1
|
Lu W, Yang X, Wang B. Carbon monoxide potentiates the effect of corticosteroids in suppressing inflammatory responses in cell culture. Bioorg Med Chem 2025; 120:118092. [PMID: 39904198 DOI: 10.1016/j.bmc.2025.118092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Inflammation is a pathology implicated in a wide range of human diseases. Recent years have seen tremendous progress in developing new types of anti-inflammatory agents for the treatment of inflammation of various origins. However, each has its own strengths and weaknesses. The very fact that there needs to have multiple types of anti-inflammatory agents underlines the complexity of inflammatory diseases and conditions, their molecular origins, and their treatment. Such complexity dictates the need to search for new approaches with improved potency and efficacy as well as reduced side effects. For these reasons, we are interested in exploring the possibility of generating synergy between carbon monoxide (CO), an endogenously produced cytoprotective agent, and known anti-inflammatory agents. Herein, we report the potentiating actions of CO on the anti-inflammatory effects of cortisone and dexamethasone as demonstrated in their ability to suppress the expression of TNF-α and IL-6 induced by either LPS or the S protein of SARS-CoV-2. Such effects are reflected in the substantially increased potency as well efficacy, when the efficacy of the corticosteroid alone does not allow for complete suppression of the expression of these cytokines. Further, increased attenuation of p65 phosphorylation is at least part of the molecular mechanism for the observed potentiating effects. We hope our work will stimulate a high level of activity along the same direction, leading to anti-inflammatory strategies with improved potency and efficacy and reduced side effects.
Collapse
Affiliation(s)
- Wen Lu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
2
|
Gagarinskiy EL, Sharapov MG, Goncharov RG, Gurin AE, Ugraitskaya SV, Fesenko EE. The effectiveness of prolonged hypothermic preservation of isolated rat hearts using oxygen, medical nitrous oxide and carbon monoxide gas mixtures. Arch Biochem Biophys 2025; 765:110295. [PMID: 39798642 DOI: 10.1016/j.abb.2025.110295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/23/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
The possibility of using an oxygen-nitrous oxide mixture for prolonged hypothermic preservation of rat heart for 24 h was investigated. A comparative analysis of restoration of functional activity of hearts in the groups of 24-h preservation at +4 °C with different gases (O2, N2) and gas mixtures (CO + O2, N2O + O2, N2+O2, N2O + N2) was carried out. It was shown that the presence of oxygen in the gas mixture was the key factor for heart preservation. No stable heart preservation was observed in oxygen-free mixtures. At the same time, preservation in pure oxygen showed a significantly lower level of cardiac recovery compared to preservation in gas mixtures O2+CO (6.5 atm.) and O2+N2O (6.5 atm.). LVDP (left ventricular developed pressure) values were 30 ± 19 mmHg and 46 ± 9 mmHg, respectively, with no significant differences found. The decrease in LDVP after 24 h of storage was 26-40 % of the intact control. The results obtained indicate the presence of pronounced synergistic effects of both gases during 24-h heart preservation, which is confirmed by data of marker genes Nfe2l2, Nox1, Prdx1, Hif1a, Nos2, Slc2a4, Ucp-1, Jun, Casp3 expression analysis and myocardial infarction damage level data. The more frequent occurrence of arrhythmias was observed in the oxygen-nitrous oxide group compared with the CO group, and the mechanism of this phenomenon is unclear. Nevertheless, the already medically approved N2O + O2 gas mixture could serve as a balanced choice for future improvements, offering a shorter duration of cardiac preservation compared to the CO + O2 mixture, while ensuring safety in its use.
Collapse
Affiliation(s)
- Evgeniy L Gagarinskiy
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Mars G Sharapov
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Ruslan G Goncharov
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Artem E Gurin
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Svetlana V Ugraitskaya
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| | - Eugeny E Fesenko
- Institute of Cell Biophysics RAS - a Separate Subdivision of Federal Research Centre "Pushchino Scientific Centre for Biological Research RAS", Institutskaya St., 3, 142290, Russia, Moscow Region, Pushchino.
| |
Collapse
|
3
|
Bauer N, Liu D, Nguyen T, Wang B. Unraveling the Interplay of Dopamine, Carbon Monoxide, and Heme Oxygenase in Neuromodulation and Cognition. ACS Chem Neurosci 2024; 15:400-407. [PMID: 38214656 PMCID: PMC10853931 DOI: 10.1021/acschemneuro.3c00742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
The dopaminergic system plays important roles in neuromodulation, including prominent roles in complex neurological functions such as cognition, reward, motivation, and memory. Understandably, the highly complex nature of such physiological functions means that their regulation is intertwined with other signaling pathways, as has been demonstrated by numerous studies. Contrary to its public perception of being poisonous at all concentrations, carbon monoxide (CO) is produced endogenously from heme degradation by heme oxygenase (HO) as part of the physiological process of red blood cell turnover. Physiological concentrations of CO can reach high micromolar ranges in the hemoglobin bound form. Low-dose CO has shown therapeutic effects in numerous animal models, including traumatic brain injury via engaging various hemoprotein targets. As such, the HO-CO axis has been shown to offer beneficial effects in organ protection, anti-inflammation, and neuroprotection, among many others. Further, a large number of publications have shown the interactions among CO, HO, and the dopaminergic system. In this review, we critically examine such experimental evidence in a holistic fashion and in the context of a possible dopamine-HO-CO signaling axis. We hope that this Perspective will stimulate additional investigations into the molecular connectivity related to this possible axis and open doors to the development of novel therapeutics that impact the dopaminergic system.
