151
|
Chinta KC, Rahman MA, Saini V, Glasgow JN, Reddy VP, Lever JM, Nhamoyebonde S, Leslie A, Wells RM, Traylor A, Madansein R, Siegal GP, Antony VB, Deshane J, Wells G, Nargan K, George JF, Ramdial PK, Agarwal A, Steyn AJC. Microanatomic Distribution of Myeloid Heme Oxygenase-1 Protects against Free Radical-Mediated Immunopathology in Human Tuberculosis. Cell Rep 2018; 25:1938-1952.e5. [PMID: 30428359 PMCID: PMC6250977 DOI: 10.1016/j.celrep.2018.10.073] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 07/18/2018] [Accepted: 10/19/2018] [Indexed: 11/26/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that controls inflammatory responses and redox homeostasis; however, its role during pulmonary tuberculosis (TB) remains unclear. Using freshly resected human TB lung tissue, we examined the role of HO-1 within the cellular and pathological spectrum of TB. Flow cytometry and histopathological analysis of human TB lung tissues showed that HO-1 is expressed primarily in myeloid cells and that HO-1 levels in these cells were directly proportional to cytoprotection. HO-1 mitigates TB pathophysiology by diminishing myeloid cell-mediated oxidative damage caused by reactive oxygen and/or nitrogen intermediates, which control granulocytic karyorrhexis to generate a zonal HO-1 response. Using whole-body or myeloid-specific HO-1-deficient mice, we demonstrate that HO-1 is required to control myeloid cell infiltration and inflammation to protect against TB progression. Overall, this study reveals that zonation of HO-1 in myeloid cells modulates free-radical-mediated stress, which regulates human TB immunopathology.
Collapse
Affiliation(s)
- Krishna C Chinta
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Vikram Saini
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Joel N Glasgow
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Vineel P Reddy
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremie M Lever
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | - Ryan M Wells
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amie Traylor
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Gene P Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Veena B Antony
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jessy Deshane
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gordon Wells
- Africa Health Research Institute, Durban 4001, South Africa
| | | | - James F George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pratistadevi K Ramdial
- Department of Anatomical Pathology, NHLS, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban 4091, South Africa
| | - Anupam Agarwal
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Veterans Affairs, Birmingham, AL 35294, USA
| | - Adrie J C Steyn
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Africa Health Research Institute, Durban 4001, South Africa; UAB Center for AIDS Research, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
152
|
Hu Y, Cai MC, Wang L, Zhang TH, Luo ZG, Zhang GW, Zuo FY. MiR-1246 is upregulated and regulates lung cell apoptosis during heat stress in feedlot cattle. Cell Stress Chaperones 2018; 23:1219-1228. [PMID: 30105590 PMCID: PMC6237691 DOI: 10.1007/s12192-018-0927-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 11/29/2022] Open
Abstract
Globally, heat stress seriously threatens productivity of cattle. The objective of this study was to identify novel miRNAs that regulated heat stress in feedlot cattle. Experiment was conducted under heat stress and normal conditions. With profiling miRNAs of each feedlot cattle, our results showed the level of miR-1246 was significantly increased in these heat-stressed cattle (P < 0.05). Furthermore, by using bioinformatics analysis and luciferase reporter assays combined with qPCR and western blot, we found miR-1246 negatively regulated poly (C) binding protein 2 (PCBP2) and cAMP response element binding protein-like 2 (CREBL2) mRNA and protein levels through binding to the 3'-UTR region (P < 0.05); further, it inhibited heat-induced apoptosis in lung cells. Finally, our results suggested that miR-1246 plays an important role in heat stress and it has the potential to be a novel modulation factor for heat stress in feedlot cattle.
Collapse
Affiliation(s)
- Yu Hu
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Ming-Chen Cai
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, 611130, Sichuan, People's Republic of China
| | - Ling Wang
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Ting-Huan Zhang
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing Academy of Animal Sciences, Chongqing, 402460, People's Republic of China
| | - Zhong-Gang Luo
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Gong-Wei Zhang
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Fu-Yuan Zuo
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China.
| |
Collapse
|
153
|
Jiang X, Tang Q, Zhang J, Wang H, Bai L, Meng P, Qin X, Xu G, Bose DD, Wang B, Chen C, Zou Z. Autophagy-dependent release of zinc ions is critical for acute lung injury triggered by zinc oxide nanoparticles. Nanotoxicology 2018; 12:1068-1091. [PMID: 30317896 DOI: 10.1080/17435390.2018.1513094] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Pulmonary exposure to zinc oxide nanoparticles (ZnONPs) could cause acute lung injury (ALI), but the underlying molecular mechanism remains unclear. Herein, we established a ZnONPs-induced ALI mouse model, characterized by the histopathological changes (edema and infiltration of inflammatory cells in lung tissues), and the elevation of total protein and cytokine interleukin-6 in bronchoalveolar lavage fluid in time- and dose-dependent manners. This model also exhibited features like the disturbance of redox-state (reduced of glutathione to glutathione disulfide ratio, elevation of heme oxygenase-1 and superoxide dismutase 2), the decrease of adenosine triphosphate synthesis and the release of zinc ions in the lung tissues. Interestingly, we found that ZnONPs exposure caused the accumulation of autophagic vacuoles and the elevation of microtubule-associated proteins 1A/1B light chain (LC)3B-II and p62, indicating the impairment of autophagic flux. Our data indicated that the above process might be regulated by the activation of AMP-activated protein kinase but not the mammalian target of rapamycin pathway. The association between ZnONPs-induced ALI and autophagy was further verified by a classical autophagy inhibitor, 3-methyladenine (3-MA). 3-MA administration reduced the accumulation of autophagic vacuoles, the expression of LC3B-II and p62, followed by a significant attenuation of histopathological changes, inflammation, and oxidative stress. More importantly, 3-MA could directly decrease the release of zinc ions in lung tissues. Taken together, our study provides the evidence that ZnONPs-induced pulmonary toxicity is autophagy-dependent, suggests that limiting the release of zinc ions by inhibiting autophagy could be a feasible strategy for the prevention of ZnONPs-associated pulmonary toxicity.
Collapse
Affiliation(s)
- Xuejun Jiang
- a Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center , Chongqing Medical University , Chongqing , People's Republic of China.,b Laboratory of Tissue and Cell Biology, Experimental Teaching and Management Center , Chongqing Medical University , Chongqing , People's Republic of China
| | - Qianghu Tang
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Jun Zhang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Hong Wang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Lulu Bai
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Pan Meng
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Xia Qin
- e Department of Pharmacy , The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Ge Xu
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Diptiman D Bose
- f Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences , Western New England University , Springfield , MA , USA
| | - Bin Wang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Chengzhi Chen
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China.,g Post-doctoral Research Stations of Nursing Science, School of Nursing , Chongqing Medical University , Chongqing , People's Republic of China
| | - Zhen Zou
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| |
Collapse
|
154
|
Heme oxygenase-2 suppresses acute inflammation and improves the survival of skin allografts. Int Immunopharmacol 2018; 63:191-197. [DOI: 10.1016/j.intimp.2018.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 11/24/2022]
|
155
|
Dunigan K, Li Q, Li R, Locy ML, Wall S, Tipple TE. The thioredoxin reductase inhibitor auranofin induces heme oxygenase-1 in lung epithelial cells via Nrf2-dependent mechanisms. Am J Physiol Lung Cell Mol Physiol 2018; 315:L545-L552. [PMID: 30024305 PMCID: PMC6230877 DOI: 10.1152/ajplung.00214.2018] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 11/22/2022] Open
Abstract
Thioredoxin reductase-1 (TXNRD1) inhibition effectively activates nuclear factor (erythroid-derived 2)-like 2 (Nrf2) responses and attenuates lung injury in acute respiratory distress syndrome (ARDS) and bronchopulmonary dysplasia (BPD) models. Upon TXNRD1 inhibition, heme oxygenase-1 (HO-1) is disproportionally increased compared with Nrf2 target NADPH quinone oxidoreductase-1 (Nqo1). HO-1 has been investigated as a potential therapeutic target in both ARDS and BPD. TXNRD1 is predominantly expressed in airway epithelial cells; however, the mechanism of HO-1 induction by TXNRD1 inhibitors is unknown. We tested the hypothesis that TXNRD1 inhibition induces HO-1 via Nrf2-dependent mechanisms. Wild-type (WT), Nrf2KO1.3, and Nrf2KO2.2 cells were morphologically indistinguishable, indicating that Nrf2 can be deleted from murine-transformed club cells (mtCCs) using CRISPR/Cas9 gene editing. Hemin, a Nrf2-independent HO-1-inducing agent, significantly increased HO-1 expression in WT, Nrf2KO1.3, and Nrf2KO2.2. Auranofin (AFN) (0.5 µM) inhibited TXNRD1 activity by 50% and increased Nqo1 and Hmox1 mRNA levels by 6- and 24-fold, respectively, in WT cells. Despite similar levels of TXNRD1 inhibition, Nqo1 mRNA levels were not different between control and AFN-treated Nrf2KO1.3 and Nrf2KO2.2. AFN slightly increased Hmox1 mRNA levels in Nrf2KO1.3 and Nrf2KO2.2 cells compared with controls. AFN failed to increase HO-1 protein in Nrf2KO1.3 and Nrf2KO2.2 compared with a 36-fold increase in WT mtCCs. Our data indicate that Nrf2 is the primary mechanism by which TXNRD1 inhibitors increase HO-1 in lung epithelia. Future studies will use ARDS and BPD models to define the role of HO-1 in attenuation of lung injury by TXNRD1 inhibitors.
Collapse
Affiliation(s)
- Katelyn Dunigan
- Neonatal Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, Alabama
- University of Alabama at Birmingham , Birmingham, Alabama
| | - Qian Li
- Neonatal Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, Alabama
- University of Alabama at Birmingham , Birmingham, Alabama
| | - Rui Li
- Neonatal Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, Alabama
- University of Alabama at Birmingham , Birmingham, Alabama
| | - Morgan L Locy
- University of Alabama at Birmingham , Birmingham, Alabama
| | - Stephanie Wall
- Neonatal Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, Alabama
- University of Alabama at Birmingham , Birmingham, Alabama
| | - Trent E Tipple
- Neonatal Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, Alabama
- University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
156
|
Modulation of the monocyte/macrophage system in heart failure by targeting heme oxygenase-1. Vascul Pharmacol 2018; 112:79-90. [PMID: 30213580 DOI: 10.1016/j.vph.2018.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/26/2018] [Accepted: 08/29/2018] [Indexed: 01/14/2023]
Abstract
Upon myocardial infarction (MI) immune system becomes activated by extensive necrosis of cardiomyocytes releasing intracellular molecules called damage-associated molecular patterns. Overactive and prolonged immune responses are likely to be responsible for heart failure development and progression in patients surviving the ischemic episode. Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. This stress-inducible enzyme is induced by various oxidative and inflammatory signals. Consequently, biological actions of HO-1 are not limited to degradation of a toxic heme released from hemoproteins, but also provide an adaptive cellular response against chronic inflammation and oxidative injury. Indeed, the immunomodulatory and anti-inflammatory properties of HO-1 were demonstrated in several experimental studies, as well as in human cases of genetic HO-1 deficiency. HO-1 was shown to suppress the production, myocardial infiltration and inflammatory properties of monocytes and macrophages what resulted in limitation of post-MI cardiac damage. This review specifically addresses the role of HO-1, heme and its degradation products in macrophage biology and post-ischemic cardiac repair. A more complete understanding of these mechanisms is essential to develop new therapeutic approaches.
