compared with mice treated with Al(2)O(3) or saline. In addition, passive transfer of AMs from silica-exposed mice to control mice increased M. tuberculosis susceptibility. These results indicate that silica exposure converts mycobacteria-resistant mice into mycobacteria-susceptible mice via a process that likely involves a new population of AMs that are more susceptible to mycobacterial infection.
Collapse
Affiliation(s)
- Rajamouli Pasula
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| | | | | | | |
Collapse
|
98
|
Pulmonary toxicity and kinetic study of Cy5.5-conjugated superparamagnetic iron oxide nanoparticles by optical imaging. Toxicol Appl Pharmacol 2009; 239:106-15. [PMID: 19520096 DOI: 10.1016/j.taap.2009.05.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 05/25/2009] [Accepted: 05/30/2009] [Indexed: 11/23/2022]
Abstract
Recent advances in the development of nanotechnology and devices now make it possible to accurately deliver drugs or genes to the lung. Magnetic nanoparticles can be used as contrast agents, thermal therapy for cancer, and be made to concentrate to target sites through an external magnetic field. However, these advantages may also become problematic when taking into account safety and toxicological factors. This study demonstrated the pulmonary toxicity and kinetic profile of anti-biofouling polymer coated, Cy5.5-conjugated thermally cross-linked superparamagnetic iron oxide nanoparticles (TCL-SPION) by optical imaging. Negatively charged, 36 nm-sized, Cy5.5-conjugated TCL-SPION was prepared for optical imaging probe. Cy5.5-conjugated TCL-SPION was intratracheally instilled into the lung by a non-surgical method. Cy5.5-conjugated TCL-SPION slightly induced pulmonary inflammation. The instilled nanoparticles were distributed mainly in the lung and excreted in the urine via glomerular filtration. Urinary excretion was peaked at 3 h after instillation. No toxicity was found under the concentration of 1.8 mg/kg and the half-lives of nanoparticles in the lung and urine were estimated to be about 14.4+/-0.54 h and 24.7+/-1.02 h, respectively. Although further studies are required, our results showed that Cy5.5-conjugated TCL-SPION can be a good candidate for use in pulmonary delivery vehicles and diagnostic probes.
Collapse
|
99
|
Inhibition of the MAP kinase ERK protects from lipopolysaccharide-induced lung injury. Biochem Pharmacol 2009; 77:1827-34. [DOI: 10.1016/j.bcp.2009.03.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 03/07/2009] [Accepted: 03/10/2009] [Indexed: 01/14/2023]
|
100
|
Kanter M. Effects of Nigella sativa seed extract on ameliorating lung tissue damage in rats after experimental pulmonary aspirations. Acta Histochem 2009; 111:393-403. [PMID: 19428057 DOI: 10.1016/j.acthis.2008.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 10/27/2008] [Accepted: 10/30/2008] [Indexed: 12/31/2022]
Abstract
Aspiration of gastric contents can cause serious lung injury, although the mechanisms of pulmonary damage are still not clear and means of amelioration of the pulmonary damage have been little investigated. The black cumin seed, Nigella sativa L. (NS) has been shown to have specific health benefits and the aim of the current study was to investigate the possible beneficial effects of NS on experimental lung injury in male Wistar rats after pulmonary aspiration of different materials. The rats were randomly allotted into one of six experimental groups (n=7 per group): (1) saline control, (2) saline+NS treated, (3) Pulmocare (a specialized nutritional supplement given to pulmonary patients), (4) Pulmocare+NS treated, (5) hydrochloric acid, (6) hydrochloric acid+NS treated. The saline, Pulmocare and hydrochloric acid were injected into the lungs in a volume of 2 ml/kg. The rats received daily oral doses of NS volatile oil (400mg/kg body weight) by means of intragastric intubation for 7 days starting immediately after the pulmonary aspiration of the materials. After 7 days, the rats were sacrificed and tissue samples from both lungs were taken for histopathological investigation. To date, no similar study investigating the potential for NS treatment to protect against lung injury after pulmonary aspiration of materials has been reported. Our study showed that NS treatment inhibits the inflammatory pulmonary responses, reducing significantly (p<0.05) peribronchial inflammatory cell infiltration, alveolar septal infiltration, alveolar edema, alveolar exudate, alveolar macrophages, interstitial fibrosis, granuloma and necrosis formation in different pulmonary aspiration models. Our data indicate a significant reduction in the activity of inducible nitric oxide synthase (iNOS) and a rise in surfactant protein D in lung tissue of different pulmonary aspiration models after NS therapy. Based on our results, we conclude that NS treatment might be beneficial in lung injury and have potential clinical use.
Collapse
|