Collapse
Affiliation(s)
- Nicola Bauer
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Dongning Liu
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - TanPhat Nguyen
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Binghe Wang
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
4
|
Franzini M, Valdenassi L, Pandolfi S, Tirelli U, Ricevuti G, Chirumbolo S. The Role of Ozone as an Nrf2-Keap1-ARE Activator in the Anti-Microbial Activity and Immunity Modulation of Infected Wounds. Antioxidants (Basel) 2023; 12:1985. [PMID: 38001838 PMCID: PMC10669564 DOI: 10.3390/antiox12111985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Ozone is an allotrope of oxygen, widely known to exert an anti-oxidant potential. The ability of low, controlled and standardized doses of ozone in the ozone adjunct treatment of bacterial infections, which occur in wounds, is engaging clinical research to deepen the role of ozone in eradicating even multidrug-resistant bacteria. Ozone activates the nuclear factor erythroid 2-related factor 2 (Nrf2), and this activation triggers a complex cascade of events, which ultimately leads to macrophage training and an improvement in their ability to operate a clearance of bacteria in the patient's anatomical districts. In this review, we try to elucidate the recent evidence about the mechanisms with which ozone can actually remove bacteria and even multi-drug-resistant (MDR) bacteria, accounting on its complex ability in modulating immunity.
Collapse
Affiliation(s)
- Marianno Franzini
- Italian Scientific Society of Oxygen-Ozone Therapy (SIOOT), 24020 Bergamo, Italy; (M.F.); (L.V.); (S.P.)
| | - Luigi Valdenassi
- Italian Scientific Society of Oxygen-Ozone Therapy (SIOOT), 24020 Bergamo, Italy; (M.F.); (L.V.); (S.P.)
| | - Sergio Pandolfi
- Italian Scientific Society of Oxygen-Ozone Therapy (SIOOT), 24020 Bergamo, Italy; (M.F.); (L.V.); (S.P.)
| | | | - Giovanni Ricevuti
- Department of Drug Science, University of Pavia, 27100 Pavia, Italy;
| | - Salvatore Chirumbolo
- Department of Engineering for Innovation Medicine, University of Verona, 37125 Verona, Italy
| |
Collapse
|
5
|
Robert B, Subramaniam S. Gasotransmitter-Induced Therapeutic Angiogenesis: A Biomaterial Prospective. ACS OMEGA 2022; 7:45849-45866. [PMID: 36570231 PMCID: PMC9773187 DOI: 10.1021/acsomega.2c05599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Gasotransmitters such as NO, H2S, and CO have emerged as key players in the regulation of various pathophysiological functions, prompting the development of gas therapy for various pathogeneses. Deficient production of gasotransmitters has been linked to various diseases such as hypertension, endothelial dysfunction, myocardial infarction, ischemia, and impaired wound healing, as they are involved in the regulatory action of angiogenesis. A better understanding of the regulatory mechanisms has given new hope to address the vascular impairment caused by the breakthroughs in gasotransmitters as therapeutics. However, the unstable nature and poor target specificity of gas donors limit the full efficacy of drugs. In this regard, biomaterials that possess excellent biocompatibility and porosity are ideal drug carriers to deliver the gas transmitters in a tunable manner for therapeutic angiogenesis. This review article provides a comprehensive discussion of biomaterial-based gasotransmitter delivery approaches for therapeutic angiogenesis. The critical role of gasotransmitters in modulating angiogenesis during tissue repair as well as their challenges and future directions are demonstrated.
Collapse
Affiliation(s)
- Becky Robert
- Biomaterials
and Bioprocess Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore 641046, India
| | - Sadhasivam Subramaniam
- Biomaterials
and Bioprocess Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore 641046, India
- Department
of Extension and Career Guidance, Bharathiar
University, Coimbatore 641046, India
| |
Collapse
|
6
|
Yuan Z, De La Cruz LK, Yang X, Wang B. Carbon Monoxide Signaling: Examining Its Engagement with Various Molecular Targets in the Context of Binding Affinity, Concentration, and Biologic Response. Pharmacol Rev 2022; 74:823-873. [PMID: 35738683 PMCID: PMC9553107 DOI: 10.1124/pharmrev.121.000564] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Carbon monoxide (CO) has been firmly established as an endogenous signaling molecule with a variety of pathophysiological and pharmacological functions, including immunomodulation, organ protection, and circadian clock regulation, among many others. In terms of its molecular mechanism(s) of action, CO is known to bind to a large number of hemoproteins with at least 25 identified targets, including hemoglobin, myoglobin, neuroglobin, cytochrome c oxidase, cytochrome P450, soluble guanylyl cyclase, myeloperoxidase, and some ion channels with dissociation constant values spanning the range of sub-nM to high μM. Although CO's binding affinity with a large number of targets has been extensively studied and firmly established, there is a pressing need to incorporate such binding information into the analysis of CO's biologic response in the context of affinity and dosage. Especially important is to understand the reservoir role of hemoglobin in CO storage, transport, distribution, and transfer. We critically review the literature and inject a sense of quantitative assessment into our analyses of the various relationships among binding affinity, CO concentration, target occupancy level, and anticipated pharmacological actions. We hope that this review presents a picture of the overall landscape of CO's engagement with various targets, stimulates additional research, and helps to move the CO field in the direction of examining individual targets in the context of all of the targets and the concentration of available CO. We believe that such work will help the further understanding of the relationship of CO concentration and its pathophysiological functions and the eventual development of CO-based therapeutics. SIGNIFICANCE STATEMENT: The further development of carbon monoxide (CO) as a therapeutic agent will significantly rely on the understanding of CO's engagement with therapeutically relevant targets of varying affinity. This review critically examines the literature by quantitatively analyzing the intricate relationships among targets, target affinity for CO, CO level, and the affinity state of carboxyhemoglobin and provide a holistic approach to examining the molecular mechanism(s) of action for CO.