Collapse
|
157
|
Agmatine attenuates rhabdomyolysis-induced acute kidney injury in rats in a dose dependent manner. Life Sci 2018; 208:79-86. [DOI: 10.1016/j.lfs.2018.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
|
158
|
Yen IC, Shi LS, Chung MC, Ahmetaj-Shala B, Chang TC, Lee SY. Antrolone, a Novel Benzoid Derived from Antrodia cinnamomea, Inhibits the LPS-Induced Inflammatory Response in RAW264.7 Macrophage Cells by Balancing the NF-
κ
B and Nrf2 Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:1297-1313. [PMID: 30149752 DOI: 10.1142/s0192415x18500684] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Antrodia cinnamomea, a medicinal mushroom, has previously demonstrated anti-inflammatory activity, although the specific compound responsible for the effect remains unclear. The present study was designed to investigate the anti-inflammatory property of antrolone, a novel benzoid derived from A. cinnamomea mycelium, and to clarify the underlying mechanisms of action. To this end, murine macrophage RAW264.7 cells were treated with antrolone (0.1-30 μ M) 30 min prior to stimulation with lipopolysaccharides (LPS, 0.1μ g/ml) for 24 h. Cell viability, nitric oxide (NO) and prostaglandin E2 (PGE2) production, levels of pro-inflammatory cytokines and chemokines, and the signaling pathways involved in the inflammatory cascades were then investigated. Our results show that antrolone significantly decreased LPS-induced NO, PGE2, pro-inflammatory cytokine, and keratinocyte chemoattractant CXCL1 (KC) production and reduced levels of the proteins inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2). These effects were independent of the effect of antrolone on macrophage cytotoxicity. Moreover, antrolone significantly inhibited the activation of the NFκ B, MAPK, and AKT pathways, while it increased nuclear factor erythroid-2-related factor (Nrf2) and heme oxygenase-1 (HO-1) levels. Our findings suggest that antrolone exhibits potent anti-inflammatory activity and may, therefore, be a lead compound for the development of an anti-inflammatory drug.
Collapse
Affiliation(s)
- I-Chuan Yen
- * School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Li-Shian Shi
- § Department of Biotechnology, National Formosa University, Yunlin, Taiwan
| | - Min-Chieh Chung
- † Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | | | - Tsu-Chung Chang
- † Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Yu Lee
- ‡ Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
159
|
Glass K, Thibault D, Guo F, Mitchel JA, Pham B, Qiu W, Li Y, Jiang Z, Castaldi PJ, Silverman EK, Raby B, Park JA, Yuan GC, Zhou X. Integrative epigenomic analysis in differentiated human primary bronchial epithelial cells exposed to cigarette smoke. Sci Rep 2018; 8:12750. [PMID: 30143676 PMCID: PMC6109173 DOI: 10.1038/s41598-018-30781-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 07/24/2018] [Indexed: 12/12/2022] Open
Abstract
Cigarette smoke (CS) is one of the major risk factors for many pulmonary diseases, including chronic obstructive pulmonary disease (COPD) and lung cancer. The first line of defense for CS exposure is the bronchial epithelial cells. Elucidation of the epigenetic changes during CS exposure is key to gaining a mechanistic understanding into how mature and differentiated bronchial epithelial cells respond to CS. Therefore, we performed epigenomic profiling in conjunction with transcriptional profiling in well-differentiated human bronchial epithelial (HBE) cells cultured in air-liquid interface (ALI) exposed to the vapor phase of CS. The genome-wide enrichment of histone 3 lysine 27 acetylation was detected by chromatin immunoprecipitation followed by next generation sequencing (ChIP-Seq) in HBE cells and suggested the plausible binding of specific transcription factors related to CS exposure. Additionally, interrogation of ChIP-Seq data with gene expression profiling of HBE cells after CS exposure for different durations (3 hours, 2 days, 4 days) suggested that earlier epigenetic changes (3 hours after CS exposure) may be associated with later gene expression changes induced by CS exposure (4 days). The integration of epigenetics and gene expression data revealed signaling pathways related to CS-induced epigenetic changes in HBE cells that may identify novel regulatory pathways related to CS-induced COPD.
Collapse
Affiliation(s)
- Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Derek Thibault
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Feng Guo
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Jennifer A Mitchel
- Department of Enviromental Health, Harvard T.H. School of Public Health, Boston, United States
| | - Betty Pham
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Weiliang Qiu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Yan Li
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Zhiqiang Jiang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States.,Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Benjamin Raby
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Jin-Ah Park
- Department of Enviromental Health, Harvard T.H. School of Public Health, Boston, United States
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biostatistics, Harvard T.H. School of Public Health, Boston, United States
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States. .,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States.
| |
Collapse
|
160
|
Malaria Parasite-Mediated Alteration of Macrophage Function and Increased Iron Availability Predispose to Disseminated Nontyphoidal Salmonella Infection. Infect Immun 2018; 86:IAI.00301-18. [PMID: 29986892 DOI: 10.1128/iai.00301-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/28/2018] [Indexed: 12/31/2022] Open
Abstract
Disseminated infections with nontyphoidal Salmonella (NTS) are a significant cause of child mortality in sub-Saharan Africa. NTS infection in children is clinically associated with malaria, suggesting that malaria compromises the control of disseminated NTS infection. To study the mechanistic basis for increased NTS susceptibility, we utilized a model of concurrent infection with Salmonella enterica serotype Typhimurium and Plasmodium yoelii nigeriensis (P. yoelii). Underlying malaria blunted monocyte expression of Ly6C, a marker for inflammatory activation, and impaired recruitment of inflammatory cells to the liver. Hepatic mononuclear phagocytes expressed lower levels of inducible nitric oxide synthase, tumor necrosis factor alpha, and granulocyte-macrophage colony-stimulating factor and showed increased levels of production of interleukin-10 and heme oxygenase-1, indicating that the underlying malaria modifies the activation state and inflammatory response of mononuclear phagocytes to NTS. P. yoelii infection also increased intracellular iron levels in liver mononuclear cells, as evidenced by elevated levels of ferritin and by the rescue of an S Typhimurium tonB feoB mutant defective for iron uptake. In addition, concurrent P. yoelii infection partially rescued the systemic colonization defect of an S Typhimurium spiB mutant defective for type III secretion system 2 (T3SS-2), indicating that the ability of phagocytic cells to limit the spread of S Typhimurium is impaired during concurrent P. yoelii infection. These results show that concurrent malaria increases susceptibility to disseminated NTS infection by blunting macrophage bactericidal mechanisms and providing an essential nutrient that enhances bacterial growth.
Collapse
|
161
|
Nakashima K, Sato T, Shigemori S, Shimosato T, Shinkai M, Kaneko T. Regulatory role of heme oxygenase-1 in silica-induced lung injury. Respir Res 2018; 19:144. [PMID: 30068325 PMCID: PMC6090697 DOI: 10.1186/s12931-018-0852-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 07/26/2018] [Indexed: 11/10/2022] Open
Abstract
Background Silicosis, a progressive inflammatory lung disease attributed mainly to occupational exposure to silica dust, shows loss of lung function even after cessation of exposure. In addition to conventional evaluation methods such as chest X-ray, computed tomography, and spirometry, we identified heme oxygenase (HO)-1, an inducible antioxidant, as a potential biomarker to identify at-risk patients. We found that HO-1 was critical in attenuating the disease progression of silicosis; however, the key signaling pathway has not yet been elucidated. Here, we report the critical pathway after silica exposure, focusing on the role of silica-derived reactive oxygen species (ROS) signaling and its attenuation, which is mediated by HO-1 induction, in vivo and in vitro. Methods Normal bronchial epithelial cells and a macrophage cell line, as well as a murine silicosis model generated by intratracheal administration of 2.5 mg of crystalline silica, were used in this study. The pathways activated in response to silica exposure, including the mitogen-activated protein kinase (MAPK) signaling pathway, were examined and compared with or without super-induction of HO-1. Results The murine silicosis model was first assessed for the evaluation of activated pathways after silica exposure, focusing on ROS-MAPK activation. In the murine model, increased expression of HO-1 in the lungs was observed after silica-instillation. Moreover, silica-medicated activation of extracellular signal-regulated kinase (ERK) in the lungs was attenuated in response to silica-induced HO-1 upregulation. Activation of other MAPKs, such as p38 and c-Jun N-terminal kinase pathways, after silica exposure was not significantly different irrespective of HO-1 induction. Further in vitro studies showed that 1) silica-induced HO-1 was significantly attenuated by inhibiting ERK activation, and 2) carbon monoxide and bilirubin as final byproducts of HO-1 could inhibit ERK activation. Taken together, silica-induced HO-1 upregulation was mediated by ERK activation, and HO-1 further regulates ERK activation via its final byproducts, carbon monoxide and bilirubin. Conclusions This is the first study to demonstrate the regulatory role of HO-1 in silicosis. This finding could contribute to the development of a treatment strategy of monitoring HO-1 levels as a marker of therapeutic intervention. Electronic supplementary material The online version of this article (10.1186/s12931-018-0852-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kentaro Nakashima
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Takashi Sato
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan.
| | - Suguru Shigemori
- Matebologenomics Core, Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | - Takeshi Shimosato
- Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Masaharu Shinkai
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 2360004, Japan
| |
Collapse
|
162
|
Castañeda AR, Pinkerton KE, Bein KJ, Magaña-Méndez A, Yang HT, Ashwood P, Vogel CFA. Ambient particulate matter activates the aryl hydrocarbon receptor in dendritic cells and enhances Th17 polarization. Toxicol Lett 2018; 292:85-96. [PMID: 29689377 PMCID: PMC5971007 DOI: 10.1016/j.toxlet.2018.04.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 03/01/2018] [Accepted: 04/18/2018] [Indexed: 02/02/2023]
Abstract
The objective of this study was to explore the role of the aryl hydrocarbon receptor (AhR) in ambient particulate matter (PM)-mediated activation of dendritic cells (DCs) and Th17-immune responses in vitro. To assess the potential role of the AhR in PM-mediated activation of DCs, co-stimulation, and cytokine expression, bone marrow (BM)-derived macrophages and DCs from C57BL/6 wildtype or AhR knockout (AhR-/-) mice were treated with PM. Th17 differentiation was assessed via co-cultures of wildtype or AhR-/- BMDCs with autologous naive T cells. PM2.5 significantly induced AhR DNA binding activity to dioxin responsive elements (DRE) and expression of the AhR repressor (AhRR), cytochrome P450 (CYP) 1A1, and CYP1B1, indicating activation of the AhR. In activated (OVA sensitized) BMDCs, PM2.5 induced interleukin (IL)-1β, CD80, CD86, and MHC class II, suggesting enhanced DC activation, co-stimulation, and antigen presentation; responses that were abolished in AhR deficient DCs. DC-T cell co-cultures treated with PM and lipopolysaccharide (LPS) led to elevated IL-17A and IL-22 expression at the mRNA level, which is mediated by the AhR. PM-treated DCs were essential in endowing T cells with a Th17-phenotype, which was associated with enhanced expression of MHC class II and cyclooxygenase (COX)-2. In conclusion, PM enhances DC activation that primes naive T cell differentiation towards a Th17-like phenotype in an AhR-dependent manner.
