1
|
Sherekar P, Suke SG, Dhok A, Malegaonkar S, Dhale SA. Global scenario of silica-associated diseases: A review on emerging pathophysiology of silicosis and potential therapeutic regimes. Toxicol Rep 2025; 14:101941. [PMID: 39989982 PMCID: PMC11847043 DOI: 10.1016/j.toxrep.2025.101941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/25/2025] Open
Abstract
Silicosis is an occupational fibrotic lung disease caused by exposure to respirable crystalline silica dust particles produced during industrial activities. Other crystalline silica-induced pulmonary disorders include a predisposition to mycobacterial infections, obstructive airway diseases, idiopathic pulmonary fibrosis, and lung cancer. This review paper discusses the burden of silicosis and associated co-morbidities in developed as well as developing countries globally using the published data of various government agencies, related organizations, and epidemiological findings. Moreover, it sheds light on diverse mechanisms of silicosis, outlining molecular events and peculiar alterations in lung parenchyma leading to this occupational lung disease. Evaluation of pathophysiological mechanisms could aid in the identification of novel target molecules and treatments; to date, there is no curative treatment for silicosis. In recent periods, a lot of attention has been focused on the development and fabrication of suitable nanocarriers for improved and sustained drug delivery in the pulmonary system. Nanoparticle-based therapeutic modality has been evaluated in in-vitro and ex-vivo silicosis models for prolongation of drug activity and improved therapeutic outcomes. The preclinical findings open the doors to clinical trials for operational and regenerative nanoformulations, which eventually create a positive change in medical practice. The following review summarizes various therapeutic approaches available and in the pipe line for silicosis and also stresses the preventive practices for effectively combating this occupational hazard.
Collapse
Affiliation(s)
- Prasad Sherekar
- Department of Biotechnology, Priyadarshini College of Engineering, Priyadarshini Campus, Hingna Road, Nagpur 440 019, India
- Department of Biochemistry, Jawaharlal Nehru Medical College, DattaMeghe Institute of Higher Education and Research (Deemed to be University), Wardha 442 005, India
| | - Sanvidhan G. Suke
- Department of Biotechnology, Priyadarshini College of Engineering, Priyadarshini Campus, Hingna Road, Nagpur 440 019, India
| | - Archana Dhok
- Department of Biochemistry, Jawaharlal Nehru Medical College, DattaMeghe Institute of Higher Education and Research (Deemed to be University), Wardha 442 005, India
| | - Srikant Malegaonkar
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, MIHAN, Nagpur 441 108, India
| | - Shrikrishna A. Dhale
- Department of Civil Engineering, Priyadarshini College of Engineering, Priyadarshini Campus, Hingna Road, Nagpur 440 019, India
| |
Collapse
|
2
|
Zaaba NE, Al-Salam S, Beegam S, Elzaki O, Aldhaheri F, Nemmar A, Ali BH, Nemmar A. Attenuation of cisplatin-induced acute kidney injury by sanguinarine: modulation of oxidative stress, inflammation, and cellular damage. Front Pharmacol 2025; 16:1567888. [PMID: 40242453 PMCID: PMC11999955 DOI: 10.3389/fphar.2025.1567888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction Cisplatin (CP)-induced acute kidney injury (AKI) is a significant side effect of CP chemotherapy, driven by oxidative stress and inflammation. Sanguinarine (SANG), an alkaloid from the rhizomes of Sanguinaria canadensis and poppy-fumaria species, exhibits antioxidant and anti-inflammatory properties. This study examined SANG's effect on CP-induced AKI in mice and its underlying mechanisms. Methods Mice were orally administered 5 mg/kg SANG for 10 days. On the seventh day, they received a single intraperitoneal CP injection (20 mg/kg) and were sacrificed on the 11th day. Results SANG significantly improved CP-induced decreases in body weight, water intake, urine volume, relative kidney weight, creatinine clearance, albumin-to-creatinine ratio, and plasma urea and creatinine levels. It also reduced elevated plasma neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, cystatin C, and adiponectin levels, as well as renal markers of inflammation and oxidative stress induced by CP administration. SANG normalized kidney mitochondrial dysfunction, DNA damage, and apoptosis caused by CP. It also inhibited the CP-induced increase in the expression of phosphorylated nuclear factor-κB and autophagy markers in the kidney. Histological analysis showed that SANG reduced acute tubular necrosis and intraluminal protein accumulation due to CP. Discussion In conclusion, SANG mitigated CP-induced AKI by reducing inflammation, oxidative stress, DNA damage, apoptosis, and autophagy. Pending more comprehensive pharmacological and toxicological assessments, SANG may be regarded as a potential therapeutic agent for mitigating CP-induced AKI.
Collapse
Affiliation(s)
- Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Fatima Aldhaheri
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Anas Nemmar
- College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Badreldin H. Ali
- Emeritus Professor, Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Science, Sultan Qaboos University, Muscat, Oman
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
3
|
Zaaba NE, Yuvarayu P, Beegam S, Elzaki O, Arafat K, Attoub S, Nemmar A. Direct Effects of Waterpipe Smoke Extract on Aortic Endothelial Cells: An In Vitro Study. Physiol Res 2025; 74:69-78. [PMID: 40126144 PMCID: PMC11995937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/03/2024] [Indexed: 03/25/2025] Open
Abstract
Waterpipe smoking (WPS) has adverse health effects that include endothelial dysfunction with mechanisms involving oxidative stress and inflammation. Nonetheless, there is a scarcity of data on the direct impact of WPS on endothelial function. In this study, we assessed the in vitro effects of waterpipe smoke extract (WPSE) on aortic endothelial cell lines, namely the TeloHAEC. The WPSE markedly caused concentration- and time-dependent decreases in cellular viability. When compared with the control, at a concentration of 20 % and an incubation period of 48 h, the WPSE significantly increased the levels of lactate dehydrogenase, and markers of oxidative stress including thiobarbituric acid reactive substances, superoxide dismutase, catalase, and reduced glutathione. Moreover, the concentrations of proinflammatory cytokine (tumor necrosis factor alpha), and adhesion molecules (E-selectin and intercellular adhesion molecule-1) were also significantly augmented. Likewise, WPSE triggered mitochondrial dysfunction, DNA oxidative damage, as well as apoptosis in TeloHAEC cells. Similarly, cells cultured with WPSE have shown increased expression of phosphorylated nuclear factor-kappaB and hypoxia-inducible factor 1-alpha (HIF-1alpha). In conclusion, our study showed that WPSE triggers endothelial inflammation, oxidative stress, DNA damage, mitochondrial dysfunction, and apoptosis via mechanisms involving the activation of nuclear factor-kappaB and HIF-1alpha. Key words Waterpipe smoking, Aortic endothelial cells, Inflammation, Oxidative Stress.
Collapse
Affiliation(s)
- N E Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| | | | | | | | | | | | | |
Collapse
|
4
|
Beegam S, Al-Salam S, Zaaba NE, Elzaki O, Nemmar A. Prothrombotic State and Vascular Damage in Angiotensin II-Induced Hypertension: Influence of Waterpipe Smoke Exposure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2025; 2025:2670738. [PMID: 39959581 PMCID: PMC11824600 DOI: 10.1155/omcl/2670738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/04/2025] [Indexed: 02/18/2025]
Abstract
Hypertension is a risk factor for vascular injury and thrombotic complications, and smoking tobacco is a risk factor for the development and exacerbation of hypertension. The influence of waterpipe smoke (WPS) on coagulation and vascular injury in hypertension is not fully understood. Here, we evaluated the effects of WPS in mice made hypertensive (HT) by infusing angiotensin II (Ang II) for 42 days. On day 14 of the infusion of Ang II or vehicle (normotensive; NT), mice were exposed either to air or WPS for four consecutive weeks. Each session was 30 min/day for 5 days/week. The concentrations of tissue factor, von Willebrand factor, fibrinogen, and plasminogen activator inhibitor-1 were elevated in the HT + WPS group versus either HT + air or NT + WPS groups. Similarly, in the HT + WPS group, thrombogenicity was increased both in vivo and in vitro, compared with either HT + air or NT + WPS groups. In aortic tissue, adhesion molecules including P-selectin, E-selectin, intercellular adhesion molecule-1, and vascular adhesion molecule-1 were increased in the HT + WPS group versus the controls. Likewise, various proinflammatory cytokines and markers of oxidative stress augmented in the HT + WPS group compared with either HT + air or NT + WPS. DNA damage, cleaved caspase-3, and cytochrome C were increased in the HT + WPS group versus the controls. The immunohistochemical expression of nuclear factor erythroid 2-related factor 2 was increased in the HT + WPS group versus either HT + air or NT + WPS. Taken together, our findings show that WPS exposure intensified thrombogenicity and vascular damage in experimentally induced hypertension. Our data suggest that vascular toxicity of WPS may be exaggerated in hypertensive patients.
Collapse
Affiliation(s)
- Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, UAE
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, UAE
| | - Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, UAE
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, UAE
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, UAE
| |
Collapse
|
5
|
Beegam S, Zaaba NE, Elzaki O, Nemmar A. α-Bisabolol alleviates diesel exhaust particle-induced lung injury and mitochondrial dysfunction by regulating inflammatory, oxidative stress, and apoptotic biomarkers through the c-Jun N-terminal kinase signaling pathway. Front Pharmacol 2025; 15:1485101. [PMID: 39830335 PMCID: PMC11738621 DOI: 10.3389/fphar.2024.1485101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/11/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Exposure to particulate matter ≤2.5 μm in diameter (PM2.5) is associated with adverse respiratory outcomes, including alterations to lung morphology and function. These associations were reported even at concentrations lower than the current annual limit of PM2.5. Inhalation of PM2.5, of which diesel exhaust particles (DEPs) is a major component, induces lung inflammation and oxidative stress. α-Bisabolol (BIS) is a bioactive dietary phytochemical with various pharmacological properties, including anti-inflammatory and antioxidant actions. Here, we evaluated the possible protective effects of BIS on DEP-induced lung injury. Methods Mice were exposed to DEPs (20 µg/mouse) or saline (control) by intratracheal instillation. BIS was administered orally at two doses (25 and 50 mg/kg) approximately 1 h before DEP exposure. Twenty-four hours after DEP administration, multiple respiratory endpoints were evaluated. Results BIS administration was observed to prevent DEP-induced airway hyperreactivity to methacholine; influx of macrophages, neutrophils, and lymphocytes in the bronchoalveolar lavage fluid; and increases in epithelial and endothelial permeabilities. DEP exposure caused increases in the levels of myeloperoxidase, proinflammatory cytokines, and oxidative stress markers in lung tissue homogenates, and all these effects were abated by BIS treatment. The activities of mitochondrial complexes I, II, III, and IV were markedly increased in the lungs of mice exposed to DEPs, and these effects were significantly reduced in the BIS-treated group. Intratracheal instillation of DEPs induced DNA damage and increase in the apoptotic marker cleaved caspase-3. The latter effects were prevented in mice treated with BIS and exposed to DEPs. Moreover, BIS mitigated DEP-induced increase in the expression of phospho-c-Jun N-terminal kinase (JNK) in a dose-dependent manner. Discussion BIS markedly alleviated DEP-induced lung injury by regulating the inflammatory, oxidative stress, and apoptotic biomarkers through the JNK signaling pathway. Following additional studies, BIS may be considered as a plausible protective agent against inhaled-particle-induced pulmonary adverse effects.