Collapse
Affiliation(s)
- Zhengnan Yuan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Ladie Kimberly De La Cruz
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| |
Collapse
|
7
|
Ranjana M, Sunil D. Naphthalimide derivatives as fluorescent probes for imaging endogenous gasotransmitters. Chem Biol Interact 2022; 363:110022. [PMID: 35753358 DOI: 10.1016/j.cbi.2022.110022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/07/2022] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Gasotransmitters have gained significant recognition attributed to their evident biological impacts, and is accepted as a promising and less-explored area with immense research scope. The three-member family comprising of nitric oxide, carbon monoxide and hydrogen sulphide as endogenous gaseous signaling molecules have been found to elicit a plethora of crucial biological functions, spawning a new research area. The sensing of these small molecules is vital to gain deeper insights into their functions, as they can act both as a friend or a foe in mammalian systems. The initial sections of the review present the physiological and pathophysiological roles of these endogenous gas transmitters and their synergistic interactions. Further, various detection approaches, especially the usage of fascinating features of 1,8-naphthalimide as fluorescent probe in the detection and monitoring of these small signaling molecules are highlighted. The current limitations and the future scope of improving the sensing of the three gasotransmitters are also discussed.
Collapse
Affiliation(s)
- M Ranjana
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India.
| |
Collapse
|
8
|
Suliman HB, Healy Z, Zobi F, Kraft BD, Welty-Wolf K, Smith J, Barkauskas C, Piantadosi CA. Nuclear respiratory factor-1 negatively regulates TGF-β1 and attenuates pulmonary fibrosis. iScience 2022; 25:103535. [PMID: 34977500 PMCID: PMC8683592 DOI: 10.1016/j.isci.2021.103535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/02/2021] [Accepted: 11/25/2021] [Indexed: 12/27/2022] Open
Abstract
The preclinical model of bleomycin-induced lung fibrosis is useful to study mechanisms related to human pulmonary fibrosis. Using BLM in mice, we find low HO-1 expression. Although a unique Rhenium-CO-releasing molecule (ReCORM) up-regulates HO-1, NRF-1, CCN5, and SMAD7, it reduces TGFβ1, TGFβr1, collagen, α-SMA, and phosphorylated Smad2/3 levels in mouse lung and in human lung fibroblasts. ChIP assay studies confirm NRF-1 binding to the promoters of TGFβ1 repressors CCN5 and Smad7. ReCORM did not blunt lung fibrosis in Hmox1-deficient alveolar type 2 cell knockout mice, suggesting this gene participates in lung protection. In human lung fibroblasts, TGFβ1-dependent production of α-SMA is abolished by ReCORM or by NRF-1 gene transfection. We demonstrate effective HO-1/NRF-1 signaling in lung AT2 cells protects against BLM induced lung injury and fibrosis by maintaining mitochondrial health, function, and suppressing the TGFβ1 pathway. Thus, protection of AT2 cell mitochondrial integrity via HO-1/NRF-1 presents an innovative therapeutic target.
Collapse
Affiliation(s)
- Hagir B. Suliman
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
- Department of Anaesthesiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Zachary Healy
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Fabio Zobi
- Department of Chemistry, University of Fribourg, Fribourg, Switzerland
| | - Bryan D. Kraft
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Karen Welty-Wolf
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Joshua Smith
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Christina Barkauskas
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Claude A. Piantadosi
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
- Department of Anaesthesiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
9
|
Zhou B, Sun X, Dong B, Yu S, Cheng L, Hu S, Liu W, Xu L, Bai X, Wang L, Song H. Antibacterial PDT nanoplatform capable of releasing therapeutic gas for synergistic and enhanced treatment against deep infections. Theranostics 2022; 12:2580-2597. [PMID: 35401821 PMCID: PMC8965476 DOI: 10.7150/thno.70277] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/16/2022] [Indexed: 11/05/2022] Open
Abstract
Antibacterial photodynamic therapy (aPDT) has emerged as an attractive treatment option for efficient removal of pathogenic bacteria. However, aPDT in deep tissue will encounter difficulties such as limited light penetration depth, insufficient oxygen (O2) supply and inability to eliminate inflammation introduced by bacteria, which hinders its clinical application. Herein, the near infrared (NIR) strategy of simultaneously generating O2 and CO was developed for aPDT based antibacterial therapy and mitigation of deep infection inflammation. Methods: We prepared NIR-mediated multifunctional aPDT nanoplatform (POS-UCNPs/ICG) producing therapeutic gas of O2 and CO. The CO, O2 and ROS generation of the nanoplatform were characterized by dye probes, respectively. The antibacterial activity and anti-inflammation of POS-UCNPs/ICG were demonstrated in vitro and in vivo. In addition, the therapeutic effects in vivo were serially analyzed by immunofluorescence staining, Masson's staining, hematoxylin and eosin staining, colony formation units (CFU) and so on. Results: NIR-mediated multifunctional aPDT nanoplatform was realized by combining the up-conversion nanoparticles (UCNPs) and partially oxidized SnS2 (POS) nanosheets (NSs) as well as indocyanine green (ICG). Using a single 808 nm light, aPDT can be achieved via ICG molecules, meanwhile, O2/CO can be generated by POS NSs through upconversion light excitation. During the aPDT process, O2 can enhance aPDT, while CO can regulate inflammation through the PI3K/NF-κB pathway. Therefore, POS-UCNPs/ICG groups had a highest percentage of healing area up to 91.55±1.26% in mouse abscess model. Conclusion: Due to enhanced aPDT and anti-inflammatory collaborative therapy, the POS-UCNPs/ICG composites showed remarkably accelerated recovery in animal abscess models. Such NIR light responsive nanoplatform with optimized antibacterial capacity and immunomodulatory functions is promising for clinical therapeutics of bacteria-induced infections.