Collapse
Affiliation(s)
| | - Kent E Pinkerton
- Center for Health and the Environment, University of California, Davis, 95616, USA; Department of Pediatrics, School of Medicine, University of California, Davis, 95817, USA
| | - Keith J Bein
- Center for Health and the Environment, University of California, Davis, 95616, USA; Air Quality Research Center, University of California, Davis, CA, 95616, USA
| | - Alfonso Magaña-Méndez
- Escuela de Ciencias de la Salud, Universidad Autónoma de Baja California, Ensenada, C.P. 22860, Mexico
| | - Houa T Yang
- M.I.N.D. Institute, University of California, Davis, 95817, USA
| | - Paul Ashwood
- M.I.N.D. Institute, University of California, Davis, 95817, USA
| | - Christoph F A Vogel
- Center for Health and the Environment, University of California, Davis, 95616, USA; Department of Environmental Toxicology, University of California, Davis, 95616, USA.
| |
Collapse
|
163
|
Shih CM, Wu CH, Wu WJ, Hsiao YM, Ko JL. Hypericin inhibits hepatitis C virus replication via deacetylation and down-regulation of heme oxygenase-1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 46:193-198. [PMID: 30097118 DOI: 10.1016/j.phymed.2017.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 07/10/2017] [Accepted: 08/14/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Hepatitis C virus (HCV) is a globally prevalent pathogen and a leading cause of death and morbidity. Traditional therapy with pegylated interferon-<alpha> and ribavirin has had only limited success, with some adverse effects. Direct-acting antivirals (DAAs) are effective in suppressing HCV replication, but are expensive. PURPOSE Hypericin has been reported to be a good antiviral agent for inhibiting HCV replication, however, little is known about its mechanisms of action. The aim of this study is to elucidate the mode of action of hypericin in Ava5 human hepatoma cell line (Huh7 derivative) harboring HCV subgenomic replicon RNA. METHODS To determine the non-structure protein 5A (NS5A) mRNA and NS3 protein expression levels, real-time PCR and Western blot analysis were performed, respectively. To investigate how hypericin inhibits HCV replication, 5-aza-2'-deoxycytidine (5-Aza-dC) and chidamide were used for determining histone modification. Furthermore, shRNA was applied to confirm the role of heme oxygenase (HO-1) in HCV repression. RESULTS Hypericin in experiment were tested and showed no cytotoxicity. Hypericin reduced HO-1 and NS5A in a time- and dose- dependent manner. Chidamide, but not 5-Aza-dc, restored hypericin-induced reduction in HCV NS3 expression and reversed HO-1 expression in Ava5 cells. LY294002 inhibited HCV replication via HO-1 down-regulation. Constitutive expressed p-AKT was not involved in hypericin-induced reduction in HCV replication. In addition, shHO-1 inhibited HCV replication. CONCLUSION In conclusion, hypericin inhibits HCV replication via down-regulation of HO-1 expression and deacetylation.
Collapse
Affiliation(s)
- Chao-Ming Shih
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Yang Ming Hospital, Chia-Yi, Taiwan
| | - Chih-Hsien Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; Basic Medical Education Center, Central Taiwan University of Science and Technology, Taichung 406, Taiwan
| | - Wen-Jun Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Min Hsiao
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan.
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
164
|
Cosola C, Sabatino A, di Bari I, Fiaccadori E, Gesualdo L. Nutrients, Nutraceuticals, and Xenobiotics Affecting Renal Health. Nutrients 2018; 10:nu10070808. [PMID: 29937486 PMCID: PMC6073437 DOI: 10.3390/nu10070808] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) affects 8–16% of the population worldwide. In developed countries, the most important risk factors for CKD are diabetes, hypertension, and obesity, calling into question the importance of educating and acting on lifestyles and nutrition. A balanced diet and supplementation can indeed support the maintenance of a general health status, including preservation of renal function, and can help to manage and curb the main risk factors for renal damage. While the concept of protein and salt restriction in nephrology is historically acknowledged, the role of some nutrients in renal health and the importance of nutrition as a preventative measure for renal care are less known. In this narrative review, we provide an overview of the demonstrated and potential actions of some selected nutrients, nutraceuticals, and xenobiotics on renal health and function. The direct and indirect effects of fiber, protein, fatty acids, curcumin, steviol glycosides, green tea, coffee, nitrates, nitrites, and alcohol on kidney health are reviewed here. In view of functional and personalized nutrition, understanding the renal and systemic effects of dietary components is essential since many chronic conditions, including CKD, are related to systemic dysfunctions such as chronic low-grade inflammation.
Collapse
Affiliation(s)
- Carmela Cosola
- Department of Emergency and Organ Transplantation-Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, 70124 Bari, Italy.
| | - Alice Sabatino
- Department of Medicine and Surgery, Parma University Medical School, 43126 Parma, Italy.
| | - Ighli di Bari
- Department of Emergency and Organ Transplantation-Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, 70124 Bari, Italy.
| | - Enrico Fiaccadori
- Department of Medicine and Surgery, Parma University Medical School, 43126 Parma, Italy.
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation-Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, 70124 Bari, Italy.
| |
Collapse
|
165
|
Khan IU, Yoon Y, Kim A, Jo KR, Choi KU, Jung T, Kim N, Son Y, Kim WH, Kweon OK. Improved Healing after the Co-Transplantation of HO-1 and BDNF Overexpressed Mesenchymal Stem Cells in the Subacute Spinal Cord Injury of Dogs. Cell Transplant 2018; 27:1140-1153. [PMID: 29909686 PMCID: PMC6158544 DOI: 10.1177/0963689718779766] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abundant expression of proinflammatory cytokines after a spinal cord injury (SCI) creates an inhibitory microenvironment for neuroregeneration. The mesenchymal stem cells help to mitigate the inflammation and improve neural growth and survival. For this purpose, we potentiated the function of adipose-derived mesenchymal stem cells (Ad-MSCs) by transfecting them with brain-derived neurotrophic factor (BDNF) and heme oxygenase-1 (HO-1), through a lentivirus, to produce BDNF overexpressed Ad-MSCs (BDNF-MSCs), and HO-1 overexpressed Ad-MSCs (HO-1-MSCs). Sixteen SCI beagle dogs were randomly assigned into four treatment groups. We injected both HO-1 and BDNF-overexpressed MSCs as a combination group, to selectively control inflammation and induce neuroregeneration in SCI dogs, and compared this with BDNF-MSCs, HO-1-MSCs, and GFP-MSCs injected dogs. The groups were compared in terms of improvement in canine Basso, Beattie, and Bresnahan (cBBB) score during 8 weeks of experimentation. After 8 weeks, spinal cords were harvested and subjected to western blot analysis, immunofluorescent staining, and hematoxylin and eosin (H&E) staining. The combination group showed a significant improvement in hindlimb functions, with a higher BBB score, and a robust increase in neuroregeneration, depicted by a higher expression of Tuj-1, NF-M, and GAP-43 due to a decreased expression of the inflammatory markers interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), and an increased expression of interleukin-10 (IL-10) (P ≤ 0.05). H&E staining showed more reduced intraparenchymal fibrosis in the combination group than in other groups (P ≤ 0.05). It was thus suggested that the cotransplantation of HO-1 and BDNF-MSCs is more effective in promoting the healing of SCI. HO-1-MSCs reduce inflammation, which favors BDNF-induced neuroregeneration in SCI of dogs.
Collapse
Affiliation(s)
- Imdad Ullah Khan
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Yongseok Yoon
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Ahyoung Kim
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Kwang Rae Jo
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Kyeung Uk Choi
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Taeseong Jung
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Namyul Kim
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - YeonSung Son
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Wan Hee Kim
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Oh-Kyeong Kweon
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| |
Collapse
|
166
|
Hara Y, Shinkai M, Taguri M, Nagai K, Hashimoto S, Kaneko T. ELISA Development for Serum Hemeoxygenase-1 and Its Application to Patients with Acute Respiratory Distress Syndrome. Can Respir J 2018; 2018:9627420. [PMID: 29849835 PMCID: PMC5932439 DOI: 10.1155/2018/9627420] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/29/2018] [Accepted: 03/13/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Hemeoxygenase-1 (HO-1) is an essential enzyme in heme catabolism and has been proposed as a biomarker of lung disease prognosis. We modified a commercial HO-1 enzyme-linked immunosorbent assay (ELISA) kit to achieve higher sensitivity and evaluated if serum HO-1 could be a biomarker to predict the prognosis of acute respiratory distress syndrome (ARDS) patients. METHODS Serum samples were collected from 15 healthy volunteers to validate the modified ELISA. In the 22 patients with ARDS who were enrolled, serum HO-1 was measured upon diagnosis (D0) and at 7 days after diagnosis (D7). RESULTS The serum HO-1 concentration could be measured in all healthy volunteers. The intra- and interassay tests and the percentage recovery test were acceptable. Compared with normal control subjects, patients with ARDS had significantly higher D0 HO-1 concentrations (75.4 ng/mL versus 31.7 ng/mL, P < 0.001). The 28-day survival was significantly better in patients with low D0 HO-1 (<75.8 ng/mL) than in those with high D0 HO-1 (≥75.8 ng/mL) (mortality rate: 18% versus 73%, P=0.016). Nonsurvivors had significantly higher D0 and D7 HO-1 concentrations than survivors (P < 0.05). CONCLUSION Serum HO-1 may be a useful biomarker to predict the prognosis of patients with ARDS.
Collapse
Affiliation(s)
- Yu Hara
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaharu Shinkai
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masataka Taguri
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kenjiro Nagai
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoru Hashimoto
- Division of Intensive Care Unit, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
167
|
Qu X, Hu M, Shang Y, Pan L, Jia P, Fu C, Liu Q, Wang Y. Liver Transcriptome and miRNA Analysis of Silver Carp ( Hypophthalmichthys molitrix) Intraperitoneally Injected With Microcystin-LR. Front Physiol 2018; 9:381. [PMID: 29692738 PMCID: PMC5902739 DOI: 10.3389/fphys.2018.00381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/28/2018] [Indexed: 01/04/2023] Open
Abstract
Next-generation sequencing was used to analyze the effects of toxic microcystin-LR (MC-LR) on silver carp (Hypophthalmichthys molitrix). Silver carps were intraperitoneally injected with MC-LR, and RNA-seq and miRNA-seq in the liver were analyzed at 0.25, 0.5, and 1 h. The expression of glutathione S-transferase (GST), which acts as a marker gene for MC-LR, was tested to determine the earliest time point at which GST transcription was initiated in the liver tissues of the MC-LR-treated silver carps. Hepatic RNA-seq/miRNA-seq analysis and data integration analysis were conducted with reference to the identified time point. Quantitative PCR (qPCR) was performed to detect the expression of the following genes at the three time points: heme oxygenase 1 (HO-1), interleukin-10 receptor 1 (IL-10R1), apolipoprotein A-I (apoA-I), and heme binding protein 2 (HBP2). Results showed that the liver GST expression was remarkably decreased at 0.25 h (P < 0.05). RNA-seq at this time point revealed that the liver tissue contained 97,505 unigenes, including 184 significantly different unigenes and 75 unknown genes. Gene Ontology (GO) term enrichment analysis suggested that 35 of the 145 enriched GO terms were significantly enriched and mainly related to the immune system regulation network. KEGG pathway enrichment analysis showed that 18 of the 189 pathways were significantly enriched, and the most significant was a ribosome pathway containing 77 differentially expressed genes. miRNA-seq analysis indicated that the longest miRNA had 22 nucleotides (nt), followed by 21 and 23 nt. A total of 286 known miRNAs, 332 known miRNA precursor sequences, and 438 new miRNAs were predicted. A total of 1,048,575 mRNA–miRNA interaction sites were obtained, and 21,252 and 21,241 target genes were respectively predicted in known and new miRNAs. qPCR revealed that HO-1, IL-10R1, apoA-I, and HBP2 were significantly differentially expressed and might play important roles in the toxicity and liver detoxification of MC-LR in fish. These results were consistent with those of high-throughput sequencing, thereby verifying the accuracy of our sequencing data. RNA-seq and miRNA-seq analyses of silver carp liver injected with MC-LR provided valuable and new insights into the toxic effects of MC-LR and the antitoxic mechanisms of MC-LR in fish. The RNA/miRNA data are available from the NCBI database Registration No. : SRP075165.