Collapse
Affiliation(s)
| | | | | | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
6
|
Cattani-Cavalieri I, Trombetta-Lima M, Yan H, Manzano-Covarrubias AL, Baarsma HA, Oun A, van der Veen MM, Oosterhout E, Dolga AM, Ostrom RS, Valenca SS, Schmidt M. Diesel exhaust particles alter mitochondrial bioenergetics and cAMP producing capacity in human bronchial epithelial cells. FRONTIERS IN TOXICOLOGY 2024; 6:1412864. [PMID: 39118833 PMCID: PMC11306203 DOI: 10.3389/ftox.2024.1412864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction: Air pollution from diesel combustion is linked in part to the generation of diesel exhaust particles (DEP). DEP exposure induces various processes, including inflammation and oxidative stress, which ultimately contribute to a decline in lung function. Cyclic AMP (cAMP) signaling is critical for lung homeostasis. The impact of DEP on cAMP signaling is largely unknown. Methods: We exposed human bronchial epithelial (BEAS-2B) cells to DEP for 24-72 h and evaluated mitochondrial bioenergetics, markers of oxidative stress and inflammation and the components of cAMP signaling. Mitochondrial bioenergetics was measured at 72 h to capture the potential and accumulative effects of prolonged DEP exposure on mitochondrial function. Results: DEP profoundly altered mitochondrial morphology and network integrity, reduced both basal and ATP-linked respiration as well as the glycolytic capacity of mitochondria. DEP exposure increased gene expression of oxidative stress and inflammation markers such as interleukin-8 and interleukin-6. DEP significantly affected mRNA levels of exchange protein directly activated by cAMP-1 and -2 (Epac1, Epac2), appeared to increase Epac1 protein, but left phospho-PKA levels unhanged. DEP exposure increased A-kinase anchoring protein 1, β2-adrenoceptor and prostanoid E receptor subtype 4 mRNA levels. Interestingly, DEP decreased mRNA levels of adenylyl cyclase 9 and reduced cAMP levels stimulated by forskolin (AC activator), fenoterol (β2-AR agonist) or PGE2 (EPR agonist). Discussion: Our findings suggest that DEP induces mitochondrial dysfunction, a process accompanied by oxidative stress and inflammation, and broadly dampens cAMP signaling. These epithelial responses may contribute to lung dysfunction induced by air pollution exposure.
Collapse
Affiliation(s)
- Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Ana L. Manzano-Covarrubias
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hoeke A. Baarsma
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Asmaa Oun
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | | | - Emily Oosterhout
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rennolds S. Ostrom
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Samuel Santos Valenca
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
7
|
Jeong S, Bae S, Shin EC, Lee JH, Ha JH. Ellagic Acid Prevents Particulate Matter-Induced Pulmonary Inflammation and Hyperactivity in Mice: A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4523. [PMID: 36901532 PMCID: PMC10001477 DOI: 10.3390/ijerph20054523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 06/18/2023]
Abstract
The inhalation of fine particulate matter (PM) is a significant health-related environmental issue. Previously, we demonstrated that repeated PM exposure causes hyperlocomotive activity in mice, as well as inflammatory and hypoxic responses in their lungs. In this study, we evaluated the potential efficacy of ellagic acid (EA), a natural polyphenolic compound, against PM-induced pulmonary and behavioral abnormalities in mice. Four treatment groups were assigned in this study (n = 8): control (CON), particulate-matter-instilled (PMI), low-dose EA with PMI (EL + PMI), and high-dose EA with PMI (EH + PMI). EA (20 and 100 mg/kg body weight for low dose and high dose, respectively) was orally administered for 14 days in C57BL/6 mice, and after the eighth day, PM (5 mg/kg) was intratracheally instilled for 7 consecutive days. PM exposure induced inflammatory cell infiltration in the lungs following EA pretreatment. Moreover, PM exposure induced inflammatory protein expression in the bronchoalveolar lavage fluid and the expression of inflammatory (tumor necrosis factor alpha (Tnfα), interleukin (Il)-1b, and Il-6) and hypoxic (vascular endothelial growth factor alpha (Vegfα), ankyrin repeat domain 37 (Ankrd37)) response genes. However, EA pretreatment markedly prevented the induction of expression of inflammatory and hypoxic response genes in the lungs. Furthermore, PM exposure significantly triggered hyperactivity by increasing the total moving distance with an increase in moving speed in the open field test. On the contrary, EA pretreatment significantly prevented PM-induced hyperactivity. In conclusion, dietary intervention with EA may be a potential strategy to prevent PM-induced pathology and activity.
Collapse
Affiliation(s)
- Sunyoung Jeong
- Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sungryong Bae
- Department of Fire Protection and Disaster Management, Chosun University, Gwangju 61452, Republic of Korea
| | - Eui-Cheol Shin
- Department of GreenBio Science/Food Science and Technology, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Jong-Hwa Lee
- Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Republic of Korea
- Research Center for Industrialization of Natural Neutralization, Dankook University, Yongin 16890, Republic of Korea
| |
Collapse
|
8
|
Waterpipe smoke inhalation potentiates cardiac oxidative stress, inflammation, mitochondrial dysfunction, apoptosis and autophagy in experimental hypertension. Biomed Pharmacother 2023; 158:114144. [PMID: 36916396 DOI: 10.1016/j.biopha.2022.114144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/04/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Cigarette smoking worsens the health of hypertensive patients. However, less is known about the actions and underlying mechanisms of waterpipe smoke (WPS) in hypertension. Therefore, we evaluated the effects of WPS inhalation in mice made hypertensive (HT) by infusing angiotensin II for six weeks. On day 14 of the infusion of angiotensin II or vehicle (normotensive; NT), mice were exposed either to air or WPS for four consecutive weeks. Each session was 30 min/day and 5 days/week. In NT mice, WPS increased systolic blood pressure (SBP) compared with NT air-exposed group. SBP increase was elevated in HT+WPS group versus either HT+air or NT+WPS. Similarly, the plasma levels of brain natriuretic peptide, C-reactive protein, 8-isoprostane and superoxide dismutase were increased in HT+WPS compared with either HT+air or NT+WPS. In the heart tissue, several markers of oxidative stress and inflammation were increased in HT+WPS group vs the controls. Furthermore, mitochondrial dysfunction in HT+WPS group was more affected than in the HT+air or HT+WPS groups. WPS inhalation in HT mice significantly increased cardiac DNA damage, cleaved caspase 3, expression of the autophagy proteins beclin 1 and microtubule-associated protein light chain 3B, and phosphorylated nuclear factor κ B, compared with the controls. Compared with HT+air mice, heart histology of WPS-exposed HT mice showed increased cardiomyocyte damage, neutrophilic and lymphocytic infiltration and focal fibrosis. We conclude that, in HT mice, WPS inhalation worsened hypertension, cardiac oxidative stress, inflammation, mitochondrial dysfunction, DNA damage, apoptosis and autophagy. The latter effects were associated with a mechanism involving NF-κB activation.
Collapse
|
9
|
Jeong S, Bae S, Yu D, Yang HS, Yang MJ, Lee JH, Ha JH. Dietary Intervention with Quercetin Attenuates Diesel Exhaust Particle-Instilled Pulmonary Inflammation and Behavioral Abnormalities in Mice. J Med Food 2023; 26:93-103. [PMID: 36723487 DOI: 10.1089/jmf.2022.k.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Exposure to diesel exhaust particles (DEPs) is inevitable and closely linked with increased health hazards, causing pulmonary abnormalities by increasing inflammation, hypoxia, and so on. Moreover, long-term exposure to DEPs may trigger whole-body toxicity with behavioral alterations. Therefore, nutritional intervention with natural components may be desirable to prevent and/or ameliorate DEP-inducible pathophysiology in mammals. Quercetin has been demonstrated to reduce metabolic complications by possessing antioxidative, anti-inflammatory, and antimutagenic effects. In this study, we investigated the effects of quercetin on pulmonary inflammation and behavioral alteration in male C57BL/6 mice against DEP instillation. The experimental mice were separated into four treatment groups (n = 8 per group), which include: vehicle control, DEP instillation, dietary intervention with a low dose of quercetin (20 mg/kg) for 14 days with DEP instillation for 7 days, or dietary intervention with a high dose of quercetin (100 mg/kg) for 14 days with DEP instillation for 7 days. Compared with the DEP-instilled group, dietary intervention with quercetin significantly attenuated eosinophils in the bronchoalveolar lavage fluid analysis, pulmonary cytokine, and hypoxic mRNA expressions regardless of quercetin concentrations. DEP instillation triggered hyperactivities in the experimental mice, while quercetin pretreatment successfully normalized DEP-inducible abnormalities regardless of the dosage. Therefore, dietary intervention with quercetin may be an applicable means to prevent DEP-triggered pulmonary and behavioral abnormalities.
Collapse
Affiliation(s)
- Sunyoung Jeong
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Korea.,Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon, Korea
| | - Sungryong Bae
- Department of Fire Protection and Disaster Management, Chosun University, Gwangju, Korea
| | - Daeung Yu
- Department of Food and Nutrition, Changwon National University, Changwon, Korea.,Interdisciplinary Program in Senior Human-Ecology, Major in Food and Nutrition, Changwon National University, Changwon, Korea
| | - Hyo-Seon Yang
- Inhalation Toxicology Center for Airborne Risk Factors, Korea Institute of Toxicology, Jeongeup, Korea
| | - Mi-Jin Yang
- Jeonbuk Pathology Research Group, Korea Institute of Toxicology, Jeongeup, Korea
| | - Jong-Hwa Lee
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Korea.,Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon, Korea
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan, Korea.,Research Center for Industrialization of Natural Neutralization, Dankook University, Yongin, Korea
| |
Collapse
|
10
|
Sharifi-Rad J, Herrera-Bravo J, Kamiloglu S, Petroni K, Mishra AP, Monserrat-Mesquida M, Sureda A, Martorell M, Aidarbekovna DS, Yessimsiitova Z, Ydyrys A, Hano C, Calina D, Cho WC. Recent advances in the therapeutic potential of emodin for human health. Biomed Pharmacother 2022; 154:113555. [PMID: 36027610 DOI: 10.1016/j.biopha.2022.113555] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/04/2022] [Accepted: 08/14/2022] [Indexed: 01/01/2023] Open
Abstract
Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is a bioactive compound, a natural anthraquinone aglycone, present mainly in herbaceous species of the families Fabaceae, Polygonaceae and Rhamnaceae, with a physiological role in protection against abiotic stress in vegetative tissues. Emodin is mainly used in traditional Chinese medicine to treat sore throats, carbuncles, sores, blood stasis, and damp-heat jaundice. Pharmacological research in the last decade has revealed other potential therapeutic applications such as anticancer, neuroprotective, antidiabetic, antioxidant and anti-inflammatory. The present study aimed to summarize recent studies on bioavailability, preclinical pharmacological effects with evidence of molecular mechanisms, clinical trials and clinical pitfalls, respectively the therapeutic limitations of emodin. For this purpose, extensive searches were performed using the PubMed/Medline, Scopus, Google scholar, TRIP database, Springer link, Wiley and SciFinder databases as a search engines. The in vitro and in vivo studies included in this updated review highlighted the signaling pathways and molecular mechanisms of emodin. Because its bioavailability is low, there are limitations in clinical therapeutic use. In conclusion, for an increase in pharmacotherapeutic efficacy, future studies with carrier molecules to the target, thus opening up new therapeutic perspectives.