Collapse
Affiliation(s)
- Bingshuai Zhou
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Xiaolin Sun
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Siyao Yu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Liang Cheng
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Songtao Hu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Wei Liu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Lin Xu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Xue Bai
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Lin Wang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Hongwei Song
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| |
Collapse
|
10
|
Ríos-Arrabal S, Puentes-Pardo JD, Moreno-SanJuan S, Szuba Á, Casado J, García-Costela M, Escudero-Feliu J, Verbeni M, Cano C, González-Puga C, Martín-Lagos Maldonado A, Carazo Á, León J. Endothelin-1 as a Mediator of Heme Oxygenase-1-Induced Stemness in Colorectal Cancer: Influence of p53. J Pers Med 2021; 11:jpm11060509. [PMID: 34199777 PMCID: PMC8227293 DOI: 10.3390/jpm11060509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an antioxidant protein implicated in tumor progression, metastasis, and resistance to therapy. Elevated HO-1 expression is associated with stemness in several types of cancer, although this aspect has not yet been studied in colorectal cancer (CRC). Using an in vitro model, we demonstrated that HO-1 overexpression regulates stemness and resistance to 5-FU treatment, regardless of p53. In samples from CRC patients, HO-1 and endothelin converting enzyme-1 (ECE-1) expression correlated significantly, and p53 had no influence on this result. Carbon monoxide (CO) activated the ECE-1/endothelin-1 (ET-1) pathway, which could account for the protumoral effects of HO-1 in p53 wild-type cells, as demonstrated after treatment with bosentan (an antagonist of both ETRA and ETRB endothelin-1 receptors). Surprisingly, in cells with a non-active p53 or a mutated p53 with gain-of-function, ECE-1-produced ET-1 acted as a protective molecule, since treatment with bosentan led to increased efficiency for spheres formation and percentage of cancer stem cells (CSCs) markers. In these cells, HO-1 could activate or inactivate certain unknown routes that could induce these contrary responses after treatment with bosentan in our cell model. However more research is warranted to confirm these results. Patients carrying tumors with a high expression of both HO-1 and ECE-1 and a non-wild-type p53 should be considered for HO-1 based-therapies instead of ET-1 antagonists-based ones.
Collapse
Affiliation(s)
- Sandra Ríos-Arrabal
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Jose D. Puentes-Pardo
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Departamento de Farmacología, Facultad de Farmacia, Universidad de Granada, 18071 Granada, Spain
| | - Sara Moreno-SanJuan
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Cytometry and Microscopy Research Service, Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
| | - Ágata Szuba
- Unidad de Gestión Clínica de Cirugía, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Jorge Casado
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - María García-Costela
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Julia Escudero-Feliu
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Michela Verbeni
- Departamento de Ciencias de la Computación e Inteligencia Artificial, E.T.S. de Ingenierías Informática y de Telecomunicación, Universidad de Granada, 18014 Granada, Spain; (M.V.); (C.C.)
| | - Carlos Cano
- Departamento de Ciencias de la Computación e Inteligencia Artificial, E.T.S. de Ingenierías Informática y de Telecomunicación, Universidad de Granada, 18014 Granada, Spain; (M.V.); (C.C.)
| | - Cristina González-Puga
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Unidad de Gestión Clínica de Cirugía, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| | - Alicia Martín-Lagos Maldonado
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| | - Ángel Carazo
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Josefa León
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
- Correspondence: ; Tel.: +34-958023199
| |
Collapse
|
11
|
Di Pietro C, Öz HH, Murray TS, Bruscia EM. Targeting the Heme Oxygenase 1/Carbon Monoxide Pathway to Resolve Lung Hyper-Inflammation and Restore a Regulated Immune Response in Cystic Fibrosis. Front Pharmacol 2020; 11:1059. [PMID: 32760278 PMCID: PMC7372134 DOI: 10.3389/fphar.2020.01059] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022] Open
Abstract
In individuals with cystic fibrosis (CF), lung hyper-inflammation starts early in life and is perpetuated by mucus obstruction and persistent bacterial infections. The continuous tissue damage and scarring caused by non-resolving inflammation leads to bronchiectasis and, ultimately, respiratory failure. Macrophages (MΦs) are key regulators of immune response and host defense. We and others have shown that, in CF, MΦs are hyper-inflammatory and exhibit reduced bactericidal activity. Thus, MΦs contribute to the inability of CF lung tissues to control the inflammatory response or restore tissue homeostasis. The non-resolving hyper-inflammation in CF lungs is attributed to an impairment of several signaling pathways associated with resolution of the inflammatory response, including the heme oxygenase-1/carbon monoxide (HO-1/CO) pathway. HO-1 is an enzyme that degrades heme groups, leading to the production of potent antioxidant, anti-inflammatory, and bactericidal mediators, such as biliverdin, bilirubin, and CO. This pathway is fundamental to re-establishing cellular homeostasis in response to various insults, such as oxidative stress and infection. Monocytes/MΦs rely on abundant induction of the HO-1/CO pathway for a controlled immune response and for potent bactericidal activity. Here, we discuss studies showing that blunted HO-1 activation in CF-affected cells contributes to hyper-inflammation and defective host defense against bacteria. We dissect potential cellular mechanisms that may lead to decreased HO-1 induction in CF cells. We review literature suggesting that induction of HO-1 may be beneficial for the treatment of CF lung disease. Finally, we discuss recent studies highlighting how endogenous HO-1 can be induced by administration of controlled doses of CO to reduce lung hyper-inflammation, oxidative stress, bacterial infection, and dysfunctional ion transport, which are all hallmarks of CF lung disease.