Collapse
Affiliation(s)
- Xiancheng Qu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.,The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, China
| | - Menghong Hu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.,The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, China
| | - Yueyong Shang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.,The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, China
| | - Lisha Pan
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Peixuan Jia
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Chunxue Fu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Qigen Liu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.,The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, China
| | - Youji Wang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.,The Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai, China.,International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, China
| |
Collapse
|
168
|
Pabelick CM, Thompson MA, Britt RD. Effects of Hyperoxia on the Developing Airway and Pulmonary Vasculature. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:179-194. [PMID: 29047087 DOI: 10.1007/978-3-319-63245-2_11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although it is necessary and part of standard practice, supplemental oxygen (40-90% O2) or hyperoxia is a significant contributing factor to development of bronchopulmonary dysplasia, persistent pulmonary hypertension, recurrent wheezing, and asthma in preterm infants. This chapter discusses hyperoxia and the role of redox signaling in the context of neonatal lung growth and disease. Here, we discuss how hyperoxia promotes dysfunction in the airway and the known redox-mediated mechanisms that are important for postnatal vascular and alveolar development. Whether in the airway or alveoli, redox pathways are important and greatly influence the neonatal lung.
Collapse
Affiliation(s)
- Christina M Pabelick
- Department of Anesthesiology, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA. .,Departments Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA.
| | - Michael A Thompson
- Department of Anesthesiology, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA
| | - Rodney D Britt
- Departments Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA
| |
Collapse
|
169
|
Zhou Y, Liu Y, Sun H, Ma J, Xiao L, Cao L, Li W, Wang B, Yuan J, Chen W. Associations of urinary polycyclic aromatic hydrocarbon metabolites with fractional exhaled nitric oxide and exhaled carbon monoxide: A cross-sectional study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 618:542-550. [PMID: 29149738 DOI: 10.1016/j.scitotenv.2017.10.294] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/26/2017] [Accepted: 10/28/2017] [Indexed: 06/07/2023]
Abstract
Exposure to Polycyclic aromatic hydrocarbons (PAHs) has been associated with inflammatory responses. Fractional exhaled nitric oxide (FeNO) and exhaled carbon monoxide (eCO) are both important inflammatory mediators especially in airways. However, few studies have investigated associations of PAH exposures with FeNO or eCO. Therefore, we aimed to quantify the associations of urinary PAH metabolites with FeNO and eCO levels, and investigate their potential effect modifiers by linear mixed models among 4133 participants from the Wuhan-Zhuhai cohort in China. We further performed stratified analyses to estimate effect modification. We found significant associations of increased urinary PAH metabolites with elevated eCO and FeNO. Among all participants, each 1% increase of 1-hydroxynaphthalene, 2-hydroxynaphthalene, 2-hydroxyfluorene, 4-hydroxyphenanthrene, 3-hydroxyphenanthrene, and total PAH metabolites was significantly associated with a 12.6% (95% confidence interval: 9.3%, 15.9%), 9.7% (6.5%, 12.9%), 7.5% (4.1%, 10.9%), 3.2% (0.2%, 6.2%), 2.7% (0.1%, 5.3%), and 6.5% (2.7%, 10.4%) increased eCO level, respectively; while each 1% increase of urinary 1-hydroxynaphthalene, 9-hydroxyphenanthrene, 3-hydroxyphenanthrene, and 2-hydroxyphenanthrene was associated with a -3.0% (-5.8%, -0.2%), 2.9% (0.3%, 5.6%), 3.2% (1.0%, 5.4%), and 4.5% (2.2%, 6.9%) change of FeNO level, respectively. Positive associations between certain urinary PAH metabolites and eCO were observed among both ever-smokers and non-smokers, and the associations were stronger among ever-smokers than that among non-smokers. Increased urinary PAH metabolites were associated with decreased FeNO among ever-smokers and elevated FeNO levels among non-smokers. Our findings suggest that PAH exposures may impair airway through inducing inflammatory response, especially among ever-smokers.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yuewei Liu
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei 430079, China
| | - Huizhen Sun
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lili Xiao
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Limin Cao
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei Li
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bin Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jing Yuan
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
170
|
Mei M, Song H, Chen L, Hu B, Bai R, Xu D, Liu Y, Zhao Y, Chen C. Early-life exposure to three size-fractionated ultrafine and fine atmospheric particulates in Beijing exacerbates asthma development in mature mice. Part Fibre Toxicol 2018. [PMID: 29540228 PMCID: PMC5851307 DOI: 10.1186/s12989-018-0249-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Epidemiological studies have suggested that elevated levels of air pollution contribute to an increased incidence or severity of asthma. Although late-onset adult asthma seems to be more attributable to environmental risk factors, limited data is available on the impact of early-life exposure to size-fractionated ambient particulate matter (PM) on asthma in adults. We aimed to determine the effect on the development and exacerbation of asthma in the adult after the mice were exposed as juveniles to three size-fractionated ambient particulates collected from Beijing. METHODS The three size-fractionated ambient particulates were collected from urban Beijing in winter, heavily affected by traffic and coal-fired emissions. The typical morphological and major chemical components of the PM were characterized first. Oxidative stress and expression of DNA methyltransferases (DNMTs) were then examined in vitro and in the lungs of mouse pups 48 h after exposure to PM by oropharyngeal aspiration. When the exposed and control juvenile mice matured to adulthood, an antigen-induced asthma model was established and relevant bio-indices were assessed. RESULTS PM with different granularities can induce oxidative stress; in particular, F1, with the smallest size (< 0.49 μm), decreased the mRNA expression of DNMTs in vitro and in vivo the most significantly. In an asthma model of adult mice, previous exposure as juveniles to size-fractionated PM caused increased peribronchiolar inflammation, increased airway mucus secretion, and increased production of Th2 cytokines and chemokines. In general, F1 and F2 (aerodynamic diameter < 0.95 μm) particulates affected murine adult asthma development more seriously than F3 (0.95-1.5 μm). Moreover, F1 led to airway inflammation in the form of both increased neutrophils and eosinophils in BALF. The activation of the TGF-β1/Smad2 and Smad3/Stat3 signaling pathways leading to airway fibrosis was more profoundly induced by F1. CONCLUSION This study demonstrated that exposure to ambient PM in juvenile mice enhanced adult asthma development, as shown by increased Th2 responses, which might be associated with the persistent effects resulting from the oxidative stress and decreased gene expression of DNMTs induced by PM exposure. The observed differences between the effects of three size-fractionated particulates were attributed to particle sizes and chemical constituents, including heavy metals and also PAHs, since the amounts of PAH associated with more severe toxicity were enriched equivalently in the F1 and F2 fractions. Relative to the often mentioned PM2.5, PM with an aerodynamic diameter smaller than 0.95 μm had a more aggravating effect on asthma development.
Collapse
Affiliation(s)
- Mei Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience & Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology of China and University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Haojun Song
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Lina Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience & Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology of China and University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Bin Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience & Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology of China and University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience & Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology of China and University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Diandou Xu
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience & Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology of China and University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience & Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology of China and University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience & Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology of China and University of Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
171
|
Long Noncoding RNAs and mRNA Regulation in Peripheral Blood Mononuclear Cells of Patients with Chronic Obstructive Pulmonary Disease. Mediators Inflamm 2018; 2018:7501851. [PMID: 29725270 PMCID: PMC5872599 DOI: 10.1155/2018/7501851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022] Open
Abstract
Background Inflammation plays a pivotal role in the pathogenesis of chronic obstructive pulmonary disease (COPD). We evaluated the lncRNA and mRNA expression profile of peripheral blood mononuclear cells (PBMCs) from healthy nonsmokers, smokers without airflow limitation, and COPD patients. Methods lncRNA and mRNA profiling of PBMCs from 17 smokers and 14 COPD subjects was detected by high-throughput microarray. The expression of dysregulated lncRNAs was validated by qPCR. The lncRNA targets in dysregulated mRNAs were predicted and the GO enrichment was analyzed. The regulatory role of lncRNA ENST00000502883.1 on CXCL16 expression and consequently the effect on PBMC recruitment were investigated by siRNA knockdown and chemotaxis analysis. Results We identified 158 differentially expressed lncRNAs in PBMCs from COPD subjects compared with smokers. The dysregulated expression of 5 selected lncRNAs NR_026891.1 (FLJ10038), ENST00000502883.1 (RP11-499E18.1), HIT000648516, XR_429541.1, and ENST00000597550.1 (CTD-2245F17.3), was validated. The GO enrichment showed that leukocyte migration, immune response, and apoptosis are the main enriched processes that previously reported to be involved in the pathogenesis of COPD. The regulatory role of ENST00000502883.1 on CXCL16 expression and consequently the effect on PBMC recruitment was confirmed. Conclusion This study may provide clues for further studies targeting lncRNAs to control inflammation in COPD.
Collapse
|
172
|
Stiefelmaier J, Ledermann B, Sorg M, Banek A, Geib D, Ulber R, Frankenberg-Dinkel N. Pink bacteria-Production of the pink chromophore phycoerythrobilin with Escherichia coli. J Biotechnol 2018; 274:47-53. [PMID: 29549003 DOI: 10.1016/j.jbiotec.2018.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/01/2018] [Accepted: 03/11/2018] [Indexed: 11/29/2022]
Abstract
Phycoerythrobilin (PEB) is an open-chain tetrapyrrole derived from heme and plays an important role as light-harvesting pigment in the phycobiliproteins of cyanobacteria and red algae. Furthermore, PEB can also function as an antioxidant with potential use as a natural acid stable food colorant. PEB is not commercially available and large, pure quantities can only be obtained by laborious methanolysis of red algae followed by liquid chromatography. Here we describe an improved method for high yield production and purification of PEB in Escherichia coli via heterologous expression where the two required enzymes heme oxygenase and PEB synthase subsequently convert the substrate heme provided by the host cell. Experiments in shaking flasks resulted in the highest product yield of 680.23 ± 42.75 μg PEB per g cell dry weight, by induction with 0.1 mM IPTG. Scale-up to batch-operated fermentation in a 2 L bioreactor reached product concentrations up to 5.02 mg PEB L-1 by adjustment of aeration, induction time, media composition and supplementation of precursors. A further approach included separation of PEB from developed foam above the culture. This enabled continuous product collection during cultivation and simplified product purification. Produced PEB was validated via UV-vis spectroscopy, high pressure liquid chromatography and mass spectrometry.