Collapse
Affiliation(s)
| | - Jesús Herrera-Bravo
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Chile; Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
| | - Senem Kamiloglu
- Department of Food Engineering, Faculty of Agriculture, Bursa Uludag University, 16059 Gorukle, Bursa, Turkey; Science and Technology Application and Research Center (BITUAM), Bursa Uludag University, 16059 Gorukle, Bursa, Turkey
| | - Katia Petroni
- Dipartimento di Bioscienze, Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy.
| | - Abhay Prakash Mishra
- Department of Pharmaceutical Chemistry, H.N.B. Garhwal (A Central) University, Srinagar Garhwal, Uttarakhand 246174, India.
| | - Margalida Monserrat-Mesquida
- Research Group in Community Nutrition and Oxidative Stress, University Research Institute of Health and Health Research Institute of Balearic Islands (IdISBa), University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain.
| | - Antoni Sureda
- Research Group in Community Nutrition and Oxidative Stress, University Research Institute of Health and Health Research Institute of Balearic Islands (IdISBa), University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain.
| | - Miquel Martorell
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Chile; Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción, 4070386 Concepción, Chile.
| | - Dossymbetova Symbat Aidarbekovna
- Almaty Tecnological University, Kazakh-Russian Medical University, Almaty 050012, str. Tole bi 100, Str. Torekulova 71, Kazakhstan.
| | - Zura Yessimsiitova
- Department of Biodiversity and Bioresource, Al-Farabi Kazakh National University, al-Farabi av. 71, 050040 Almaty, Kazakhstan.
| | - Alibek Ydyrys
- Biomedical Research Centre, Al-Farabi Kazakh National University, al-Farabi av. 71, 050040 Almaty, Kazakhstan.
| | - Christophe Hano
- Department of Biological Chemistry, University of Orleans, Eure et Loir Campus, 28000 Chartres, France.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong.
| |
Collapse
|
11
|
Jimoh YA, Lawal AO, Kade IJ, Olatunde DM, Oluwayomi O. Diphenyl diselenide modulate antioxidant status, inflammatory and redox-sensitive genes in diesel exhaust particle-induced neurotoxicity. Chem Biol Interact 2022; 367:110196. [PMID: 36174737 DOI: 10.1016/j.cbi.2022.110196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/03/2022]
Abstract
This study seeks to determine the influence of diphenyl diselenide (DPDSe) on redox status, inflammatory and redox-sensitive genes in diesel exhaust particle (DEP)-induced neurotoxicity in male albino rats. Male Wistar albino rats were administered nasally with DEP (30 and 60 μg/kg) and treated with intraperitoneal administration of 10 mg/kg DPDSe. Non-enzymatic (lipid peroxidation and conjugated diene concentrations) and enzymatic (catalase, superoxide dismutase, glutathione peroxidase) antioxidant indices and activity of acetylcholinesterase enzyme were evaluated in brain tissues of the rats. Furthermore, the expression of genes linked to oxidative stress (HO-1, Nrf2), pro-inflammatory (NF-KB, IL-8, TNF-α) anti-inflammatory (IL-10) and brain-specific (GFAP, ENO-2) genes were also determined. The results indicated that DPDSe caused a notable reduction in the high levels of thiobarbituric acid reactive substances and conjugated diene observed in the brain of DEP-administered rats. DPDSe also reversed the observed reduction in catalase, superoxide dismutase and glutathione peroxidase enzyme activities in the brain of DEP-administered rats. Lastly, the downregulation of genes associated with redox homeostasis, anti-inflammatory and brain-specific genes and upregulation of pro-inflammatory genes observed in the DEP-treated groups were ameliorated by DPDSe. The immediate restoration of altered biochemical conditions and molecular expression in the brain of DEP-treated rats by DPDSe further validates its use as a promising therapeutic candidate for restoring neurotoxicity linked with DEP-induced oxidative stress.
Collapse
Affiliation(s)
- Yomade Ayodeji Jimoh
- Department of Biochemistry, Federal University of Technology, Akure, 340252, Nigeria
| | - Akeem Olalekan Lawal
- Department of Biochemistry, Federal University of Technology, Akure, 340252, Nigeria.
| | - Ige Joseph Kade
- Department of Biochemistry, Federal University of Technology, Akure, 340252, Nigeria
| | | | - Oluwafunso Oluwayomi
- Department of Biochemistry, Federal University of Technology, Akure, 340252, Nigeria
| |
Collapse
|
12
|
Wu X, Ciminieri C, Bos IST, Woest ME, D'Ambrosi A, Wardenaar R, Spierings DCJ, Königshoff M, Schmidt M, Kistemaker LEM, Gosens R. Diesel exhaust particles distort lung epithelial progenitors and their fibroblast niche. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119292. [PMID: 35439594 PMCID: PMC11251497 DOI: 10.1016/j.envpol.2022.119292] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/01/2022] [Accepted: 04/09/2022] [Indexed: 06/14/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung disease characterized by inflammation and impaired tissue regeneration, and is reported as the fourth leading cause of death worldwide by the Centers for Disease Control and Prevention (CDC). Environmental pollution and specifically motor vehicle emissions are known to play a role in the pathogenesis of COPD, but little is still known about the molecular mechanisms that are altered following diesel exhaust particles (DEP) exposure. Here we used lung organoids derived from co-culture of alveolar epithelial progenitors and fibroblasts to investigate the effect of DEP on the epithelial-mesenchymal signaling niche in the distal lung, which is essential for tissue repair. We found that DEP treatment impaired the number as well as the average diameter of both airway and alveolar type of lung organoids. Bulk RNA-sequencing of re-sorted epithelial cells and fibroblasts following organoid co-culture shows that the Nrf2 pathway, which regulates antioxidants' activity, was upregulated in both cell populations in response to DEP; and WNT/β-catenin signaling, which is essential to promote epithelial repair, was downregulated in DEP-exposed epithelial cells. We show that pharmacological treatment with anti-oxidant agents such as N-acetyl cysteine (NAC) or Mitoquinone mesylate (MitoQ) reversed the effect of DEP on organoids growth. Additionally, a WNT/β-catenin activator (CHIR99021) successfully restored WNT signaling and promoted organoid growth upon DEP exposure. We propose that targeting oxidative stress and specific signaling pathways affected by DEP in the distal lung may represent a strategy to restore tissue repair in COPD.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Chiara Ciminieri
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - I Sophie T Bos
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Manon E Woest
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Aquilo BV, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands
| | - Angela D'Ambrosi
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - René Wardenaar
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713AV, Groningen, the Netherlands
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713AV, Groningen, the Netherlands
| | - Melanie Königshoff
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Martina Schmidt
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Aquilo BV, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Aquilo BV, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands.
| |
Collapse
|
13
|
Effects of Anthraquinones on Immune Responses and Inflammatory Diseases. Molecules 2022; 27:molecules27123831. [PMID: 35744949 PMCID: PMC9230691 DOI: 10.3390/molecules27123831] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
The anthraquinones (AQs) and derivatives are widely distributed in nature, including plants, fungi, and insects, with effects of anti-inflammation and anti-oxidation, antibacterial and antiviral, anti-osteoporosis, anti-tumor, etc. Inflammation, including acute and chronic, is a comprehensive response to foreign pathogens under a variety of physiological and pathological processes. AQs could attenuate symptoms and tissue damages through anti-inflammatory or immuno-modulatory effects. The review aims to provide a scientific summary of AQs on immune responses under different pathological conditions, such as digestive diseases, respiratory diseases, central nervous system diseases, etc. It is hoped that the present paper will provide ideas for future studies of the immuno-regulatory effect of AQs and the therapeutic potential for drug development and clinical use of AQs and derivatives.
Collapse
|
14
|
The Nephroprotective Effects of α-Bisabolol in Cisplatin-Induced Acute Kidney Injury in Mice. Biomedicines 2022; 10:biomedicines10040842. [PMID: 35453592 PMCID: PMC9032774 DOI: 10.3390/biomedicines10040842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 01/24/2023] Open
Abstract
Cisplatin (CP) treatment has been long associated with the development of acute kidney injury (AKI) through mechanisms involving inflammation and oxidative stress. α-Bisabolol (BIS), a sesquiterpene alcohol isolated from the essential oil of various plants, including chamomile, has garnered popularity lately due to its antioxidant, anti-inflammatory, and anticancer properties. Therefore, we investigated the nephroprotective effects of BIS in the murine model of CP-induced AKI and the underlying mechanism of action. BALB/c mice were given BIS orally at 25 mg/kg for 7 days. On day 7, they were given a single dose of CP at 20 mg/kg intraperitoneally. BIS treatment continued for 3 more days. The animals were sacrificed at the end of the experiment (day 11). Kidneys, plasma, and urine were collected, and subsequently, various physiological, biochemical, and histological parameters were assessed. BIS has significantly normalized the alterations of water intake, urine volume, relative kidney weight, and the concentrations of urea and creatinine, as well as the creatinine clearance induced by CP treatment. BIS significantly mitigated the effects of CP-induced kidney injury by reducing kidney injury molecule-1, neutrophil gelatinase-associated lipocalin, adiponectin, and cystatin C. Likewise, the renal concentrations of proinflammatory cytokines, tumor necrosis factor α, interleukin (IL)-6 and IL-1β that were elevated in CP group were significantly reduced in mice treated with BIS and CP. A similar significant reduction was also observed in the CP-induced augmented levels of markers of oxidative stress, as well as the metabolite pteridine. Moreover, BIS significantly reduced the CP–induced renal DNA damage, and markedly lessened the acute tubular necrosis observed in kidney histology. Additionally, BIS significantly reduced the CP-induced increase in the phosphorylated nuclear factor κB (NFκB) in the kidney. These data strongly suggest that BIS exerts a protective action against CP-induced nephrotoxicity by mitigating inflammation and oxidative stress through the inhibition of NFκB activation. No overt adverse effects were noted with BIS treatment. Additional investigations should be done to consider BIS as an efficacious nephroprotective agent against CP.