Collapse
Affiliation(s)
| | | | | | - Emanuela M. Bruscia
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
12
|
Sarkar A, Fouzder C, Chakraborty S, Ahmmed E, Kundu R, Dam S, Chattopadhyay P, Dhara K. A Nuclear-Localized Naphthalimide-Based Fluorescent Light-Up Probe for Selective Detection of Carbon Monoxide in Living Cells. Chem Res Toxicol 2020; 33:651-656. [PMID: 31944672 DOI: 10.1021/acs.chemrestox.9b00462] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A nuclear-localized fluorescent light-up probe, NucFP-NO2, was designed and synthesized that can detect CO selectively in an aqueous buffer (pH 7.4, 37 °C) through the CO-mediated transformation of the nitro group into an amino-functionalized moiety. This probe triggered a more than 55-fold "turn-on" fluorescence response to CO without using any metal ions, e.g., Pd, Rh, Fe, etc. The enhanced response is highly selective over a variety of relevant reactive oxygen, nitrogen, and sulfur species and also various biologically important cationic, anionic, and neutral species. The detection limit of this probe for CO is as low as 0.18 μM with a linear range of 0-70 μM. Also, this fluorogenic probe is an efficient candidate for monitoring intracellular CO in living cells (RAW 264.7, A549 cells), and the fluorescence signals predominantly localize in the nuclear region.
Collapse
Affiliation(s)
- Arnab Sarkar
- Department of Chemistry , The University of Burdwan , Golapbag, Burdwan 713104 , West Bengal , India
| | - Chandrani Fouzder
- Cell Signaling Laboratory, Department of Zoology, Siksha-Bhavana , Visva-Bharati University , Santiniketan 731235 , India
| | - Sujaya Chakraborty
- Department of Chemistry , The University of Burdwan , Golapbag, Burdwan 713104 , West Bengal , India
| | - Ejaj Ahmmed
- Department of Chemistry , The University of Burdwan , Golapbag, Burdwan 713104 , West Bengal , India
| | - Rakesh Kundu
- Cell Signaling Laboratory, Department of Zoology, Siksha-Bhavana , Visva-Bharati University , Santiniketan 731235 , India
| | - Somasri Dam
- Department of Microbiology , University of Burdwan , Burdwan 713104 , West Bengal , India
| | - Pabitra Chattopadhyay
- Department of Chemistry , The University of Burdwan , Golapbag, Burdwan 713104 , West Bengal , India
| | - Koushik Dhara
- Department of Chemistry , Sambhu Nath College , Labpur, Birbhum 731303 , West Bengal , India
| |
Collapse
|
13
|
Zimmer TS, Ciriminna G, Arena A, Anink JJ, Korotkov A, Jansen FE, van Hecke W, Spliet WG, van Rijen PC, Baayen JC, Idema S, Rensing NR, Wong M, Mills JD, van Vliet EA, Aronica E. Chronic activation of anti-oxidant pathways and iron accumulation in epileptogenic malformations. Neuropathol Appl Neurobiol 2020; 46:546-563. [PMID: 31869431 PMCID: PMC7308211 DOI: 10.1111/nan.12596] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/18/2019] [Indexed: 12/29/2022]
Abstract
Aims Oxidative stress is evident in resected epileptogenic brain tissue of patients with developmental brain malformations related to mammalian target of rapamycin activation: tuberous sclerosis complex (TSC) and focal cortical dysplasia type IIb (FCD IIb). Whether chronic activation of anti‐oxidant pathways is beneficial or contributes to pathology is not clear. Methods We investigated oxidative stress markers, including haem oxygenase 1, ferritin and the inflammation associated microRNA‐155 in surgically resected epileptogenic brain tissue of TSC (n = 10) and FCD IIb (n = 8) patients and in a TSC model (Tsc1GFAP−/− mice) using immunohistochemistry, in situ hybridization, real‐time quantitative PCR and immunoblotting. Using human foetal astrocytes we performed an in vitro characterization of the anti‐oxidant response to acute and chronic oxidative stress and evaluated overexpression of the disease‐relevant pro‐inflammatory microRNA‐155. Results Resected TSC or FCD IIb tissue displayed higher expression of oxidative stress markers and microRNA‐155. Tsc1GFAP−/− mice expressed more microRNA‐155 and haem oxygenase 1 in the brain compared to wild‐type, preceding the typical development of spontaneous seizures in these animals. In vitro, chronic microRNA‐155 overexpression induced haem oxygenase 1, iron regulatory elements and increased susceptibility to oxidative stress. Overexpression of iron regulatory genes was also detected in patients with TSC, FCD IIb and Tsc1GFAP−/− mice. Conclusion Our results demonstrate that early and sustained activation of anti‐oxidant signalling and dysregulation of iron metabolism are a pathological hallmark of FCD IIb and TSC. Our findings suggest novel therapeutic strategies aimed at controlling the pathological link between both processes.