Collapse
Affiliation(s)
- Judith Stiefelmaier
- Lehrgebiet Bioverfahrenstechnik, Technische Universität Kaiserslautern, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany.
| | - Benjamin Ledermann
- Abteilung für Mikrobiologie, Technische Universität Kaiserslautern, Erwin-Schroedinger-Straße 56, 67663 Kaiserslautern, Germany.
| | - Michael Sorg
- Lehrgebiet Bioverfahrenstechnik, Technische Universität Kaiserslautern, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany.
| | - Angela Banek
- Lehrgebiet Bioverfahrenstechnik, Technische Universität Kaiserslautern, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany; Abteilung für Mikrobiologie, Technische Universität Kaiserslautern, Erwin-Schroedinger-Straße 56, 67663 Kaiserslautern, Germany.
| | - Doris Geib
- Lehrgebiet Bioverfahrenstechnik, Technische Universität Kaiserslautern, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany.
| | - Roland Ulber
- Lehrgebiet Bioverfahrenstechnik, Technische Universität Kaiserslautern, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany.
| | - Nicole Frankenberg-Dinkel
- Abteilung für Mikrobiologie, Technische Universität Kaiserslautern, Erwin-Schroedinger-Straße 56, 67663 Kaiserslautern, Germany.
| |
Collapse
|
173
|
Cytoprotective effect and purification of novel antioxidant peptides from hazelnut (C. heterophylla Fisch) protein hydrolysates. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
174
|
Adebambo OA, Shea D, Fry RC. Cadmium disrupts signaling of the hypoxia-inducible (HIF) and transforming growth factor (TGF-β) pathways in placental JEG-3 trophoblast cells via reactive oxygen species. Toxicol Appl Pharmacol 2018; 342:108-115. [DOI: 10.1016/j.taap.2018.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/09/2018] [Accepted: 01/17/2018] [Indexed: 12/16/2022]
|
175
|
Tomas-Hernández S, Blanco J, Rojas C, Roca-Martínez J, Ojeda-Montes MJ, Beltrán-Debón R, Garcia-Vallvé S, Pujadas G, Arola L, Mulero M. Resveratrol Potently Counteracts Quercetin Starvation-Induced Autophagy and Sensitizes HepG2 Cancer Cells to Apoptosis. Mol Nutr Food Res 2018; 62. [PMID: 29336118 DOI: 10.1002/mnfr.201700610] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 12/21/2017] [Indexed: 12/19/2022]
Abstract
SCOPE Resveratrol (RSV) has been described as a potent antioxidant, antisteatotic, and antitumor compound, and it has also been identified as a potent autophagy inducer. On the other hand, quercetin (QCT) is a dietary flavonoid with known antitumor, anti-inflammatory, and antidiabetic effects. Additionally, QCT increases autophagy. To study the hypothetical synergistic effect of both compounds, we test the combined effect of QCT and RSV on the autophagy process in HepG2 cells. METHODS AND RESULTS Autophagy is studied by western blotting, real-time RT-PCR, and cellular staining. Our results clearly indicate a bifunctional molecular effect of RSV. Both polyphenols are individually able to promote autophagy. Strikingly, when RSV is combined with QCT, it promotes a potent reduction of QCT-induced autophagy and influences proapoptotic signaling. CONCLUSION RSV acts differentially on the autophagic process depending on the cellular energetic state. We further characterize the molecular mechanisms related to this effect, and we observe that AMP-activated protein kinase (AMPK) phosphorylation, heme oxygenase 1 (HO-1) downregulation, lysosomal membrane permeabilization (LMP), and Zinc (Zn2+ ) dynamics could be important modulators of such RSV-related effects and could globally represent a promising strategy to sensitize cancer cells to QCT treatment.
Collapse
Affiliation(s)
- Sarah Tomas-Hernández
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain
| | - Jordi Blanco
- Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - Cristina Rojas
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain
| | - Joel Roca-Martínez
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain
| | - María José Ojeda-Montes
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain
| | - Raúl Beltrán-Debón
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain
| | - Santiago Garcia-Vallvé
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain.,Technological Unit of Nutrition and Health, EURECAT-Technological Center of Catalonia, Reus, Spain
| | - Gerard Pujadas
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain.,Technological Unit of Nutrition and Health, EURECAT-Technological Center of Catalonia, Reus, Spain
| | - Lluís Arola
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain.,Technological Unit of Nutrition and Health, EURECAT-Technological Center of Catalonia, Reus, Spain
| | - Miquel Mulero
- Cheminformatic and Nutrition Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, Tarragona, Spain
| |
Collapse
|
176
|
Chi L, Gao B, Tu P, Liu CW, Xue J, Lai Y, Ru H, Lu K. Individual susceptibility to arsenic-induced diseases: the role of host genetics, nutritional status, and the gut microbiome. Mamm Genome 2018; 29:63-79. [PMID: 29429126 DOI: 10.1007/s00335-018-9736-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/17/2018] [Indexed: 01/16/2023]
Abstract
Arsenic (As) contamination in water or food is a global issue affecting hundreds of millions of people. Although As is classified as a group 1 carcinogen and is associated with multiple diseases, the individual susceptibility to As-related diseases is highly variable, such that a proportion of people exposed to As have higher risks of developing related disorders. Many factors have been found to be associated with As susceptibility. One of the main sources of the variability found in As susceptibility is the variation in the host genome, namely, polymorphisms of many genes involved in As transportation, biotransformation, oxidative stress response, and DNA repair affect the susceptibility of an individual to As toxicity and then influence the disease outcomes. In addition, lifestyles and many nutritional factors, such as folate, vitamin C, and fruit, have been found to be associated with individual susceptibility to As-related diseases. Recently, the interactions between As exposure and the gut microbiome have been of particular concern. As exposure has been shown to perturb gut microbiome composition, and the gut microbiota has been shown to also influence As metabolism, which raises the question of whether the highly diverse gut microbiota contributes to As susceptibility. Here, we review the literature and summarize the factors, such as host genetics and nutritional status, that influence As susceptibility, and we also present potential mechanisms of how the gut microbiome may influence As metabolism and its toxic effects on the host to induce variations in As susceptibility. Challenges and future directions are also discussed to emphasize the importance of characterizing the specific role of these factors in interindividual susceptibility to As-related diseases.
Collapse
Affiliation(s)
- Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Bei Gao
- NIH West Coast Metabolomics Center, University of California, Davis, CA, 95616, USA
| | - Pengcheng Tu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chih-Wei Liu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jingchuan Xue
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yunjia Lai
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hongyu Ru
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, 27607, USA
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
177
|
Salerno L, Amata E, Romeo G, Marrazzo A, Prezzavento O, Floresta G, Sorrenti V, Barbagallo I, Rescifina A, Pittalà V. Potholing of the hydrophobic heme oxygenase-1 western region for the search of potent and selective imidazole-based inhibitors. Eur J Med Chem 2018; 148:54-62. [PMID: 29454190 DOI: 10.1016/j.ejmech.2018.02.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/29/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
Abstract
Here we report the design, synthesis, and molecular modeling of new potent and selective imidazole-based HO-1 inhibitors in which the imidazole nucleus and the hydrophobic groups are linked by a phenylethanolic spacer. Most of the tested compounds showed a good inhibitor activity with IC50 values in the low micromolar range, with two of them (1b and 1j) exhibiting also high selectivity toward HO-2. These results were obtained by the idea of potholing the entire volume of the principal hydrophobic western region with an appropriate ligand volume. Molecular modeling studies showed that these molecules bind to the HO-1 in the consolidated fashion where the imidazolyl moiety coordinates the heme iron while the aromatic groups are stabilized by hydrophobic interaction in the western region of the binding pocket. Finally, the synthesized compounds were analyzed for in silico ADME-Tox properties to establish oral drug-like behavior and showed satisfactory results.
Collapse
Affiliation(s)
- Loredana Salerno
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Romeo
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Floresta
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy; Department of Chemical Sciences, University of Catania, V.le A. Doria, 95125, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy
| | - Ignazio Barbagallo
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy
| | - Antonio Rescifina
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy.
| | - Valeria Pittalà
- Department of Drug Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy.
| |
Collapse
|
178
|
Kuang Y, Sechi M, Nurra S, Ljungman M, Neamati N. Design and Synthesis of Novel Reactive Oxygen Species Inducers for the Treatment of Pancreatic Ductal Adenocarcinoma. J Med Chem 2018; 61:1576-1594. [PMID: 29328656 DOI: 10.1021/acs.jmedchem.7b01463] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Altering redox homeostasis provides distinctive therapeutic opportunities for the treatment of pancreatic cancer. Quinazolinediones (QDs) are novel redox modulators that we previously showed to induce potent growth inhibition in pancreatic ductal adenocarcinoma (PDAC) cell lines. Our lead optimization campaign yielded QD325 as the most potent redox modulator candidate inducing substantial reactive oxygen species (ROS) in PDAC cells. Nascent RNA sequencing following treatments with the QD compounds revealed induction of stress responses in nucleus, endoplasmic reticulum, and mitochondria of pancreatic cancer cells. Furthermore, the QD compounds induced Nrf2-mediated oxidative stress and unfolded protein responses as demonstrated by dose-dependent increases in RNA synthesis of representative genes such as NQO1, HMOX1, DDIT3, and HSPA5. At higher concentrations, the QDs blocked mitochondrial function by inhibiting mtDNA transcription and downregulating the mtDNA-encoded OXPHOS enzymes. Importantly, treatments with QD325 were well tolerated in vivo and significantly delayed tumor growth in mice. Our study supports the development of QD325 as a new therapeutic in the treatment of PDAC.
Collapse
Affiliation(s)
- Yuting Kuang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan , 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States.,Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California , 1985 Zonal Avenue, Los Angeles, California 90033, United States
| | - Mario Sechi
- Department of Chemistry and Pharmacy, University of Sassari , Via Vienna 2, 07100 Sassari, Italy
| | - Salvatore Nurra
- Department of Chemistry and Pharmacy, University of Sassari , Via Vienna 2, 07100 Sassari, Italy
| | - Mats Ljungman
- Department of Radiation Oncology, University of Michigan , 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan , 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
179
|
Brum AM, van de Peppel J, Nguyen L, Aliev A, Schreuders-Koedam M, Gajadien T, van der Leije CS, van Kerkwijk A, Eijken M, van Leeuwen JPTM, van der Eerden BCJ. Using the Connectivity Map to discover compounds influencing human osteoblast differentiation. J Cell Physiol 2018; 233:4895-4906. [PMID: 29194609 DOI: 10.1002/jcp.26298] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/28/2017] [Indexed: 12/24/2022]
Abstract
Osteoporosis is a common skeletal disorder characterized by low bone mass leading to increased bone fragility and fracture susceptibility. Identification of factors influencing osteoblast differentiation and bone formation is very important. Previously, we identified parbendazole to be a novel compound that stimulates osteogenic differentiation of human mesenchymal stromal cells (hMSCs), using gene expression profiling and bioinformatic analyzes, including the Connectivity Map (CMap), as an in-silico approach. The aim for this paper is to identify additional compounds affecting osteoblast differentiation using the CMap. Gene expression profiling was performed on hMSCs differentiated to osteoblasts using Illumina microarrays. Our osteoblast gene signature, the top regulated genes 6 hr after induction by dexamethasone, was uploaded into CMap (www.broadinstitute.org/cmap/). Through this approach we identified compounds with gene signatures positively correlating (withaferin-A, calcium folinate, amylocaine) or negatively correlating (salbutamol, metaraminol, diprophylline) to our osteoblast gene signature. All positively correlating compounds stimulated osteogenic differentiation, as indicated by increased mineralization compared to control treated cells. One of three negatively correlating compounds, salbutamol, inhibited dexamethasone-induced osteoblastic differentiation, while the other two had no effect. Based on gene expression data of withaferin-A and salbutamol, we identified HMOX1 and STC1 as being strongly differentially expressed . shRNA knockdown of HMOX1 or STC1 in hMSCs inhibited osteoblast differentiation. These results confirm that the CMap is a powerful approach to identify positively compounds that stimulate osteogenesis of hMSCs, and through this approach we can identify genes that play an important role in osteoblast differentiation and could be targets for novel bone anabolic therapies.