Collapse
|
15
|
Ferdous Z, Beegam S, Zaaba NE, Elzaki O, Tariq S, Greish YE, Ali BH, Nemmar A. Exacerbation of Thrombotic Responses to Silver Nanoparticles in Hypertensive Mouse Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2079630. [PMID: 35111278 PMCID: PMC8802099 DOI: 10.1155/2022/2079630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022]
Abstract
With advent of nanotechnology, silver nanoparticles, AgNPs owing majorly to their antibacterial properties, are used widely in food industry and biomedical applications implying human exposure by various routes including inhalation. Several reports have suggested AgNPs induced pathophysiological effects in a cardiovascular system. However, cardiovascular diseases such as hypertension may interfere with AgNPs-induced response, yet majority of them are understudied. The aim of this work was to evaluate the thrombotic complications in response to polyethylene glycol- (PEG-) coated AgNPs using an experimental hypertensive (HT) mouse model. Saline (control) or PEG-AgNPs (0.5 mg/kg) were intratracheally (i.t.) instilled four times, i.e., on days 7, 14, 21, and 28 post-angiotensin II-induced HT, or vehicle (saline) infusion. On day 29, various parameters were assessed including thrombosis in pial arterioles and venules, platelet aggregation in whole blood in vitro, plasma markers of coagulation, and fibrinolysis and systemic oxidative stress. Pulmonary exposure to PEG-AgNPs in HT mice induced an aggravation of in vivo thrombosis in pial arterioles and venules compared to normotensive (NT) mice exposed to PEG-AgNPs or HT mice given saline. The prothrombin time, activated partial thromboplastin time, and platelet aggregation in vitro were exacerbated after exposure to PEG-AgNPs in HT mice compared with either NT mice exposed to nanoparticles or HT mice exposed to saline. Elevated concentrations of fibrinogen, plasminogen activator inhibitor-1, and von Willebrand factor were seen after the exposure to PEG-AgNPs in HT mice compared with either PEG-AgNPs exposed NT mice or HT mice given with saline. Likewise, the plasma levels of superoxide dismutase and nitric oxide were augmented by PEG-AgNPs in HT mice compared with either NT mice exposed to nanoparticles or HT mice exposed to saline. Collectively, these results demonstrate that PEG-AgNPs can potentially exacerbate the in vivo and in vitro procoagulatory and oxidative stress effect in HT mice and suggest that population with hypertension are at higher risk of the toxicity of PEG-AgNPs.
Collapse
Affiliation(s)
- Zannatul Ferdous
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Nur E. Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Saeed Tariq
- Department of Anatomy, College of Medicine and Health Science, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Yaser E. Greish
- Department of Chemistry, College of Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Badreldin H. Ali
- Department of Pharmacology and Clinical Pharmacy, Sultan Qaboos University, P.O. Box 35, Muscat 123, Al-Khod, Oman
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, UAE
| |
Collapse
|
16
|
Nemmar A, Beegam S, Zaaba NE, Alblooshi S, Alseiari S, Ali BH. The Salutary Effects of Catalpol on Diesel Exhaust Particles-Induced Thrombogenic Changes and Cardiac Oxidative Stress, Inflammation and Apoptosis. Biomedicines 2022; 10:99. [PMID: 35052780 PMCID: PMC8773344 DOI: 10.3390/biomedicines10010099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022] Open
Abstract
Inhaled particulate air pollution exerts pulmonary inflammation and cardiovascular toxicity through secondary systemic effects due to oxidative stress and inflammation. Catalpol, an iridiod glucoside, extracted from the roots of Rehmannia glutinosa Libosch, has been reported to possess anti-inflammatory and antioxidant properties. Yet, the potential ameliorative effects of catalpol on particulate air pollution-induced cardiovascular toxicity, has not been studied so far. Hence, we evaluated the possible mitigating mechanism of catalpol (5 mg/kg) which was administered to mice by intraperitoneal injection one hour before the intratracheal (i.t.) administration of a relevant type of pollutant particle, viz. diesel exhaust particles (DEPs, 30 µg/mouse). Twenty-four hours after the lung deposition of DEPs, several cardiovascular endpoints were evaluated. DEPs caused a significant shortening of the thrombotic occlusion time in pial microvessels in vivo, induced platelet aggregation in vitro, and reduced the prothrombin time and the activated partial thromboplastin time. All these actions were effectively mitigated by catalpol pretreatment. Likewise, catalpol inhibited the increase of the plasma concentration of C-reactive proteins, fibrinogen, plasminogen activator inhibitor-1 and P- and E-selectins, induced by DEPs. Moreover, in heart tissue, catalpol inhibited the increase of markers of oxidative (lipid peroxidation and superoxide dismutase) and nitrosative (nitric oxide) stress, and inflammation (tumor necrosis factor α, interleukin (IL)-6 and IL-1β) triggered by lung exposure to DEPs. Exposure to DEPs also caused heart DNA damage and increased the levels of cytochrome C and cleaved caspase, and these effects were significantly diminished by the catalpol pretreatment. Moreover, catalpol significantly reduced the DEPs-induced increase of the nuclear factor κB (NFκB) in the heart. In conclusion, catalpol significantly ameliorated DEPs-induced procoagulant events and heart oxidative and nitrosative stress, inflammation, DNA damage and apoptosis, at least partly, through the inhibition of NFκB activation.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.B.); (N.E.Z.); (S.A.); (S.A.)
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.B.); (N.E.Z.); (S.A.); (S.A.)
| | - Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.B.); (N.E.Z.); (S.A.); (S.A.)
| | - Salem Alblooshi
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.B.); (N.E.Z.); (S.A.); (S.A.)
| | - Saleh Alseiari
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.B.); (N.E.Z.); (S.A.); (S.A.)
| | - Badreldin H. Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine & Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman;
| |
Collapse
|
17
|
Valderrama A, Zapata MI, Hernandez JC, Cardona-Arias JA. Systematic review of preclinical studies on the neutrophil-mediated immune response to air pollutants, 1980-2020. Heliyon 2022; 8:e08778. [PMID: 35128092 PMCID: PMC8810373 DOI: 10.1016/j.heliyon.2022.e08778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/24/2021] [Accepted: 01/12/2022] [Indexed: 12/11/2022] Open
Abstract
Preclinical evidence about the neutrophil-mediated response in exposure to air pollutants is scattered and heterogeneous. This has prevented the consolidation of this research field around relevant models that could advance towards clinical research. The purpose of this study was to systematic review the studies of the neutrophils response to air pollutants, following the recommendations of the Cochrane Collaboration and the PRISMA guide, through 54 search strategies in nine databases. We include 234 studies (in vitro, and in vivo), being more frequent using primary neutrophils, Balb/C and C57BL6/J mice, and Sprague-Dawley and Wistar rats. The most frequent readouts were cell counts, cytokines and histopathology. The temporal analysis showed that in the last decade, the use of mice with histopathological and cytokine measurement have predominated. This systematic review has shown that study of the neutrophils response to air pollutants started 40 years ago, and composed of 100 different preclinical models, 10 pollutants, and 11 immunological outcomes. Mechanisms of neutrophils-mediated immunopathology include cellular activation, ROS production, and proinflammatory effects, leading to cell-death, oxidative stress, and inflammatory infiltrates in lungs. This research will allow consolidating the research efforts in this field, optimizing the study of causal processes, and facilitating the advance to clinical studies.
Collapse
Affiliation(s)
- Andrés Valderrama
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Colombia
| | - Maria Isabel Zapata
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Colombia
| | - Juan C. Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Colombia
| | | |
Collapse
|
18
|
Nemmar A, Beegam S, Yuvaraju P, Yasin J, Ali BH, Adeghate E. Nose-Only Water-Pipe Smoke Exposure in Mice Elicits Renal Histopathological Alterations, Inflammation, Oxidative Stress, DNA Damage, and Apoptosis. Front Physiol 2020; 11:46. [PMID: 32116758 PMCID: PMC7026484 DOI: 10.3389/fphys.2020.00046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of water-pipe tobacco smoking is increasing worldwide, and is relatively high among youth and young adults. Exposure to water-pipe smoke (WPS) has been reported to affect various systems including the respiratory, cardiovascular and reproductive systems. However, the impact of WPS exposure on the kidney has received only scant attention. Here, we assessed the effect of nose-only WPS exposure for one or four consecutive weeks on renal histology, inflammation, oxidative stress, DNA damage, and apoptosis. The duration of the session was 30 min/day and 5 days/week. Control mice were exposed to air. Light and electron microcopy analysis revealed that the WPS exposure (especially at 4-week time point) caused degeneration of the endothelial cells of the glomerular capillaries and vacuolar degenerations of the proximal convoluted tubules. WPS exposure also significantly decreased the creatinine clearance, and significantly increased proteinuria and urinary kidney injury molecule-1 (KIM-1) concentration. Kidney lipid peroxidation, reactive oxygen species, and oxidized glutathione were significantly increased. WPS exposure also affected the concentration of reduced glutathione and the activity of catalase. Likewise, renal concentrations of interleukin (IL)-6, IL-1β and KIM-1 were augmented by WPS exposure. Moreover, WPS caused DNA damage as evaluated by comet assay, and increased the expression of cleaved caspase-3 and cytochrome C in the kidney. We conclude that exposure of mice to WPS caused renal histopathological alterations, inflammation, oxidative stress, DNA damage, and apoptosis. If the latter findings could be substantiated by controlled human studies, it would be an additional cause for disquiet about an established public health concern.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Priya Yuvaraju
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Javed Yasin
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
19
|
Comparative Study on Pulmonary Toxicity in Mice Induced by Exposure to Unflavoured and Apple- and Strawberry-Flavoured Tobacco Waterpipe Smoke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6450450. [PMID: 32025277 PMCID: PMC6983288 DOI: 10.1155/2020/6450450] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 12/24/2022]
Abstract
The use of flavoured tobacco products in waterpipe smoking (WPS) has increased its attractiveness and consumption. Nonetheless, the influence of flavourings on pulmonary toxicity caused by WPS remains unclear. Here, the pulmonary toxicity induced by plain (P)-WPS, apple-flavoured (AF)-WPS, and strawberry-flavoured (SF)-WPS (30 minutes/day, 5 days/week for 1 month) was investigated in mice. Control mice were exposed to air. Exposure to P-WPS or AF-WPS or SF-WPS induced a dose-dependent increase of airway hyperreactivity to methacholine. The histological evaluation of the lungs in all the WPS groups revealed the presence focal areas of dilated alveolar spaces and foci of widening of interalveolar spaces with inflammatory cells. In the lung, the activity of neutrophil elastase and myeloperoxidase and the concentrations of tumor necrosis factor-α and glutathione were increased by the exposure to P-WPS, AF-WPS, or SF-WPS. However, the levels of interleukin-6 and catalase were only increased in the AF-WPS and SF-WPS groups, while nitric oxide activity was only increased in the SF-WPS group. DNA injury was increased in all the WPS groups, but the concentration of cleaved caspase-3 was only elevated in the SF-WPS group. The exposure to either P-WPS or AF-WPS or SF-WPS increased the expression of nuclear factor kappa-B (NF-κB) in the lung. In conclusion, the exposure to P-WPS or AF-WPS or SF-WPS induces alterations in lung function and morphology and causes oxidative stress and inflammation via mechanisms that include activation of NF-κB. Overall, the toxicity of flavoured tobacco WPS, in particular SF-WPS, was found to be greater than that of unflavoured WPS.
Collapse
|
20
|
Song Y, Cui X, Zhao R, Hu L, Li Y, Liu C. Emodin protects against lipopolysaccharide-induced inflammatory injury in HaCaT cells through upregulation of miR-21. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2654-2661. [PMID: 31250665 DOI: 10.1080/21691401.2019.1629951] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background/aim: Pressure ulcers are a disastrous health issue in which inflammation is involved. Emodin possesses biological properties in inflammation. Our study investigated functions of emodin in lipopolysaccharide (LPS)-treated HaCaT cells. Methods: LPS was used to induce cell inflammation. MTT and flow cytometry were applied for cell viability and apoptosis assays, respectively. Moreover, apoptotic proteins were detected by western blot. Similarly, inflammatory factors and signalling related proteins were also determined by western blot. Results: Emodin increased cell viability and diminished apoptosis in LPS-treated HaCaT cells. Moreover, cleaved-PARP, cleaved-caspase-3 and cleaved-caspase-9 were all downregulated by emodin. Furthermore, inflammatory factors IL-1β, IL-6, Cox-2 and iNOS were inhibited by emodin in LPS-treated cells. In addition, emodin decreased phosphorylation of p65 and IκBα and the level of PTEN while enhanced phosphorylation of PI3K and AKT. Importantly, emodin increased expression of miR-21 suppressed by LPS and miR-21 downregulation negated the protective functions of emodin. Conclusions: Emodin promoted cell growth presented by increasing viability and blocking apoptosis process with inflammation inhibition. The protective activity of emodin was mediated by miR-21 up-regulation.