Collapse
Affiliation(s)
- T S Zimmer
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - G Ciriminna
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - A Arena
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - J J Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - A Korotkov
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - F E Jansen
- Department of Paediatric Neurology, University Medical Center Utrecht, The Netherlands
| | - W van Hecke
- Department of Pathology, University Medical Center Utrecht, The Netherlands
| | - W G Spliet
- Department of Pathology, University Medical Center Utrecht, The Netherlands
| | - P C van Rijen
- Department of Neurosurgery, Brain Centre, Rudolf Magnus Institute for Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J C Baayen
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - S Idema
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - N R Rensing
- Department of Neurology, Washington University, Saint Louis, MO, USA
| | - M Wong
- Department of Neurology, Washington University, Saint Louis, MO, USA
| | - J D Mills
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - E A van Vliet
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - E Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
14
|
Fürstenau CR, de Souza ICC, de Oliveira MR. The effects of kahweol, a diterpene present in coffee, on the mitochondria of the human neuroblastoma SH-SY5Y cells exposed to hydrogen peroxide. Toxicol In Vitro 2019; 61:104601. [DOI: 10.1016/j.tiv.2019.104601] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/07/2019] [Accepted: 07/11/2019] [Indexed: 10/26/2022]
|
15
|
Zhang Z, Zhang L, Wang B, Zhu X, Zhao L, Chu C, Guo Q, Wei R, Yin X, Zhang Y, Li X. RNF144B inhibits LPS-induced inflammatory responses via binding TBK1. J Leukoc Biol 2019; 106:1303-1311. [PMID: 31509299 PMCID: PMC6899866 DOI: 10.1002/jlb.2a0819-055r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/19/2019] [Accepted: 08/30/2019] [Indexed: 01/02/2023] Open
Abstract
Innate immune responses need to be precisely controlled to avoid prolonged inflammation and prevent unwanted damage to the host. Here, we report that RNF144B responded dynamically to LPS stimulation and negatively regulated LPS‐induced inflammation. We found that RNF144B interacted with the scaffold/dimerization domain (SDD) of TANK binding kinase 1 (TBK1) through the in between RING (IBR) domain to inhibit its phosphorylation and K63‐linked polyubiquitination, which led to TBK1 inactivation, IRF3 dephosphorylation, and IFN‐β reduction. RNF144B knockdown with siRNA increased IRF3 activation and IFN‐β production in response to LPS stimulation. Our study identifies that RNF144B interaction with TBK1 is sufficient to inactivate TBK1 and delineates a previously unrecognized role for RNF144B in innate immune responses.
Collapse
Affiliation(s)
- Zhen Zhang
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Luoyan Zhang
- Key Lab of Plant Stress Research, College of Life Science, Shandong Normal University, Jinan, Shandong, China
| | - Bin Wang
- Department of Peripheral Vascular Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaoxiao Zhu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lin Zhao
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chu Chu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qiang Guo
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ran Wei
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xunqiang Yin
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yunhong Zhang
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xia Li
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
16
|
Wang QH, Li W, Jiang YX, Lu XH, Wang GG. The extract from Agkistrodon halys venom protects against lipopolysaccharide (LPS)-induced myocardial injury. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:176. [PMID: 31315617 PMCID: PMC6637617 DOI: 10.1186/s12906-019-2595-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/09/2019] [Indexed: 01/20/2023]
Abstract
BACKGROUND Snake venoms contain various bioactive constituents which possess potential therapeutic effects. The aim of this work was to investigate the effect of the extract from Agkistrodon halys venom on lipopolysaccharide (LPS)-induced myocardial injury. METHODS Thirty male Sprague-Dawley rats were randomly assigned to three groups (10 rats per group): control group, LPS group and LPS + extract group. Rats in control and the LPS groups were intravenously injected with sterile saline solution, and rats in the LPS + extract group with the extract. After 2 h, rats of the control group were intraperitoneally injected sterile saline solution, and rats in the LPS and the LPS + extract groups were treated with LPS (20 mg per kg body weight). Levels of creatine kinase (CK) and lactate dehydrogenase (LDH) in serum were determined. Anti-inflammation of the extract was analyzed via determination of TNF-α and IL-6 in serum, and expression of TNF-α, IL-6, COX-2 and p-ERK protein in hearts. Heme oxygenase-1 (HO-1) and p-NF-κB protein expression in hearts, superoxide dismutase (SOD) activity and malondialdehyde (MDA) level in serum were used to evaluate the anti-oxidative properties of the extract. RESULTS Extract pretreatment significantly decreased the level of serum CK and LDH, reduced the generation of inflammatory cytokines such as TNF-α and IL-6, and also reduced serum level of MDA in the LPS + extract group compared with the LPS group. In addition, the extract increased SOD activity in serum, HO-1 protein expression in hearts, and decreased TNF-α, IL-6, COX-2, p-NF-κB and p-ERK1/2 protein expression. CONCLUSION Our results suggested that beneficial effect of this extract might be associated with an improved anti-oxidation and anti-inflammatory effect via downregulation of NF-κB/COX-2 signaling by activating HO-1/CO in hearts.
Collapse
Affiliation(s)
- Quan-Hai Wang
- Department of Histology and Embryology, Wannan Medical College, Wuhu, 241002, People's Republic of China
| | - Wei Li
- Department of Pathophysiology, Wannan Medical College, 22 West Wenchang Road, Yijiang District, Wuhu, 241002, China
| | - Yu-Xin Jiang
- Department of Physiology, Wannan Medical College, Wuhu, 241002, People's Republic of China
| | - Xiao-Hua Lu
- Department of Pathophysiology, Wannan Medical College, 22 West Wenchang Road, Yijiang District, Wuhu, 241002, China
| | - Guo-Guang Wang
- Department of Pathophysiology, Wannan Medical College, 22 West Wenchang Road, Yijiang District, Wuhu, 241002, China.
| |
Collapse
|
17
|
Dong J, Li W, Cheng LM, Wang GG. Lycopene attenuates LPS-induced liver injury by inactivation of NF-κB/COX-2 signaling. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:817-825. [PMID: 31933889 PMCID: PMC6945183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/26/2018] [Indexed: 06/10/2023]
Abstract
AIM This study aimed to investigate the effect of lycopene on LPS-induced liver injury in mice and its mechanisms. METHODS Male C57bl/6 mice were randomly assigned to three groups: sham control group (S-C), LPS control group (L-C), lycopene treatment group (L-T). The mice from the L-T were treated with lycopene for 2 weeks, and the remaining mice with solvent. Afterwards, the mice from the L-C and the L-T received an intraperitoneal injection of LPS (20 mg/kg, dissolved in sterile saline), and the S-C mice were injected with sterile saline. Serum levels of alanine transaminase (ALT) and aspartate aminotransferase (AST) were determined for analysis of liver function. Levels of inflammatory cytokines including tumor necrosis factor (TNF)-α and interleukin (IL)-6, malondialdehyde (MDA) content, and the activity of superoxide dismutase (SOD), were detected in serum. Liver tissues were operated for morphologic analysis and determination of protein by western blot. RESULTS Pretreatment with lycopene significantly decreased levels of ALT, AST, and TNF-α and IL-6, reduced MDA content, and increased activity of SOD in serum compared with the L-C mice. Lycopene increased expression of nuclear factor-erythroid 2 related factor 2 (Nrf2), and reduced expression of cyclooxygenase (COX)-2, and phosphorylation of nuclear factor-kappa B (NF-κB) and extracellular regulated protein kinases 1/2 (ERK1/2). CONCLUSION The results showed that lycopene attenuates LPS-induced liver injury by reducing NF-κB/COX-2 signaling by upregulation of Nrf2/HO-1 activation.