Collapse
Affiliation(s)
- Andrea M Brum
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Linh Nguyen
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Abidin Aliev
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | - Tarini Gajadien
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | | | | | | | - B C J van der Eerden
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
180
|
Elbirt KK, Bonkovsky HL. Heme Oxygenase: Recent Advances in Understanding Its Regulation and Role. ACTA ACUST UNITED AC 2018. [DOI: 10.1111/paa.1999.111.5.438] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
181
|
Herroon MK, Rajagurubandara E, Diedrich JD, Heath EI, Podgorski I. Adipocyte-activated oxidative and ER stress pathways promote tumor survival in bone via upregulation of Heme Oxygenase 1 and Survivin. Sci Rep 2018; 8:40. [PMID: 29311669 PMCID: PMC5758829 DOI: 10.1038/s41598-017-17800-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/30/2017] [Indexed: 01/26/2023] Open
Abstract
Metastatic tumor cells engage the local tumor microenvironment and activate specific pro-survival mechanisms to thrive and progress in the harsh bone marrow niche. Here we show that the major contributors to the survival of carcinoma cells that have colonized the bone marrow are the adipocyte-induced oxidative stress and ER stress pathways. We demonstrate that upon exposure to adipocyte-rich environments in vitro or in vivo, bone-trophic prostate and breast tumor cells upregulate the oxidative stress enzyme, HO-1. We also show that HO-1 levels are significantly increased in human metastatic prostate cancer tissues and that stable HO-1 overexpression in tumor cells promotes growth and invasiveness. Co-incident with the adipocyte-induced expression of HO-1, there is an upregulation of ER chaperone BIP and splicing of XBP1, indicating adipocyte-driven unfolded protein response, a process that we show to be sensitive to antioxidant treatment. Importantly, we also demonstrate that triggering of the oxidative stress and ER stress responses, or HO-1 induction by adipocyte exposure result in the activation of pro-survival pathways, involving survivin. Collectively, our findings reveal a new link between HO-1 and survivin expression in tumor cells, and provide a new insight into potentially targetable survival pathways in bone-metastatic disease.
Collapse
Affiliation(s)
- Mackenzie K Herroon
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Jonathan D Diedrich
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Elisabeth I Heath
- Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA.
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
182
|
Genome-wide association and pathway analysis of left ventricular function after anthracycline exposure in adults. Pharmacogenet Genomics 2018; 27:247-254. [PMID: 28542097 DOI: 10.1097/fpc.0000000000000284] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Anthracyclines are important chemotherapeutic agents, but their use is limited by cardiotoxicity. Candidate gene and genome-wide studies have identified putative risk loci for overt cardiotoxicity and heart failure, but there has been no comprehensive assessment of genomic variation influencing the intermediate phenotype of anthracycline-related changes in left ventricular (LV) function. The purpose of this study was to identify genetic factors influencing changes in LV function after anthracycline chemotherapy. METHODS We conducted a genome-wide association study (GWAS) of change in LV function after anthracycline exposure in 385 patients identified from BioVU, a resource linking DNA samples to de-identified electronic medical record data. Variants with P values less than 1×10 were independently tested for replication in a cohort of 181 anthracycline-exposed patients from a prospective clinical trial. Pathway analysis was performed to assess combined effects of multiple genetic variants. RESULTS Both cohorts were middle-aged adults of predominantly European descent. Among 11 candidate loci identified in discovery GWAS, one single nucleotide polymorphism near PR domain containing 2, with ZNF domain (PRDM2), rs7542939, had a combined P value of 6.5×10 in meta-analysis. Eighteen Kyoto Encyclopedia of Gene and Genomes pathways showed strong enrichment for variants associated with the primary outcome. Identified pathways related to DNA repair, cellular metabolism, and cardiac remodeling. CONCLUSION Using genome-wide association we identified a novel candidate susceptibility locus near PRDM2. Variation in genes belonging to pathways related to DNA repair, metabolism, and cardiac remodeling may influence changes in LV function after anthracycline exposure.
Collapse
|
183
|
Barzegar M, Shaygannejad V, Mirmosayyeb O, Valipour S, Akbari R, Vesal S, Ashtar F, Dehghani L, Mohammadi A, Esmaeil N. Serum Bilirubin Level Changes in Multiple Sclerosis Patients. CASPIAN JOURNAL OF NEUROLOGICAL SCIENCES 2018. [DOI: 10.29252/nirp.cjns.4.12.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
184
|
Secher N, Østergaard L, Tønnesen E, Hansen FB, Granfeldt A. Impact of age on cardiovascular function, inflammation, and oxidative stress in experimental asphyxial cardiac arrest. Acta Anaesthesiol Scand 2018; 62:49-62. [PMID: 29072303 DOI: 10.1111/aas.13014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/23/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Advanced age is an independent predictor of poor outcome after cardiac arrest (CA). From experimental studies of regional ischemia-reperfusion injury, advanced age is associated with larger infarct size, reduced organ function, and augmented oxidative stress. The objective of this study was to investigate the effect of age on cardiovascular function, oxidative stress, inflammation, and endothelial activation after CA representing global ischemia-reperfusion. METHODS Aged (26 months) and young (5 months) rats were subjected to 8 min of asphyxia induced CA, resuscitated and observed for 360 min. Left ventricular pressure-derived cardiac function was measured at baseline and 360 min after CA. Blood samples obtained at baseline, 120 min, and 360 min after CA were analyzed for IL-1β, IL-6, IL-10, TNF-α, elastase, sE-selectin, sL-selectin, sI-CAM1, hemeoxygenase-1 (HO-1) and protein carbonyl. Tissue samples of brain, heart, kidney, and lung were analyzed for HO-1. RESULTS Cardiac function, evaluated by dP/dtmax and dP/dtmin , was decreased after CA in both young and aged rats, with no group differences. Mean arterial pressure increased after CA in young, but not old rats. Aged rats showed significantly higher plasma levels of elastase and sE-selectin after CA, and there was a significant different development over time between groups for IL-6 and IL-10. Young rats showed higher levels of HO-1 in plasma and renal tissue after CA. CONCLUSION In a rat model of asphyxial CA, advanced age is associated with an attenuated hyperdynamic blood pressure response and increased endothelial activation.
Collapse
Affiliation(s)
- N. Secher
- Department of Anaesthesiology and Intensive Care Medicine; Aarhus University Hospital; Aarhus C Denmark
- Department of Internal Medicine; Horsens Regional Hospital; Horsens Denmark
| | - L. Østergaard
- Center of Functionally Integrative Neuroscience; Aarhus University; Aarhus C Denmark
| | - E. Tønnesen
- Department of Anaesthesiology and Intensive Care Medicine; Aarhus University Hospital; Aarhus C Denmark
| | - F. B. Hansen
- Department of Anaesthesiology and Intensive Care Medicine; Aarhus University Hospital; Aarhus C Denmark
| | - A. Granfeldt
- Department of Anaesthesiology and Intensive Care Medicine; Aarhus University Hospital; Aarhus C Denmark
| |
Collapse
|
185
|
Wang B, Zhang J, Chen C, Xu G, Qin X, Hong Y, Bose DD, Qiu F, Zou Z. The size of zinc oxide nanoparticles controls its toxicity through impairing autophagic flux in A549 lung epithelial cells. Toxicol Lett 2017; 285:51-59. [PMID: 29289694 DOI: 10.1016/j.toxlet.2017.12.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
Zinc oxide nanoparticles (ZnONPs) widely used in various products, have been concerned with its impact on human health, in particular, on the risk of pulmonary toxicity. Our previous study indicated that ZnONPs could harness autophagy and impair the autophagic flux, which was positively linked to ZnONPs-induced toxicity. The objective of this study was to investigate whether ZnONPs-induced impairment of autophagic flux and cell death in lung epithelial cells is related to the size of ZnONPs. We demonstrate that ZnONPs with the average size of 50 nm could induce toxic effects in A549 lung epithelial cells, including accumulation of autophagosomes (the elevation of LC3B-II/LC3B-I ratio), impaired autophagic flux (the increase of p62 expression), the release of intracellular zinc ions (the increase of FluoZin-3 signal and ZnT1 mRNA expression), mitochondrial damage (the decrease of TMRE signal), lysosomal dysfunction (the aberrant expression of LAMP-2), oxidative stress (the increase of DCFH-DA signal and HO-1 expression) and cell death. Interestingly, ZnONPs with the average size of 200 nm failed to induce autophagy-mediated toxicity. Taken together, our results indicate that the size of ZnONPs is closely correlated with its toxicity, which is probably mediated by induction of impaired autophagic flux. This finding provides an insight into better understating of ZnONPs-associated toxicity, and mitigating the risk to humans and allowing the safer application.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing 400016, China; Post-doctoral Research Stations of Nursing Science, School of Nursing, Chongqing Medical University, Chongqing 400016, China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yueling Hong
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Diptiman D Bose
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
186
|
Lin CC, Chiang YC, Cho RL, Lin WN, Yang CC, Hsiao LD, Yang CM. Up-regulation of PYK2/PKCα-dependent haem oxygenase-1 by CO-releasing molecule-2 attenuates TNF-α-induced lung inflammation. Br J Pharmacol 2017; 175:456-468. [PMID: 29139546 DOI: 10.1111/bph.14094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/18/2017] [Accepted: 11/02/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Haem oxygenase-1 (HO-1) could provide cytoprotection against various inflammatory diseases. However, the mechanisms underlying the protective effect of CO-releasing molecule-2 (CORM-2)-induced HO-1 expression against TNF-α-induced inflammatory responses in human pulmonary alveolar epithelial cells (HPAEpiCs) remain unknown. EXPERIMENTAL APPROACH CORM-2-induced HO-1 protein and mRNA expression, and signalling pathways were determined by Western blot and real-time PCR, coupled with respective pharmacological inhibitors or transfection with siRNAs. The effect of CORM-2 on TNF-α-induced increase in leukocyte counts in BAL fluid and VCAM-1 expression in lung was determined by cell counting and Western blot analysis. KEY RESULTS CORM-2 attenuated the TNF-α-induced pulmonary haematoma, VCAM-1 expression and increase in leukocytes through an up-regulation of HO-1 in mice; this effect of CORM-2 was reversed by the HO-1 inhibitor zinc protoporphyrin IX. Furthermore, CORM-2 increased HO-1 protein and mRNA expression as well as the phosphorylation of PYK2, PKCα and ERK1/2 (p44/p42 MAPK) in HPAEpiCs; these effects were attenuated by their respective pharmacological inhibitors or transfection with siRNAs. Inhibition of PKCα by Gö6976 or Gö6983 attenuated CORM-2-induced stimulation of PKCα and ERK1/2 phosphorylation but had no effect on PYK2 phosphorylation. Moreover, inhibition of PYK2 by PF431396 reduced the phosphorylation of all three protein kinases. Finally, PYK2/PKCα/ERK1/2-mediated stimulation of activator protein 1 was shown to play a key role in CORM-2-induced HO-1 expression via an up-regulation of c-Fos mRNA. CONCLUSIONS AND IMPLICATIONS CORM-2 activates a PYK2/PKCα/ERK1/2/AP-1 pathway leading to HO-1 expression in HPAEpiCs. This HO-1/CO system might have potential as a therapeutic target in pulmonary inflammation.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anaesthetics, Chang Gung Memorial Hospital at Linkuo and Chang Gung University, Tao-Yuan, Taiwan
| | - Yu-Ching Chiang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Rou-Ling Cho
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chien-Chung Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anaesthetics, Chang Gung Memorial Hospital at Linkuo and Chang Gung University, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Anaesthetics, Chang Gung Memorial Hospital at Linkuo and Chang Gung University, Tao-Yuan, Taiwan.,Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Research Centre for Chinese Herbal Medicine and Research Centre for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| |
Collapse
|
187
|
Nway NC, Fujitani Y, Hirano S, Mar O, Win-Shwe TT. Role of TLR4 in olfactory-based spatial learning activity of neonatal mice after developmental exposure to diesel exhaust origin secondary organic aerosol. Neurotoxicology 2017; 63:155-165. [DOI: 10.1016/j.neuro.2017.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 01/24/2023]
|
188
|
Nakamura K, Zhang M, Kageyama S, Ke B, Fujii T, Sosa RA, Reed EF, Datta N, Zarrinpar A, Busuttil RW, Araujo JA, Kupiec-Weglinski JW. Macrophage heme oxygenase-1-SIRT1-p53 axis regulates sterile inflammation in liver ischemia-reperfusion injury. J Hepatol 2017; 67:1232-1242. [PMID: 28842295 PMCID: PMC5884687 DOI: 10.1016/j.jhep.2017.08.010] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Hepatic ischemia-reperfusion injury (IRI), characterized by exogenous antigen-independent local inflammation and hepatocellular death, represents a risk factor for acute and chronic rejection in liver transplantation. We aimed to investigate the molecular communication involved in the mechanism of liver IRI. METHODS We analyzed human liver transplants, primary murine macrophage cell cultures and IR-stressed livers in myeloid-specific heme oxygenase-1 (HO-1) gene mutant mice, for anti-inflammatory and cytoprotective functions of macrophage-specific HO-1/SIRT1 (sirtuin 1)/p53 (tumor suppressor protein) signaling. RESULTS Decreased HO-1 expression in human post-reperfusion liver transplant biopsies correlated with a deterioration in hepatocellular function (serum ALT; p<0.05) and inferior patient survival (p<0.05). In the low HO-1 liver transplant biopsy group, SIRT1/Arf (alternative reading frame)/p53/MDM2 (murine double minute 2) expression levels decreased (p<0.05) while cleaved caspase 3 and frequency of TUNEL+cells simultaneously increased (p<0.05). Immunofluorescence showed macrophages were the principal source of HO-1 in human and mouse IR-stressed livers. In vitro macrophage cultures revealed that HO-1 induction positively regulated SIRT1 signaling, whereas SIRT1-induced Arf inhibited ubiquitinating activity of MDM2 against p53, which in turn attenuated macrophage activation. In a murine model of hepatic warm IRI, myeloid-specific HO-1 deletion lacked SIRT1/p53, exacerbated liver inflammation and IR-hepatocellular death, whereas adjunctive SIRT1 activation restored p53 signaling and rescued livers from IR-damage. CONCLUSION This bench-to-bedside study identifies a new class of macrophages activated via the HO-1-SIRT1-p53 signaling axis in the mechanism of hepatic sterile inflammation. This mechanism could be a target for novel therapeutic strategies in liver transplant recipients. LAY SUMMARY Post-transplant low macrophage HO-1 expression in human liver transplants correlates with reduced hepatocellular function and survival. HO-1 regulates macrophage activation via the SIRT1-p53 signaling network and regulates hepatocellular death in liver ischemia-reperfusion injury. Thus targeting this pathway in liver transplant recipients could be of therapeutic benefit.