Collapse
Affiliation(s)
- Yanping Song
- a Department of Health Management, Heze Medical College , Heze , China
| | - Xueling Cui
- b Department of Breast and Thyroid Surgery, Heze Municipal Hospital , Heze , China
| | - Ruilan Zhao
- c Department of General Medicine, Heze Municipal Hospital , Heze , China
| | - Lanying Hu
- d Department of Joint Surgery, Heze Municipal Hospital , Heze , China
| | - Yanjun Li
- e e Department of Nursing, Heze Medical College , Heze , China
| | - Cuiling Liu
- b Department of Breast and Thyroid Surgery, Heze Municipal Hospital , Heze , China
| |
Collapse
|
21
|
Ji S, Bai Q, Wu X, Zhang DW, Wang S, Shen JL, Fei GH. Unique synergistic antiviral effects of Shufeng Jiedu Capsule and oseltamivir in influenza A viral-induced acute exacerbation of chronic obstructive pulmonary disease. Biomed Pharmacother 2019; 121:109652. [PMID: 31734578 DOI: 10.1016/j.biopha.2019.109652] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/25/2019] [Accepted: 11/06/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The aim of the present study was to investigate the synergistic effects and interactive mechanisms of Shufeng Jiedu Capsule (SFJDC) combined with oseltamivir in the treatment of acute exacerbation of chronic obstructive pulmonary disease (AECOPD) induced by the influenza A virus (IAV). METHODS The extraction of SFJDC was analyzed by UHPLC/ESI Q-Orbitrap Mass Spectrometry. Human bronchial epithelial cells were isolated from COPD (DHBE) bronchial tissues, co-cultured with IAV for 24 h, and were subsequently treated with SFJDC and/or oseltamivir. Cell viability was detected by MTT assay. A rat model of COPD with IAV infection was established and treated with SFJDC and/or oseltamivir. Interleukin (IL)-1β and IL-18 in serum and bronchoalveolar lavage fluid (BALF) were measured by ELISA. Additionally, mRNA and protein levels of NLRP3 inflammasome pathway were measured by quantitative real-time PCR and Western blotting, respectively. RESULTS SFJDC and/or oseltamivir, at their optimal concentrations, had no significant cytotoxicity against DHBEs. The levels of NLRP3-inflammasome-associated components were significantly elevated after cells were inoculated with IAV, whereas the mRNA and protein levels of these components were significantly decreased after treatment with SFJDC and/or oseltamivir in vitro. Moreover, in vivo, the combination of SFJDC and oseltamivir improved survival rates, attenuated clinical symptoms, induced weight gain, alleviated lung damage, and significantly reduced IL-1β and IL-18 levels in serum and BALF, as well as reduced the expression levels of NLRP3-associated components and viral titers in lung homogenates. CONCLUSION SFJDC combined with oseltamivir treatment significantly attenuated IAV-induced airway inflammation and lung viral titers. Hence, our findings may provide a novel therapeutic strategy for IAV-induced respiratory infection.
Collapse
Affiliation(s)
- Shuang Ji
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, PR China
| | - Qin Bai
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, PR China
| | - Xu Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, PR China
| | - Da-Wei Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, PR China
| | - Sheng Wang
- The Center for Scientific Research of Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Ji-Long Shen
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Guang-He Fei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, PR China.
| |
Collapse
|
22
|
Honda A, Ito S, Tanaka M, Sawahara T, Hayashi T, Fukushima W, Kitamura G, Kudo H, Chowdhury PH, Okano H, Onishi T, Kawaryu Y, Higashihara M, Nakayama H, Ueda K, Takano H. Extract of curry powder and its components protect against diesel exhaust particle-induced inflammatory responses in human airway epithelial cells. FOOD AGR IMMUNOL 2019. [DOI: 10.1080/09540105.2019.1682526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Akiko Honda
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Sho Ito
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Michitaka Tanaka
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Takahiro Sawahara
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Tomohiro Hayashi
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Wataru Fukushima
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Gaku Kitamura
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Hitomi Kudo
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Pratiti Home Chowdhury
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Hitoshi Okano
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Toshinori Onishi
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yusuke Kawaryu
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Makoto Higashihara
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Hideki Nakayama
- Research & Development Institute, House Foods Corporation, Yotsukaido, Japan
| | - Kayo Ueda
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Hirohisa Takano
- Environmental Health Division, Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| |
Collapse
|
23
|
Nemmar A, Al-Salam S, Beegam S, Yuvaraju P, Ali BH. Aortic Oxidative Stress, Inflammation and DNA Damage Following Pulmonary Exposure to Cerium Oxide Nanoparticles in a Rat Model of Vascular Injury. Biomolecules 2019; 9:biom9080376. [PMID: 31426470 PMCID: PMC6722935 DOI: 10.3390/biom9080376] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 12/21/2022] Open
Abstract
Pulmonary exposure to cerium oxide nanoparticles (CeO2 NPs) can occur either at the workplace, or due to their release in the environment. Inhaled CeO2 NPs are known to cross the alveolar–capillary barrier and reach various parts of the body, including the vasculature. The anticancer drug cisplatin (CP) causes vascular damage. However, the effects CeO2 NPs on vascular homeostasis in a rat model of CP-induced vascular injury remain unclear. Here, we assessed the impact and underlying mechanism of pulmonary exposure to CeO2 NPs on aorta in rats given a single intraperitoneal injection of cisplatin (CP, 6 mg/kg) to induce vascular damage. Six days later, the rats were intratracheally instilled with either CeO2 NPs (1 mg/kg) or saline (control), and various variables were studied 24 h thereafter in the aortic tissue. The concentration of reduced glutathione and the activity of catalase were significantly increased in the CP + CeO2 NPs group compared with both the CP + saline and the CeO2 NPs groups. The activity of superoxide dismutase was significantly decreased in the CP + CeO2 NPs group compared with both the CP + saline and CeO2 NPs groups. The expression of nuclear factor erythroid-derived 2-like 2 (Nrf2) by the nuclei of smooth muscles and endocardial cells assessed by immunohistochemistry was significantly augmented in CeO2 NPs versus saline, in CP + saline versus saline, and in CP + CeO2 NPs versus CeO2 NPs. Moreover, the concentrations of total nitric oxide, lipid peroxidation and 8-hydroxy-2-deoxyguanosine were significantly elevated in the CP + CeO2 NPs group compared with both the CP + saline and the CeO2 NPs groups. Similarly, compared with both the CP + saline and CeO2 NPs groups, the combination of CP and CeO2 NPs significantly elevated the concentrations of interleukin-6 and tumour necrosis factor-α. Additionally, aortic DNA damage assessed by Comet assay was significantly increased in CeO2 NPs compared with saline, and in CP + saline versus saline, and all these effects were significantly aggravated by the combination of CP and CeO2 NPs. We conclude that pulmonary exposure to CeO2 NPs aggravates vascular toxicity in animal model of vascular injury through mechanisms involving oxidative stress, Nrf2 expression, inflammation and DNA damage.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE
- Correspondence:
| | - Suhail Al-Salam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE
| | - Priya Yuvaraju
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE
| | - Badreldin H. Ali
- Department of Pharmacology, College of Medicine & Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Al-Khoud, Oman
| |
Collapse
|
24
|
Acute Exposure to Diesel-Biodiesel Particulate Matter Promotes Murine Lung Oxidative Stress by Nrf2/HO-1 and Inflammation Through the NF-kB/TNF-α Pathways. Inflammation 2019; 42:526-537. [PMID: 30411213 DOI: 10.1007/s10753-018-0910-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Air pollution caused by fuel burning contributes to respiratory impairments that may lead to death. We aimed to investigate the effects of biodiesel (DB) burning in mouse lungs. DB particulate matter was collected from the exhaust pipes of a bus engine. Mice were treated with 250 μg or 1000 μg of DB particulate matter by intranasal instillation over 5 consecutive days. We demonstrated that DB particulate matter penetrated the lung in the 250-μg and 1000-μg groups. In addition, the DB particulate matter number in pulmonary parenchyma was 175-fold higher in the 250-μg group and 300-fold higher in the 1000-μg group compared to control mice. The instillation of DB particulate matter increased the macrophage number and protein levels of TNF-alpha in murine lungs. DB particulate matter enhanced ROS production in both exposed groups and the malondialdehyde levels compared to the control group. The protein expression levels of Nrf2, p-NF-kB, and HO-1 were higher in the 250-μg group and lower in the 1000-μg group than in control mice and the 250-μg group. In conclusion, DB particulate matter instillation promotes oxidative stress by activating the Nrf2/HO-1 and inflammation by p-NF-kB/TNF-alpha pathways.
Collapse
|
25
|
Waterpipe Smoke Exposure Triggers Lung Injury and Functional Decline in Mice: Protective Effect of Gum Arabic. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8526083. [PMID: 31178975 PMCID: PMC6501418 DOI: 10.1155/2019/8526083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/08/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
Abstract
The prevalence of waterpipe (shisha) tobacco smoking has recently seen a substantial increase worldwide and is becoming a public health problem. Both human and animal studies have established that waterpipe smoke (WPS) increases airway reactivity and inflammation. Gum Arabic (GA) is a prebiotic agent that possesses antioxidant and anti-inflammatory properties. However, its effects on lung toxicity induced by WPS exposure are unknown. Thus, the aim of this study was to investigate the possible salutary effects and underlying mechanisms of GA on WPS-induced pulmonary pathophysiologic effects. C57BL/6 mice were exposed to air or WPS (30 minutes/day for one month) with or without GA treatment in drinking water (15%, w/v). Exposure to WPS induced an influx of neutrophil polymorphs in the peribronchiolar and interstitial spaces and an increase of tumor necrosis factor-α and 8-isoprostane, a marker of lipid peroxidation, concentrations in lung homogenates. The latter effects were significantly mitigated by GA treatment. Likewise, the lung DNA damage induced by WPS exposure was prevented by GA administration. Western blot analysis of the lung showed that GA inhibited nuclear factor kappa-B (NF-κB) expression caused by WPS and augmented that of nuclear factor erythroid 2-related factor 2 (Nrf2). Similarly, immunohistochemical analysis of bronchial epithelial cells and alveolar cells showed a parallel and significant increase in the nuclear expression of Nrf2 and cytoplasmic expression of glutathione in mice treated with GA and exposed to WPS. Moreover, GA administration has significantly prevented airway hyperreactivity to methacholine induced by WPS. We conclude that GA administration significantly declined the physiological, histological, biochemical, and molecular indices of lung toxicity caused by WPS exposure, indicating its beneficial respiratory impact. Considering that GA is a safe agent with health benefits in humans, our data suggest its potential usage in waterpipe smokers.