Collapse
Affiliation(s)
- Juan Dong
- Experimental Center for Function Subjects, Wannan Medical CollegeWuhu 241002, Anhui, China
| | - Wei Li
- Department of Pathophysiology, Wannan Medical CollegeWuhu 241002, Anhui, China
| | - Li-Min Cheng
- Experimental Center of Morphology, Wannan Medical CollegeWuhu 241002, Anhui, China
| | - Guo-Guang Wang
- Department of Pathophysiology, Wannan Medical CollegeWuhu 241002, Anhui, China
| |
Collapse
|
18
|
N P, Ss A, Pv M. Comprehensive biology of antipyretic pathways. Cytokine 2019; 116:120-127. [PMID: 30711851 DOI: 10.1016/j.cyto.2019.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 01/10/2023]
Abstract
Pyrogens, the fever inducing substances accidently enter into a human body through contamination from medical or pharmaceutical products may create mild to severe complications including septicaemia and shocking syndromes. To avoid such drastic situations all the pharmaceuticals and medical devices are analysed for presence of pyrogens prior to their release into market. The entry of exogenous pyrogens like bacterial endotoxins induces the release of endogenous pyrogens or inflammatory cytokines that activate immune system to defend against these pathogens. Generation of heat is considered as one of the important defence mechanism of body achieved through receptor mediated interaction of endogenous pyrogens at the thermoregulatory centre of hypothalamus. However, uncontrolled fever and febrile reaction may cause lethal effects to the subject itself. So a well sophistically functioning antipyretic mechanism is necessary to achieve thermoregulation. The coordinated interaction of antipyretic cytokines and other mediators are active in human immune system which play a crucial role in maintaining thermal homeostasis. The multiple interacting antipyretic signals and their mechanism are the major subjects of this review.
Collapse
Affiliation(s)
- Prajitha N
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojapura, Trivandrum 695 012, Kerala, India
| | - Athira Ss
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojapura, Trivandrum 695 012, Kerala, India
| | - Mohanan Pv
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojapura, Trivandrum 695 012, Kerala, India.
| |
Collapse
|
19
|
The inhibition of heme oxigenase-1 (HO-1) abolishes the mitochondrial protection induced by sesamol in LPS-treated RAW 264.7 cells. Chem Biol Interact 2018; 296:171-178. [PMID: 30261164 DOI: 10.1016/j.cbi.2018.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/09/2018] [Accepted: 09/18/2018] [Indexed: 12/28/2022]
Abstract
Redox impairment and mitochondrial dysfunction have been seen in inflammation. Thus, there is interest in studies aiming to find molecules that would exert mitochondrial protection in mammalian tissues undergoing inflammation. Sesamol (SES) is an antioxidant and anti-inflammatory molecule as demonstrated in both in vitro and in vivo experimental models. Nonetheless, it was not previously demonstrated whether and how SES would cause mitochondrial protection during inflammation. Thus, we investigated here whether a pretreatment (for 1 h) with SES (1-100 μM) would prevent mitochondrial impairment in lipopolysaccharide (LPS)-treated RAW 264.7 cells. It was also evaluated whether the heme oxigenase-1 (HO-1) would be involved in the effects on mitochondria induced by SES. We found that SES reduced the levels of lipid peroxidation and protein nitration in the membranes of mitochondria obtained from LPS-treated RAW 264.7 cells. SES also attenuated the production of superoxide anion radical (O2-•) and nitric oxide (NO•) in this experimental model. SES suppressed the LPS-elicited mitochondrial dysfunction, as assessed through the analyses of the activities of the mitochondrial complexes I and V. SES also abrogated the LPS-induced decrease in the levels of adenosine triphosphate (ATP) and in the mitochondrial membrane potential (MMP). SES induced mitochondria-related anti-apoptotic effects in LPS-treated cells. Besides, SES pretreatment abrogated the LPS-triggered inflammation by decreasing the levels of pro-inflammatory proteins. The SES-induced mitochondria-associated protection was blocked by the specific inhibitor of HO-1, ZnPP IX (20 μM). Therefore, SES induced mitochondrial protection in LPS-treated cells by a mechanism involving HO-1.