Collapse
Affiliation(s)
- Kojiro Nakamura
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Min Zhang
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Shoichi Kageyama
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Bibo Ke
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Takehiro Fujii
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Rebecca A. Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Nakul Datta
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Ali Zarrinpar
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Ronald W. Busuttil
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Jesus A. Araujo
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA,Corresponding authors. Addresses: Dumont-UCLA Transplant Center, 10833 Le Conte Ave, 77-120 CHS, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 4196; fax: +1 (310) 267 2358 (J.W. Kupiec-Weglinski) and UCLA Division of Cardiology, 10833 Le Conte Ave, CHS 43-264, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 3222; fax: +1 (310) 206 9133 (J.A. Araujo). (J.A. Araujo), (J.W. Kupiec-Weglinski)
| | - Jerzy W. Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA,Corresponding authors. Addresses: Dumont-UCLA Transplant Center, 10833 Le Conte Ave, 77-120 CHS, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 4196; fax: +1 (310) 267 2358 (J.W. Kupiec-Weglinski) and UCLA Division of Cardiology, 10833 Le Conte Ave, CHS 43-264, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 3222; fax: +1 (310) 206 9133 (J.A. Araujo). (J.A. Araujo), (J.W. Kupiec-Weglinski)
| |
Collapse
|
189
|
Alcalde-Estévez E, Arroba AI, Sánchez-Fernández EM, Mellet CO, García Fernández JM, Masgrau L, Valverde ÁM. The sp 2-iminosugar glycolipid 1-dodecylsulfonyl-5N,6O-oxomethylidenenojirimycin (DSO 2-ONJ) as selective anti-inflammatory agent by modulation of hemeoxygenase-1 in Bv.2 microglial cells and retinal explants. Food Chem Toxicol 2017; 111:454-466. [PMID: 29191728 DOI: 10.1016/j.fct.2017.11.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
Abstract
Neuroinflammation is an early event during diabetic retinopathy (DR) that impacts the dynamics of microglia polarization. Gliosis is a hallmark of DR and we have reported the beneficial effects of 1R-DSO-ONJ, a member of the sp2-iminosugar glycolipid (sp2-IGL) family, in targeting microglia and reducing gliosis in diabetic db/db mice. Herein, we analyzed the effect of DSO2-ONJ, another family compound incorporating a sulfone group that better mimics the phosphate group of phosphatidylinositol ether lipid analogues (PIAs), in Bv.2 microglial cells treated with bacterial lipopolysaccaride (LPS) and in retinal explants from db/db mice. In addition to decreasing iNOS and inflammasome activation, the anti-inflammatory effect of DSO2-ONJ was mediated by direct p38α MAPK activation. Computational docking experiments demonstrated that DSO2-ONJ binds to p38α MAPK at the same site where PIAs and the alkyl phospholipid perifosine activators do, suggesting similar mechanism of action. Moreover, treatment of microglial cells with DSO2-ONJ increased both heme-oxygenase (HO)-1 and Il10 expression regardless the presence of LPS. In retinal explants from db/db mice, DSO2-ONJ also induced HO-1 and reduced gliosis. Since IL-10-mediated induction of HO-1 expression is mediated by p38α MAPK activation, our results suggest that this molecular mechanism is involved in the anti-inflammatory effects of DSO2-ONJ in microglia.
Collapse
Affiliation(s)
- Elena Alcalde-Estévez
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm) (CSIC/UAM), C/ Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029 Madrid, Spain.
| | - Ana I Arroba
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm) (CSIC/UAM), C/ Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029 Madrid, Spain.
| | | | - Carmen Ortiz Mellet
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain.
| | - Jose M García Fernández
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Avda. Américo Vespucio 49, 41092 Sevilla, Spain.
| | - Laura Masgrau
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm) (CSIC/UAM), C/ Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029 Madrid, Spain.
| |
Collapse
|
190
|
Lewallen EA, Salib CG, Trousdale WH, Berry CE, Hanssen GM, Robin JX, Tibbo ME, Viste A, Reina N, Morrey ME, Sanchez-Sotelo J, Hanssen AD, Berry DJ, van Wijnen AJ, Abdel MP. Molecular pathology of total knee arthroplasty instability defined by RNA-seq. Genomics 2017; 110:247-256. [PMID: 29174847 DOI: 10.1016/j.ygeno.2017.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 11/15/2022]
Abstract
Total knee arthroplasty (TKA) is a durable and reliable procedure to alleviate pain and improve joint function. However, failures related to flexion instability sometimes occur. The goal of this study was to define biological differences between tissues from patients with and without flexion instability of the knee after TKA. Human knee joint capsule tissues were collected at the time of primary or revision TKAs and analyzed by RT-qPCR and RNA-seq, revealing novel patterns of differential gene expression between the two groups. Interestingly, genes related to collagen production and extracellular matrix (ECM) degradation were higher in samples from patients with flexion instability. Partitioned clustering analyses further emphasized differential gene expression patterns between sample types that may help guide clinical interpretations of this complication. Future efforts to disentangle the effects of physical and biological (e.g., transcriptomic modifications) risk factors will aid in further characterizing and avoiding flexion instability after TKA.
Collapse
Affiliation(s)
- Eric A Lewallen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States; Department of Biological Sciences, Hampton University, Hampton, VA, United States.
| | - Christopher G Salib
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - William H Trousdale
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Charlotte E Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Joseph X Robin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Meagan E Tibbo
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Anthony Viste
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Nicolas Reina
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Mark E Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | | | - Arlen D Hanssen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Daniel J Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States; Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, United States.
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
191
|
Kim KW, Kim HJ, Sohn JH, Yim JH, Kim YC, Oh H. Anti-neuroinflammatory effect of 6,8,1'-tri-O-methylaverantin, a metabolite from a marine-derived fungal strain Aspergillus sp., via upregulation of heme oxygenase-1 in lipopolysaccharide-activated microglia. Neurochem Int 2017; 113:8-22. [PMID: 29174381 DOI: 10.1016/j.neuint.2017.11.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/06/2017] [Accepted: 11/20/2017] [Indexed: 12/30/2022]
Abstract
In the course of searching for anti-neuroinflammatory metabolites from marine-derived fungi, three fungal metabolites, 6,8,1'-tri-O-methylaverantin, 6,8-di-O-methylaverufin, and 5-methoxysterigmatocystin were isolated from a marine-derived fungal strain Aspergillus sp. SF-6796. Among these, 6,8,1'-tri-O-methylaverantin induced the expression of heme oxygenase (HO)-1 protein in BV2 microglial cells. The induction of HO-1 protein was mediated by the activation of nuclear transcription factor erythroid-2 related factor 2 (Nrf2), and was regulated by the p38 mitogen-activated protein kinase and phosphatidylinositol 3-kinase/protein kinase B signaling pathways. Furthermore, 6,8,1'-tri-O-methylaverantin suppressed the overproduction of pro-inflammatory mediators, such as nitric oxide, prostaglandin E2, inducible nitric oxide synthase, and cyclooxygenase-2 in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. These anti-neuroinflammatory effects were mediated through the negative regulation of the nuclear factor kappa B pathway, repressing the phosphorylation and degradation of inhibitor kappa B-α, translocation into the nucleus of p65/p50 heterodimer, and DNA-binding activity of p65 subunit. The anti-neuroinflammatory effect of 6,8,1'-tri-O-methylaverantin was partially blocked by a selective HO-1 inhibitor, suggesting that its anti-neuroinflammatory effect is at least partly mediated by HO-1 induction. In this study, 6,8,1'-tri-O-methylaverantin also induced HO-1 protein expression in primary microglial cells, and this correlated with anti-neuroinflammatory effects observed in LPS-stimulated primary microglial cells. In conclusion, 6,8,1'-tri-O-methylaverantin represents a potential candidate for use in the development of therapeutic agents for the regulation of neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Kwan-Woo Kim
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
| | - Hye Jin Kim
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
| | - Jae Hak Sohn
- College of Medical and Life Sciences, Silla University, Busan 46958, Republic of Korea
| | - Joung Han Yim
- Korea Polar Research Institute, KORDI, Yeonsu-gu, Incheon 21990, Republic of Korea
| | - Youn-Chul Kim
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
| | - Hyuncheol Oh
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea.