Collapse
|
26
|
Ferdous Z, Al-Salam S, Greish YE, Ali BH, Nemmar A. Pulmonary exposure to silver nanoparticles impairs cardiovascular homeostasis: Effects of coating, dose and time. Toxicol Appl Pharmacol 2019; 367:36-50. [PMID: 30639276 DOI: 10.1016/j.taap.2019.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 12/16/2022]
Abstract
Pulmonary exposure to silver nanoparticles (AgNPs) revealed the potential of nanoparticles to cause pulmonary toxicity, cross the alveolar-capillary barrier, and distribute to remote organs. However, the mechanism underlying the effects of AgNPs on the cardiovascular system remains unclear. Hence, we investigated the cardiovascular mechanisms of pulmonary exposure to AgNPs (10 nm) with varying coatings [polyvinylpyrrolidone (PVP) and citrate (CT)], concentrations (0.05, 0.5 and 5 mg/kg body weight), and time points (1 and 7 days) in BALB/C mice. Silver ions (Ag+) were used as ionic control. Exposure to AgNPs induced lung inflammation. In heart, tumor necrosis factor α, interleukin 6, total antioxidants, reduced glutathione and 8-isoprostane significantly increased for both AgNPs. Moreover, AgNPs caused oxidative DNA damage and apoptosis in the heart. The plasma concentration of fibrinogen, plasminogen activation inhibitor-1 and brain natriuretic peptide were significantly increased for both coating AgNPs. Likewise, the prothrombin time and activated partial thromboplastin time were significantly decreased. Additionally, the PVP- and CT- AgNPs induced a significant dose-dependent increase in thrombotic occlusion time in cerebral microvessels at both time points. In vitro study on mice whole blood exhibited significant platelet aggregation for both particle types. Compared with AgNPs, Ag+ increased thrombogenicity and markers of oxidative stress, but did not induce either DNA damage or apoptosis in the heart. In conclusion, pulmonary exposure to AgNPs caused cardiac oxidative stress, DNA damage and apoptosis, alteration of coagulation markers and thrombosis. Our findings provide a novel mechanistic insight into the cardiovascular pathophysiological effects of lung exposure to AgNPs.
Collapse
Affiliation(s)
- Zannatul Ferdous
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, P.O Box 17666, Al Ain, United Arab Emirates
| | - Yaser E Greish
- Department of Chemistry, College of Science, United Arab Emirates University, P.O. Box 17551, United Arab Emirates
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Al-Khod, Oman
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates.
| |
Collapse
|
27
|
Liang Z, Ren C. Emodin attenuates apoptosis and inflammation induced by LPS through up-regulating lncRNA TUG1 in murine chondrogenic ATDC5 cells. Biomed Pharmacother 2018; 103:897-902. [DOI: 10.1016/j.biopha.2018.04.085] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 12/31/2022] Open
|
28
|
Label-free quantitative proteomics reveals fibrinopeptide B and heparin cofactor II as potential serum biomarkers in respiratory syncytial virus-infected mice treated with Qingfei oral liquid formula. Chin J Nat Med 2018; 16:241-251. [DOI: 10.1016/s1875-5364(18)30054-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Indexed: 01/28/2023]
|
29
|
Exercise Training Mitigates Water Pipe Smoke Exposure-Induced Pulmonary Impairment via Inhibiting NF- κB and Activating Nrf2 Signalling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7459612. [PMID: 29692875 PMCID: PMC5859847 DOI: 10.1155/2018/7459612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 01/16/2018] [Indexed: 12/29/2022]
Abstract
Water pipe smoking is a tobacco smoking method commonly used in Eastern countries and is gaining popularity in Europe and North America, in particular among adolescents and young adults. Several clinical and experimental studies have reported that exposure to water pipe smoke (WPS) induces lung inflammation and impairment of pulmonary function. However, the mechanisms of such effects are not understood, as are data on the possible palliative effect of exercise training. The present study evaluated the effects of regular aerobic exercise training (treadmill: 5 days/week, 40 min/day) on subchronic exposure to WPS (30 minutes/day, 5 days/week for 2 months). C57BL/6 mice were exposed to air or WPS with or without exercise training. Airway resistance measured using forced oscillation technique was significantly and dose-dependently increased in the WPS-exposed group when compared with the air-exposed one. Exercise training significantly prevented the effect of WPS on airway resistance. Histologically, the lungs of WPS-exposed mice had focal moderate interstitial inflammatory cell infiltration consisting of neutrophil polymorphs, plasma cells, and lymphocytes. There was a mild increase in intra-alveolar macrophages and a focal damage to alveolar septae in some foci. Exercise training significantly alleviated these effects and also decreased the WPS-induced increase of tumor necrosis factor α and interleukin 6 concentrations and attenuated the increase of 8-isoprostane in lung homogenates. Likewise, the lung DNA damage induced by WPS was significantly inhibited by exercise training. Moreover, exercise training inhibited nuclear factor kappa-B (NF-κB) expression induced by WPS and increased that of nuclear factor erythroid 2-related factor 2 (Nrf2). Our findings suggest that exercise training significantly mitigated WPS-induced increase in airway resistance, inflammation, oxidative stress, and DNA damage via mechanisms that include inhibiting NF-κB and activating Nrf2 signalling pathways.
Collapse
|
30
|
In Vivo Protective Effects of Nootkatone against Particles-Induced Lung Injury Caused by Diesel Exhaust Is Mediated via the NF-κB Pathway. Nutrients 2018; 10:nu10030263. [PMID: 29495362 PMCID: PMC5872681 DOI: 10.3390/nu10030263] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/18/2018] [Accepted: 01/23/2018] [Indexed: 12/21/2022] Open
Abstract
Numerous studies have shown that acute particulate air pollution exposure is linked with pulmonary adverse effects, including alterations of pulmonary function, inflammation, and oxidative stress. Nootkatone, a constituent of grapefruit, has antioxidant and anti-inflammatory effects. However, the effect of nootkatone on lung toxicity has not been reported so far. In this study we evaluated the possible protective effects of nootkatone on diesel exhaust particles (DEP)-induced lung toxicity, and the possible mechanisms underlying these effects. Mice were intratracheally (i.t.) instilled with either DEP (30 µg/mouse) or saline (control). Nootkatone was given to mice by gavage, 1 h before i.t. instillation, with either DEP or saline. Twenty-four hours following DEP exposure, several physiological and biochemical endpoints were assessed. Nootkatone pretreatment significantly prevented the DEP-induced increase in airway resistance in vivo, decreased neutrophil infiltration in bronchoalveolar lavage fluid, and abated macrophage and neutrophil infiltration in the lung interstitium, assessed by histolopathology. Moreover, DEP caused a significant increase in lung concentrations of 8-isoprostane and tumor necrosis factor α, and decreased the reduced glutathione concentration and total nitric oxide activity. These actions were all significantly alleviated by nootkatone pretreatment. Similarly, nootkatone prevented DEP-induced DNA damage and prevented the proteolytic cleavage of caspase-3. Moreover, nootkatone inhibited nuclear factor-kappaB (NF-κB) induced by DEP. We conclude that nootkatone prevented the DEP-induced increase in airway resistance, lung inflammation, oxidative stress, and the subsequent DNA damage and apoptosis through a mechanism involving inhibition of NF-κB activation. Nootkatone could possibly be considered a beneficial protective agent against air pollution-induced respiratory adverse effects.
Collapse
|
31
|
Nemmar A, Karaca T, Beegam S, Yuvaraju P, Yasin J, Ali BH. Lung Oxidative Stress, DNA Damage, Apoptosis, and Fibrosis in Adenine-Induced Chronic Kidney Disease in Mice. Front Physiol 2017; 8:896. [PMID: 29218013 PMCID: PMC5703828 DOI: 10.3389/fphys.2017.00896] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022] Open
Abstract
It is well-established that there is a crosstalk between the lung and the kidney, and several studies have reported association between chronic kidney disease (CKD) and pulmonary pathophysiological changes. Experimentally, CKD can be caused in mice by dietary intake of adenine. Nevertheless, the consequence of such intervention on the lung received only scant attention. Here, we assessed the pulmonary effects of adenine (0.2% w/w in feed for 4 weeks)-induced CKD in mice by assessing various physiological histological and biochemical endpoints. Adenine treatment induced a significant increase in urine output, urea and creatinine concentrations, and it decreased the body weight and creatinine clearance. It also increased proteinuria and the urinary levels of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin. Compared with control group, the histopathological evaluation of lungs from adenine-treated mice showed polymorphonuclear leukocytes infiltration in alveolar and bronchial walls, injury, and fibrosis. Moreover, adenine caused a significant increase in lung lipid peroxidation and reactive oxygen species and decreased the antioxidant catalase. Adenine also induced DNA damage assessed by COMET assay. Similarly, adenine caused apoptosis in the lung characterized by a significant increase of cleaved caspase-3. Moreover, adenine induced a significant increase in the expression of nuclear factor erythroid 2–related factor 2 (Nrf2) in the lung. We conclude that administration of adenine in mice induced CKD is accompanied by lung oxidative stress, DNA damage, apoptosis, and Nrf2 expression and fibrosis.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Turan Karaca
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Priya Yuvaraju
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Javed Yasin
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
32
|
Wang H, Duan H, Meng T, Yang M, Cui L, Bin P, Dai Y, Niu Y, Shen M, Zhang L, Zheng Y, Leng S. Local and Systemic Inflammation May Mediate Diesel Engine Exhaust–Induced Lung Function Impairment in a Chinese Occupational Cohort. Toxicol Sci 2017; 162:372-382. [DOI: 10.1093/toxsci/kfx259] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Haitao Wang
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Huawei Duan
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Tao Meng
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Mo Yang
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Lianhua Cui
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Ping Bin
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yufei Dai
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yong Niu
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Meili Shen
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Liping Zhang
- Department of Environmental Health, Faculty of Public Health, Weifang Medical University, Weifang 261053, China
| | - Yuxin Zheng
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Shuguang Leng
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266021, China
| |
Collapse
|
33
|
Taurine ameliorates particulate matter-induced emphysema by switching on mitochondrial NADH dehydrogenase genes. Proc Natl Acad Sci U S A 2017; 114:E9655-E9664. [PMID: 29078374 PMCID: PMC5692577 DOI: 10.1073/pnas.1712465114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Exposure to high levels of particulate matter (PM) poses a major threat to human health. Cigarette smoke is the most common irritant that causes chronic obstructive pulmonary disease (COPD); however, at least one-fourth of patients with COPD are nonsmokers, and their disease is largely attributed to air pollution. The occurrence of pollution episodes in China has raised an emergent question of how PM leads to the pathogenesis of COPD. In this paper, we show that deregulation of mitochondrial NADH dehydrogenase gene expression levels plays a key role in the aggravation of COPD during air pollutant exposure, which can be rescued by taurine and 3-MA treatments in both mammalian cells and animals. Chronic obstructive pulmonary disease (COPD) has been linked to particulate matter (PM) exposure. Using transcriptomic analysis, we demonstrate that diesel exhaust particles, one of the major sources of particulate emission, down-regulated genes located in mitochondrial complexes I and V and induced experimental COPD in a mouse model. 1-Nitropyrene was identified as a major toxic component of PM-induced COPD. In the panel study, COPD patients were found to be more susceptible to PM than individuals with normal lung function due to an increased inflammatory response. Mechanistically, exposure to PM in human bronchial epithelial cells led to a decline in CCAAT/enhancer-binding protein alpha (C/EBPα), which triggered aberrant expression of NADH dehydrogenase genes and ultimately led to enhanced autophagy. ATG7-deficient mice, which have lower autophagy rates, were protected from PM-induced experimental COPD. Using metabolomics analysis, we further established that treatment with taurine and 3-methyladenine completely restored mitochondrial gene expression levels, thereby ameliorating the PM-induced emphysema. Our studies suggest a potential therapeutic intervention for the C/EBPα/mitochondria/autophagy axis in PM-induced COPD.
Collapse
|
34
|
Role of tumor necrosis factor-α and its receptors in diesel exhaust particle-induced pulmonary inflammation. Sci Rep 2017; 7:11508. [PMID: 28912506 PMCID: PMC5599628 DOI: 10.1038/s41598-017-11991-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/29/2017] [Indexed: 01/05/2023] Open
Abstract
Inhalation of diesel exhaust particles (DEP) induces an inflammatory reaction in the lung. However, the underlying mechanisms remain to be elucidated. Tumor necrosis factor alpha (TNF-α) is a pro-inflammatory cytokine that operates by binding to tumor necrosis factor receptor 1 (TNFR1) and tumor necrosis factor receptor 2 (TNFR2). The role of TNF-α signaling and the importance of either TNFR1 or TNFR2 in the DEP-induced inflammatory response has not yet been elucidated. TNF-α knockout (KO), TNFR1 KO, TNFR2 KO, TNFR1/TNFR2 double KO (TNFR-DKO) and wild type (WT) mice were intratracheally exposed to saline or DEP. Pro-inflammatory cells and cytokines were assessed in the bronchoalveolar lavage fluid (BALF). Exposure to DEP induced a dose-dependent inflammation in the BALF in WT mice. In addition, levels of TNF-α and its soluble receptors were increased upon exposure to DEP. The DEP-induced inflammation in the BALF was decreased in TNF-α KO, TNFR-DKO and TNFR2 KO mice. In contrast, the inflammatory response in the BALF of DEP-exposed TNFR1 KO mice was largely comparable with WT controls. In conclusion, these data provide evidence for a regulatory role of TNF-α in DEP-induced pulmonary inflammation and identify TNFR2 as the most important receptor in mediating these inflammatory effects.
Collapse
|
35
|
Gruber-Dorninger C, Novak B, Nagl V, Berthiller F. Emerging Mycotoxins: Beyond Traditionally Determined Food Contaminants. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7052-7070. [PMID: 27599910 DOI: 10.1021/acs.jafc.6b03413] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Modern analytical techniques can determine a multitude of fungal metabolites contaminating food and feed. In addition to known mycotoxins, for which maximum levels in food are enforced, also currently unregulated, so-called "emerging mycotoxins" were shown to occur frequently in agricultural products. The aim of this review is to critically discuss the relevance of selected emerging mycotoxins to food and feed safety. Acute and chronic toxicity as well as occurrence data are presented for enniatins, beauvericin, moniliformin, fusaproliferin, fusaric acid, culmorin, butenolide, sterigmatocystin, emodin, mycophenolic acid, alternariol, alternariol monomethyl ether, and tenuazonic acid. By far not all of the detected compounds are toxicologically relevant at their naturally occurring levels and are therefore of little or no health concern to consumers. Still, gaps in knowledge have been identified for several compounds. These gaps should be closed by the scientific community in the coming years to allow a proper risk assessment.
Collapse
Affiliation(s)
| | - Barbara Novak
- BIOMIN Research Center , Technopark 1, 3430 Tulln, Austria
| | - Veronika Nagl
- BIOMIN Research Center , Technopark 1, 3430 Tulln, Austria
| | - Franz Berthiller
- Christian Doppler Laboratory for Mycotoxin Metabolism and Center for Analytical Chemistry, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU) , Konrad-Lorenz-Strasse 20, 3430 Tulln, Austria
| |
Collapse
|
36
|
Niemann B, Rohrbach S, Miller MR, Newby DE, Fuster V, Kovacic JC. Oxidative Stress and Cardiovascular Risk: Obesity, Diabetes, Smoking, and Pollution: Part 3 of a 3-Part Series. J Am Coll Cardiol 2017; 70:230-251. [PMID: 28683970 DOI: 10.1016/j.jacc.2017.05.043] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 04/25/2017] [Accepted: 05/10/2017] [Indexed: 12/16/2022]
Abstract
Oxidative stress occurs whenever the release of reactive oxygen species (ROS) exceeds endogenous antioxidant capacity. In this paper, we review the specific role of several cardiovascular risk factors in promoting oxidative stress: diabetes, obesity, smoking, and excessive pollution. Specifically, the risk of developing heart failure is higher in patients with diabetes or obesity, even with optimal medical treatment, and the increased release of ROS from cardiac mitochondria and other sources likely contributes to the development of cardiac dysfunction in this setting. Here, we explore the role of different ROS sources arising in obesity and diabetes, and the effect of excessive ROS production on the development of cardiac lipotoxicity. In parallel, contaminants in the air that we breathe pose a significant threat to human health. This paper provides an overview of cigarette smoke and urban air pollution, considering how their composition and biological effects have detrimental effects on cardiovascular health.
Collapse
Affiliation(s)
- Bernd Niemann
- Department of Adult and Pediatric Cardiovascular Surgery, University Hospital Giessen, Giessen, Germany
| | - Susanne Rohrbach
- Institute of Physiology, Justus-Liebig University, Giessen, Germany.
| | - Mark R Miller
- BHF/University of Edinburgh Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David E Newby
- BHF/University of Edinburgh Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Valentin Fuster
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, New York; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
37
|
Li Y, Chang N, Han Y, Zhou M, Gao J, Hou Y, Jiang M, Zhang T, Bai G. Anti-inflammatory effects of Shufengjiedu capsule for upper respiratory infection via the ERK pathway. Biomed Pharmacother 2017; 94:758-766. [PMID: 28802227 DOI: 10.1016/j.biopha.2017.07.118] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/12/2017] [Accepted: 07/24/2017] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Shufengjiedu Capsule (SFJD) is a type of Chinese traditional medicine compound for the treatment of acute upper respiratory tract infection. The present work aims to decipher the mechanism of SFJD. METHODS In this study, we used target prediction and RNA sequence (RNA-Seq) based on transcriptome analysis to clarify the inflammation-eliminating mechanism of SFJD. Firstly, Pseudomonas aeruginosa (PAK) was used to induce acute lung injury in KM mice. After being treated by SFJD, the differently expressed genes were analyzed by RNA-Seq. Secondly, the chemical constituents of SFJD were identified by ultra-performance liquid chromatography quadrupole/time of flight mass spectrometry (UPLC/Q-TOF-MS) and submitted to PharmMapper to predict targets. The Kyoto Encyclopedia of Genes and Genomes (KEGG) and String 9.1 websites were employed to establish the interaction network of inflammation of these targets. RESULTS The results indicated that SFJD alleviated PAK induced lung injury in KM mice. We infer that the mechanism is a complex network containing 15 pathways related to inflammation regulated by 16 types of components from six types of herbs via 29 proteins. The ERK signaling pathway was a key pathway among them, which was predicted to be regulated by 14 types of components in SFJD. Phillyrin, emodin, and verbenalin were screened out by binding capacity, and the synergistic effect of them was further confirmed. CONCLUSIONS Various components of SFJD ameliorated PAK induced upper respiratory tract infection via multiple targets, of which ERK phosphorylation might be the key event regulated specifically by verbenalin, phillyrin and emodin.
Collapse
Affiliation(s)
- Yanmei Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Nianwei Chang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Yanqi Han
- Department of Traditional Chinese Medicine, Tianjin Institute of Pharmaceutical Research Co. Ltd., Tianjin 300193, People's Republic of China
| | - Mengge Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China.
| | - Tiejun Zhang
- Department of Traditional Chinese Medicine, Tianjin Institute of Pharmaceutical Research Co. Ltd., Tianjin 300193, People's Republic of China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| |
Collapse
|
38
|
Chen H, Huang RS, Yu XX, Ye Q, Pan LL, Shao GJ, Pan J. Emodin protects against oxidative stress and apoptosis in HK-2 renal tubular epithelial cells after hypoxia/reoxygenation. Exp Ther Med 2017; 14:447-452. [PMID: 28672952 DOI: 10.3892/etm.2017.4473] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 02/24/2017] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to determine the effects of emodin, a natural compound with antioxidant properties, on oxidative stress and apoptosis induced by hypoxia/reoxygenation (H/R) in HK-2 human renal tubular cells. In HK-2 cells subjected to H/R, it was observed that pre-treatment with emodin lead to an increase in cellular viability and a reduction in the rate of apoptosis and the B-cell lymphoma 2 (Bcl-2)-associated X protein/Bcl-2 ratio. H/R alone caused a significant increase in the levels of reactive oxygen species and malondialdehyde (P<0.05) and a significant decrease in the activities of superoxide dismutase, catalase and glutathione peroxidase (P<0.05), relative to normoxic cells. In turn, parameters of oxidative stress were improved by emodin pre-treatment. In addition, emodin pre-treatment significantly inhibited the phosphorylation of extracellular signal-regulated protein kinase and c-Jun N-terminal kinase mitogen-activated protein kinases (MAPKs) induced by H/R (P<0.05). These data suggest that emodin may prevent H/R-induced apoptosis in human renal tubular cells through the regulation of cellular oxidative stress, MAPK activation and restoration of the Bax/Bcl-2 ratio.
Collapse
Affiliation(s)
- Hui Chen
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Ri-Sheng Huang
- Department of Thoracic Surgery, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Xian-Xian Yu
- Department of Nephrology, Yueqing People's Hospital, Wenzhou, Zhejiang 325600, P.R. China
| | - Qiong Ye
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Lu-Lu Pan
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Guo-Jian Shao
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Jing Pan
- Department of Cadre Health Care, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
39
|
Nemmar A, Al-Salam S, Beegam S, Yuvaraju P, Ali BH. The acute pulmonary and thrombotic effects of cerium oxide nanoparticles after intratracheal instillation in mice. Int J Nanomedicine 2017; 12:2913-2922. [PMID: 28435267 PMCID: PMC5391826 DOI: 10.2147/ijn.s127180] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cerium oxide nanoparticles (CeO2 NPs), used as a diesel fuel catalyst, can be emitted into the ambient air, resulting in exposure to humans by inhalation. Recent studies have reported the development of lung toxicity after pulmonary exposure to CeO2 NPs. However, little is known about the possible thrombotic effects of these NPs. The present study investigated the acute (24 hours) effect of intratracheal (IT) instillation of either CeO2 NPs (0.1 or 0.5 mg/kg) or saline (control) on pulmonary and systemic inflammation and oxidative stress and thrombosis in mice. CeO2 NPs induced a significant increase of neutrophils into the bronchoalveolar lavage (BAL) fluid with an elevation of tumor necrosis factor α (TNFα) and a decrease in the activity of the antioxidant catalase. Lung sections of mice exposed to CeO2 NPs showed a dose-dependent infiltration of inflammatory cells consisting of macrophages and neutrophils. Similarly, the plasma levels of C-reactive protein and TNFα were significantly increased, whereas the activities of catalase and total antioxidant were significantly decreased. Interestingly, CeO2 NPs significantly and dose dependently induced a shortening of the thrombotic occlusion time in pial arterioles and venules. Moreover, the plasma concentrations of fibrinogen and plasminogen activator inhibitor-1 were significantly elevated by CeO2 NPs. The direct addition of CeO2 NPs (1, 5, or 25 μg/mL) to mouse whole blood, collected from the inferior vena cava, in vitro neither caused significant platelet aggregation nor affected prothrombin time or partial thromboplastin time, suggesting that the thrombotic events observed in vivo may have resulted from systemic inflammation and/or oxidative stress induced by CeO2 NPs. This study concludes that acute pulmonary exposure to CeO2 NPs induces pulmonary and systemic inflammation and oxidative stress and promotes thrombosis in vivo.