| |
Collapse
|
192
|
Lin CC, Yang CC, Hsiao LD, Chen SY, Yang CM. Heme Oxygenase-1 Induction by Carbon Monoxide Releasing Molecule-3 Suppresses Interleukin-1β-Mediated Neuroinflammation. Front Mol Neurosci 2017; 10:387. [PMID: 29209167 PMCID: PMC5701945 DOI: 10.3389/fnmol.2017.00387] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/07/2017] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative disorders and brain damage are initiated by excessive production of reactive oxygen species (ROS), which leads to tissue injury, cellular death and inflammation. In cellular anti-oxidant systems, heme oxygenase-1 (HO-1) is an oxidative-sensor protein induced by ROS generation or carbon monoxide (CO) release. CO releasing molecules (CORMs), including CORM-3, exert anti-oxidant and anti-inflammatory effects. However, the molecular mechanisms of CORM-3-induced HO-1 expression and protection against interleukin (IL)-1β-induced inflammatory responses have not been fully elucidated in rat brain astrocytes (RBA-1). To study the regulation of CORM-3-induced HO-1 expression, signaling pathways, promoter activity, mRNA and protein expression were assessed following treatment with pharmacological inhibitors and gene-specific siRNA knockdown. We found that CORM-3 mediated HO-1 induction via transcritional and translational processes. Furthermore, CORM-3-induced HO-1 expression was mediated by phosphorylation of several protein kinases, such as c-Src, Pyk2, protein kinase Cα (PKCα) and p42/p44 mitogen-activated protein kinase (MAPK), which were inhibited by respective pharmacological inhibitors or by gene-specific knockdown with siRNA transfections. Next, we found that CORM-3 sequentially activated the c-Src/Pyk2/PKCα/p42/p44 MAPK pathway, thereby up-regulating mRNA for the activator protein (AP)-1 components c-Jun and c-Fos; these effects were attenuated by an AP-1 inhibitor (Tanshinone IIA; TSIIA) and other relevant inhibitors. Moreover, CORM-3-induced upregulation of HO-1 attenuated the IL-1β-induced cell migration and matrix metallopeptidase-9 mRNA expression in RBA-1 cells. These effects were reversed by an matrix metalloproteinase (MMP)2/9 inhibitor or by transfection with HO-1 siRNA.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou, and College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan.,Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou, and College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Ssu-Yu Chen
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou, and College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Research Center for Industry of Human Ecology, Research Center for Chinese Herbal Medicine, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| |
Collapse
|
193
|
Turolo S, Edefonti A, Syren ML, Marangoni F, Morello W, Agostoni C, Montini G. Fatty Acids in Nephrotic Syndrome and Chronic Kidney Disease. J Ren Nutr 2017; 28:145-155. [PMID: 29153556 DOI: 10.1053/j.jrn.2017.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/14/2022] Open
Abstract
The role of fatty acids (FAs) in inflammation and in the related chronic diseases has been demonstrated. However, there is a lack of consistent and agreed knowledge about the role of FA profile and renal physiology and pathology, most articles focusing on the effect of polyunsaturated FAs supplementation, without considering the impact of basal FA metabolism on the efficacy of the supplementation. Here, we have summarized the specific literature concerning the assessment of circulating FA in 2 renal diseases, namely nephrotic syndrome and chronic kidney disease, also under hemodialytic treatment, and have received the most significant contributions in the last years. The effects of changes of FA profile and metabolism and the possible involvement of polyunsaturated FA metabolites in raising and modulating inflammation are discussed.
Collapse
Affiliation(s)
- Stefano Turolo
- Pediatric Department of Nephrology Dialysis and Transplantation, IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Alberto Edefonti
- Pediatric Department of Nephrology Dialysis and Transplantation, IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marie Louise Syren
- Pediatric Clinic IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - William Morello
- Pediatric Department of Nephrology Dialysis and Transplantation, IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Carlo Agostoni
- Pediatric Clinic IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giovanni Montini
- Pediatric Department of Nephrology Dialysis and Transplantation, IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Pediatric Clinic IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
194
|
Hargitai R, Boross N, Hámori S, Neuberger E, Nyiri Z. Eggshell Biliverdin and Protoporphyrin Pigments in a Songbird: Are They Derived from Erythrocytes, Blood Plasma, or the Shell Gland? Physiol Biochem Zool 2017; 90:613-626. [DOI: 10.1086/694297] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
195
|
Gallic acid, a natural polyphenol, protects against tert-butyl hydroperoxide- induced hepatotoxicity by activating ERK-Nrf2-Keap1-mediated antioxidative response. Food Chem Toxicol 2017; 119:479-488. [PMID: 29066411 DOI: 10.1016/j.fct.2017.10.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 02/07/2023]
Abstract
Gallic acid (GA), a natural polyphenol, has been shown to exert a variety of heath promoting effects. We herein investigated the critical role of nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response in the protection of GA against tert-butyl hydroperoxide (t-BHP)-induced hepatotoxicity in L02 cells. Pretreatment of GA prevented the hepatocytotoxicity induced by t-BHP, as evidenced by the facts that GA suppressed t-BHP-induced cytotoxicity and reactive oxygen species (ROS) generation. GA induced nuclear translocation of Nrf2 along with expression of target proteins, including heme oxygenase-1 (HO-1) and glutamate cysteine ligase catalytic modify subunit (GCLC), and increased intracellular glutathione (GSH) content. Additionally, GA induced phosphorylated activation of extracellular regulated kinase (ERK), and ERK inhibitor PD98059 partially decreased GA-induced hepatoprotection, and downregulated the increased protein expressions of Nrf2, GCLC and HO-1 induced by GA. Interestingly, we found that GA could enhance the thermal stability of Keap1, which indicated the potential interaction between GA and Keap1. Furthermore, molecular docking indicated that GA possibly competed with Nrf2 for binding to Keap1. Collectively, GA effectively protects against t-BHP-induced hepatotoxicity via inducing ERK/Nrf2-mediated antioxidative signaling pathway. Meanwhile, GA disturbs protein-protein interaction between Keap1 and Nrf2 which might also contribute to nuclear translocation of Nrf2.
Collapse
|
196
|
Almeida BFM, Silva KLO, Venturin GL, Chiku VM, Leal AAC, Bosco AM, Ciarlini PC, Lima VMF. Induction of haem oxygenase-1 increases infection of dog macrophages by L. infantum. Parasite Immunol 2017; 39. [PMID: 28929503 DOI: 10.1111/pim.12494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/13/2017] [Indexed: 11/28/2022]
Abstract
We aimed to induce and inhibit HO-1, ascertaining its effect on infection rate, parasite load and the levels of superoxide, reactive oxygen species (ROS), nitric oxide (NO), TNF-alpha and IL-10 in cultured macrophages from healthy dogs infected by Leishmania infantum. Macrophages obtained from 15 healthy dogs were cultured alone or infected with L. infantum, with or without association of HO-1 inducer and inhibitor. The infection rate and the parasite load were determined by the number of infected macrophages and number of promastigotes per macrophage, respectively. HO-1 levels and gene expression, as well as IL-10 and TNF-alpha levels were also measured in these cultures. Superoxide, ROS and NO levels in macrophages were measured through flow cytometry. Induction of HO-1 increased the infection rate and parasite load, while its inhibition decreased the infection rate and IL-10 production. There was a positive correlation between HO-1 and infection rate or parasite load. Increased infection rate was associated with decreased superoxide, ROS and NO levels. Induction of HO-1 metabolism in dogs infected by L. infantum is possibly one of the mechanisms responsible for increasing the infection of macrophages, mainly through reduction in the oxidative and nitrosative metabolisms of these cells.
Collapse
Affiliation(s)
- B F M Almeida
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| | - K L O Silva
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| | - G L Venturin
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| | - V M Chiku
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| | - A A C Leal
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| | - A M Bosco
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| | - P C Ciarlini
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| | - V M F Lima
- Immunology Laboratory, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil.,Department of Animal Internal Medicine, Surgery and Reproduction, School of Veterinary Medicine, Araçatuba Campus, São Paulo State University (UNESP), Araçatuba, Brazil
| |
Collapse
|
197
|
Miltonprabu S, Tomczyk M, Skalicka-Woźniak K, Rastrelli L, Daglia M, Nabavi SF, Alavian SM, Nabavi SM. Hepatoprotective effect of quercetin: From chemistry to medicine. Food Chem Toxicol 2017; 108:365-374. [DOI: 10.1016/j.fct.2016.08.034] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/04/2016] [Accepted: 08/30/2016] [Indexed: 12/21/2022]
|
198
|
Propolis reversed cigarette smoke-induced emphysema through macrophage alternative activation independent of Nrf2. Bioorg Med Chem 2017; 25:5557-5568. [DOI: 10.1016/j.bmc.2017.08.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/07/2017] [Accepted: 08/15/2017] [Indexed: 01/01/2023]
|
199
|
Kim JY, Lee H, Lee EJ, Kim M, Kim TG, Kim HP, Oh SH. Keap1 knockdown in melanocytes induces cell proliferation and survival via HO-1-associated β-catenin signaling. J Dermatol Sci 2017; 88:85-95. [PMID: 28583303 DOI: 10.1016/j.jdermsci.2017.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/21/2017] [Accepted: 05/15/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Nrf2-Keap1 signaling pathway protects cells against photo-oxidative stress. Yet in recent works, its role in melanogenesis together with cell protection functions against oxidative stress has been gaining interest. However, its effect on melanogenesis still has contradictory results from different studies. OBJECTIVE The aims of our study were to investigate the effect of Keap1 silencing in melanocyte on melanogenesis and its associated mechanism. METHODS Primary human epidermal melanocytes and melan-a cell line were used for this experiment. RNA sequencing was done to identify genes involved in melanocyte biology using Keap1 knockdown through siRNA techniques. And melanogenesis and the expression of melanogenesis-associated molecules were evaluated in Keap1 silenced melanocyte to examine the effects of Keap1 on melanogenesis, melanocyte growth, and related pathways. RESULTS RNA-sequencing data revealed that Keap1 knockdown in primary human epidermal melanocytes (PHEMs) induced cell survival-related gene expression. Additionally, siRNA-mediated inhibition of Keap1 led to upregulation of MITF and melanogenesis-associated molecules along with Nrf2 activation in PHEMs. HO-1, a major gene that is upregulated in RNA-sequencing using Keap1-silenced PHEMs, protected melanocytes against H2O2-induced cell death and upregulated MITF and β-catenin expression. Further, increased expression of melanogenesis-associated molecules after Keap1 silencing was validated to occur through HO-1-associated β-catenin activation in a Keap1 and HO-1 double knockdown experiment. CONCLUSION This work suggests that Keap1 silencing in melanocytes induced melanogenesis and the expression of melanogenesis-associated molecules through HO-1-associated β-catenin activation. Keap1 downregulation in melanocytes is important for cell proliferation and survival.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hemin Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mikyoung Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Gyun Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Ho Oh
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
200
|
Zhao L, Feng Y, Shi A, Zhang L, Guo S, Wan M. Neuroprotective Effect of Low-Intensity Pulsed Ultrasound Against MPP +-Induced Neurotoxicity in PC12 Cells: Involvement of K2P Channels and Stretch-Activated Ion Channels. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1986-1999. [PMID: 28583325 DOI: 10.1016/j.ultrasmedbio.2017.04.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 06/07/2023]
Abstract
Parkinson's disease is the second most common neurodegenerative disease. It is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta. 1-Methyl-4-phenylpyridinium (MPP+) is a dopaminergic neuronal toxin that is widely used in constructing Parkinson's disease models in vitro. Low-intensity pulsed ultrasound (LIPUS) is a non-invasive therapeutic approach that has neuromodulation and neuroprotective effects in the central neural system; however, whether LIPUS can provide protection for dopaminergic neurons against MPP+-induced neurocytotoxicity remains unknown. In this study, we found that pre-treatment with LIPUS (1 MHz, 50 mW/cm2, 20% duty cycle and 100-Hz pulse repetition frequency, 10 min) inhibited MPP+-induced neurotoxicity and mitochondrial dysfunction in PC12 cells. LIPUS decreased MPP+-induced oxidative stress by modulating antioxidant proteins, including thioredoxin-1 and heme oxygenase-1, and prevented neurocytotoxicity via the phosphoinositide 3-kinase (PI3K)-Akt and ERK1/2 pathways. Furthermore, these beneficial effects were attributed to the activation of K2P channels and stretch-activated ion channels by LIPUS. These data indicate that LIPUS protects neuronal cells from MPP+-induced cell death through the K2P channel- and stretch-activated ion channel-mediated downstream pathways. The data also suggest that LIPUS could be a promising therapeutic method in halting or retarding the degeneration of dopaminergic neurons in Parkinson's disease in a non-invasive manner.
Collapse
Affiliation(s)
- Lu Zhao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yi Feng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Aiwei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Lei Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shifang Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Mingxi Wan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|