Collapse
Affiliation(s)
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | | | - Priya Yuvaraju
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Al-Khod, Sultanate of Oman
| |
Collapse
|
40
|
Santos ÊR, Oliveira HN, Oliveira EJ, Azevedo SH, Jesus AA, Medeiros AM, Dariva C, Sousa EM. Supercritical fluid extraction of Rumex Acetosa L. roots: Yield, composition, kinetics, bioactive evaluation and comparison with conventional techniques. J Supercrit Fluids 2017. [DOI: 10.1016/j.supflu.2016.11.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
41
|
Cerium Oxide Nanoparticles in Lung Acutely Induce Oxidative Stress, Inflammation, and DNA Damage in Various Organs of Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9639035. [PMID: 28392888 PMCID: PMC5368370 DOI: 10.1155/2017/9639035] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/26/2017] [Accepted: 02/09/2017] [Indexed: 12/21/2022]
Abstract
CeO2 nanoparticles (CeO2 NPs) which are used as a diesel fuel additive are emitted in the particulate phase in the exhaust, posing a health concern. However, limited information exists regarding the in vivo acute toxicity of CeO2 NPs on multiple organs. Presently, we investigated the acute (24 h) effects of intratracheally instilled CeO2 NPs in mice (0.5 mg/kg) on oxidative stress, inflammation, and DNA damage in major organs including lung, heart, liver, kidneys, spleen, and brain. Lipid peroxidation measured by malondialdehyde production was increased in the lungs only, and reactive oxygen species were increased in the lung, heart, kidney, and brain. Superoxide dismutase activity was decreased in the lung, liver, and kidney, whereas glutathione increased in lung but it decreased in the kidney. Total nitric oxide was increased in the lung and spleen but it decreased in the heart. Tumour necrosis factor-α increased in all organs studied. Interleukin- (IL-) 6 increased in the lung, heart, liver, kidney, and spleen. IL-1β augmented in the lung, heart, kidney, and spleen. Moreover, CeO2 NPs induced DNA damage, assessed by COMET assay, in all organs studied. Collectively, these findings indicate that pulmonary exposure to CeO2 NPs causes oxidative stress, inflammation, and DNA damage in multiple organs.
Collapse
|
42
|
Huang J, Gong W, Chen Z, Huang J, Chen Q, Huang H, Zhao C. Emodin self-emulsifying platform ameliorates the expression of FN, ICAM-1 and TGF-β1 in AGEs-induced glomerular mesangial cells by promoting absorption. Eur J Pharm Sci 2017; 99:128-136. [PMID: 27988328 DOI: 10.1016/j.ejps.2016.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/13/2016] [Accepted: 12/10/2016] [Indexed: 10/20/2022]
Abstract
Emodin, a potential anti-diabetic nephropathy agent, is limited by its oral use due to the poor water solubility. The present study aimed to enhance the absorption and the suppressive effects of emodin on renal fibrosis by developing a self-microemulsifying drug delivery system (SMEDDS). Solubility studies, compatibility tests, pseudo-ternary phase diagrams analysis and central composite design were carried out to obtain the optimized formulation. The average droplet size of emodin-loaded SMEDDS was about 18.31±0.12nm, and the droplet size and zeta potential remained stable at different dilution ratios of water and different values of pH varying from 1.2 to 7.2. Enhanced cellular uptake in both the Caco-2 cells and glomerular mesangial cells (GMCs) is great advantageous for the formulation. The AUC0-24h of emodin-loaded SMEDDS was 1.87-fold greater than that of emodin suspension, which may be attributed to enhanced uptake in Caco-2 cells. Moreover, emodin-loaded SMEDDS showed better suppressive effects on the protein level of fibronectin (FN), transforming growth factor-beta 1 (TGF-β1) and intercellular adhesion molecule 1 (ICAM-1) than the crude emodin in advanced glycation-end products (AGEs)-induced GMCs and renal tubular epithelial cells (NRK-52E). Our study illustrated that developed SMEDDS improved the oral absorption of emodin, and attained better suppressive effects on the protein level of renal fibrosis compositions in AGEs-induced GMCs and NRK-52E cells.
Collapse
Affiliation(s)
- Jiani Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenyan Gong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiquan Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junying Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qiuhong Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Heqing Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
43
|
Liu J, Yang Y, Zeng X, Bo L, Jiang S, Du X, Xie Y, Jiang R, Zhao J, Song W. Investigation of selenium pretreatment in the attenuation of lung injury in rats induced by fine particulate matters. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:4008-4017. [PMID: 27921246 DOI: 10.1007/s11356-016-8173-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 11/28/2016] [Indexed: 06/06/2023]
Abstract
Selenium (Se) is vital for health because of its antioxidative and anti-inflammation functions. The aim of this study was to determine if dietary selenium could inhibit the rat lung injury induced by ambient fine particulate matter (PM2.5). Sprague-Dawley rats were randomly allocated in seven groups (n = 8). The rats in PM2.5 exposure group were intratracheally instilled with 40 mg/kg of body weight (b.w.) of PM2.5 suspension. The rats in Se prevention groups were pretreated with 17.5, 35, or 70 μg/kg b.w. of Se for 4 weeks, respectively. Then, the rats were exposed to 40 mg/kg b.w. of PM2.5 in the fifth week. The bronchoalveolar lavage fluid (BALF) was collected to count the neutrophil numbers and to analyze the cytokines (tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), soluble intercellular adhesion molecule-1 (sICAM-1)) related to inflammation, the markers related to oxidative stress (total antioxidant capacity (T-AOC), total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA)), and the indicators related to cell damage (lactate dehydrogenase (LDH), total protein (TP), alkaline phosphatase (AKP)). The lung lobe that has not undergone bronchoalveolar lavage was processed for light microscopic examination. The results showed that the proportions of neutrophils in the BALF and the pathologic scores of the lung in PM2.5-exposed groups were higher than that in the control group (P < 0.05). Se pretreatment caused a dose-dependent decrease in TNF-α, IL-1β, sICAM-1, LDH, TP, AKP, and MDA when compared with the PM2.5-only exposure group. Meanwhile, the dose-dependent increase in T-AOC, T-SOD, and GSH-Px activities were observed in rats pretreated with Se. In conclusion, Se pretreatment may protect rat lungs against inflammation and oxidative stress induced by PM2.5, which suggests that Se plays an important role as a kind of potential preventative agent to inhibit the PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Jie Liu
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China
| | - Yingying Yang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China
| | - Liang Bo
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China
| | - Shuo Jiang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China
| | - Xihao Du
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China
| | - Yuquan Xie
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200032, China
| | - Rongfang Jiang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China.
| | - Weimin Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Fudan University, 138 Yixueyuan Road, Box 249, Shanghai, 200032, China.
| |
Collapse
|
44
|
Kang S, Zhao X, Yue L, Liu L. Main anthraquinone components in Aloe vera
and their inhibitory effects on the formation of advanced glycation end-products. J FOOD PROCESS PRES 2016. [DOI: 10.1111/jfpp.13160] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Shimo Kang
- The College of Food Science; Shenyang Agricultural University; Shenyang Liaoning 110866 China
| | - Xin Zhao
- The College of Food Science; Shenyang Agricultural University; Shenyang Liaoning 110866 China
| | - Lu Yue
- The College of Food Science; Shenyang Agricultural University; Shenyang Liaoning 110866 China
| | - Ling Liu
- The College of Food Science; Shenyang Agricultural University; Shenyang Liaoning 110866 China
| |
Collapse
|
45
|
Monisha BA, Kumar N, Tiku AB. Emodin and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 928:47-73. [DOI: 10.1007/978-3-319-41334-1_3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
46
|
Nemmar A, Beegam S, Yuvaraju P, Yasin J, Tariq S, Attoub S, Ali BH. Ultrasmall superparamagnetic iron oxide nanoparticles acutely promote thrombosis and cardiac oxidative stress and DNA damage in mice. Part Fibre Toxicol 2016; 13:22. [PMID: 27138375 PMCID: PMC4852430 DOI: 10.1186/s12989-016-0132-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/18/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Ultrasmall superparamagnetic iron oxide nanoparticles (USPIO) are being developed for several biomedical applications including drug delivery and imaging. However, little is known about their possible adverse effects on thrombosis and cardiac oxidative and DNA damage. METHODS Presently, we investigated the acute (1 h) effect of intravenously (i.v.) administered USPIO in mice (0.4, 2 and 10 μg/kg). Diesel exhaust particles (DEP; 400 μg/kg) were used as positive control. RESULTS USPIO induced a prothrombotic effect in pial arterioles and venules in vivo and increased the plasma plasminogen activator inhibitor-1 (PAI-1). Both thrombogenicity and PAI-1 concentration were increased by DEP. The direct addition of USPIO (0.008, 0.04 and 0.2 μg/ml) to untreated mouse blood dose-dependently induced in vitro platelet aggregation. USPIO caused a shortening of activated partial thromboplastin time (aPTT) and prothrombin time (PT). Similarly, DEP administration (1 μg/ml) triggered platelet aggregation in vitro in whole blood. DEP also reduced PT and aPTT. The plasma levels of creatine phosphokinase-MB isoenzyme (CK-MB), lactate dehydrogenase (LDH) and troponin-I were increased by USPIO. DEP induced a significant increase of CK-MB, LDH and troponin I levels in plasma. The cardiac levels of markers of oxidative stress including lipid peroxidation, reactive oxygen species and superoxide dismutase activity were increased by USPIO. Moreover, USPIO caused DNA damage in the heart. Likewise, DEP increased the markers of oxidative stress and induced DNA damage in the heart. CONCLUSION We conclude that acute i.v. administration of USPIO caused thrombosis and cardiac oxidative stress and DNA damage. These findings provide novel insight into the pathophysiological effects of USPIO on cardiovascular homeostasis, and highlight the need for a thorough evaluation of their toxicity.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Departments of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates.
| | - Sumaya Beegam
- Departments of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Priya Yuvaraju
- Departments of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Javed Yasin
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Saeed Tariq
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Samir Attoub
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Badreldin H Ali
- Department of Pharmacology, College of Medicine & Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Al-Khod, Sultanate of Oman
| |
Collapse
|
47
|
Shi Y, Li J, Ren Y, Wang H, Cong Z, Wu G, Du L, Li H, Zhang X. Pharmacokinetics and tissue distribution of emodin loaded nanoemulsion in rats. J Drug Deliv Sci Technol 2015. [DOI: 10.1016/j.jddst.2015.10.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|