Collapse
|
20
|
Murakami Y, Kawata A, Fujisawa S. Expression of Cyclooxygenase-2, Nitric Oxide Synthase 2 and Heme Oxygenase-1 mRNA Induced by Bis-Eugenol in RAW264.7 Cells and their Antioxidant Activity Determined Using the Induction Period Method. ACTA ACUST UNITED AC 2018; 31:819-831. [PMID: 28882947 DOI: 10.21873/invivo.11135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIM To clarify the mechanisms responsible for the anti-inflammatory/proinflammatory activities of eugenol-related compounds, we investigated the cytotoxicity and up-regulatory/down-refgulatory effects of the biphenols curcumin, bis-eugenol, magnolol and honokiol, and the monophenols eugenol and isoeugenol, on major regulators of cyclooxygenase-2 (Cox-2), nitric oxide synthase 2 (Nos2) and heme oxygenase-1 (HO-1) mRNA in RAW264.7 cells. MATERIALS AND METHODS mRNA expression was investigated using real-time reverse transcriptase-polymerase chain reaction (RT-PCR), and the theoretical parameters were calculated using the DFT/B3LYP/6-31* method. Also, the antioxidant activity of eugenol-related compounds in combination with 2-mercapto-1-methylimidazole (MMI, as a model for glutathione (GSH)) was investigated using the induction period method for polymerization of methyl methacrylate initiated by benzoyl peroxide (BPO). RESULTS The cytotoxicity of eugenol-related compounds showed a linear relationship with their softness (σ) and electrophilicity (ω). At a concentration of 50 μM, biphenols except for bis-eugenol elicited the expression of mRNA for both Cox-2 and Nos2, but monophenols did not. In contrast, bis-eugenol elicited Cox-2 gene expression, but down-regulated Nos2 gene expression. bis-Eugenol alone induced the expression of HO-1 mRNA, and when combined with MMI it showed a potent antagonistic effect on BPO-induced antioxidant activity. The ability of methoxyphenols to inhibit LPS-stimulated Cox-2 gene expression declined in the order curcumin >> isoeugenol > bis-eugenol >> eugenol, and the rank of ability was related to their ω value. CONCLUSION Most eugenol-related compounds had proinflammatory activity at high concentrations. However, they had also anti-inflammatory activity at lower concentrations. Eugenol-related compounds may exert antioxidant and anti-inflammatory activity in LPS-stimulated RAW264.7 cells possibly by inhibiting the activation of nuclear factor-kappa B (Nf-ĸB), whereas bis-eugenol requires induction of HO-1 expression. bis-Eugenol as well as curcumin, may have anti-inflammatory and anticancer therapeutic applications.
Collapse
Affiliation(s)
- Yukio Murakami
- Division of Oral Diagnosis and General Dentistry, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, Sakado, Japan
| | - Akifumi Kawata
- Division of Oral Diagnosis and General Dentistry, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, Sakado, Japan
| | - Seiichiro Fujisawa
- Division of Oral Diagnosis and General Dentistry, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, Sakado, Japan
| |
Collapse
|
21
|
Mahapatra PS, Jain S, Shrestha S, Senapati S, Puppala SP. Ambient endotoxin in PM 10 and association with inflammatory activity, air pollutants, and meteorology, in Chitwan, Nepal. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 618:1331-1342. [PMID: 29033055 DOI: 10.1016/j.scitotenv.2017.09.249] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 09/23/2017] [Accepted: 09/23/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Endotoxin associated with ambient PM (particulate matter) has been linked to adverse respiratory symptoms, but there have been few studies of ambient endotoxin and its association with co-pollutants and inflammation. OBJECTIVES Our aim was to measure endotoxin associated with ambient PM10 (particulate matter with aerodynamic diameter<10μm) in summer 2016 at four locations in Chitwan, Nepal, and investigate its association with meteorology, co-pollutants, and inflammatory activity. METHODS PM10 concentrations were recorded and filter paper samples were collected using E-samplers; PM1, PM2.5, black carbon (BC), methane (CH4), and carbon monoxide (CO) were also measured. The Limulus amebocyte lysate (LAL) assay was used for endotoxin quantification and the nuclear factor kappa B (NFκB) activation assay to assess inflammatory activity. RESULTS The mean concentration of PM10 at the different locations ranged from 136 to 189μg/m3, and of endotoxin from 0.29 to 0.53EU/m3. Pollutant presence was positively correlated with endotoxin. Apart from relative humidity, meteorological variations had no significant impact on endotoxin concentration. NF-κB activity was negatively correlated with endotoxin concentration. CONCLUSIONS To the best of our knowledge, this study provides the first measurements of ambient endotoxin associated with PM10 in Nepal. Endotoxin and co-pollutants were positively associated indicating a similar source. Endotoxin was negatively correlated with inflammatory activity as a result of a time-limited forest fire event during the sampling period. Studies of co-pollutants suggested that the higher levels of endotoxin related to biomass burning were accompanied by increased levels of anti-inflammatory agents, which suppressed the endotoxin inflammatory effect.
Collapse
Affiliation(s)
- Parth Sarathi Mahapatra
- International Centre for Integrated Mountain Development (ICIMOD), G.P.O. Box 3226, Kathmandu, Nepal.
| | - Sumeet Jain
- Institute of Life Sciences, Bhubaneswar, Odisha, India; Manipal University, Manipal, Karnataka, India
| | - Sujan Shrestha
- International Centre for Integrated Mountain Development (ICIMOD), G.P.O. Box 3226, Kathmandu, Nepal
| | | | - Siva Praveen Puppala
- International Centre for Integrated Mountain Development (ICIMOD), G.P.O. Box 3226, Kathmandu, Nepal.
| |
Collapse
|
22
|
Liu Y, Li F, Zhang L, Wu J, Wang Y, Yu H. Taurine alleviates lipopolysaccharide‑induced liver injury by anti‑inflammation and antioxidants in rats. Mol Med Rep 2017; 16:6512-6517. [PMID: 28901400 PMCID: PMC5865819 DOI: 10.3892/mmr.2017.7414] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 04/20/2017] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to investigate the protective effect of taurine on lipopolysaccharide (LPS)-induced liver injury and its mechanisms. Male rats were randomly divided into three groups: Normal saline, LPS model and taurine treatment. Experimental animals were treated with saline or taurine (dissolved in saline, 200 mg/kg/day) via intravenous injection. After 2 h, saline or LPS (0.5 mg/kg) was administrated via intraperitoneal injection. Markers of liver injury, pro-inflammatory cytokines and superoxide dismutase (SOD) activity were determined in plasma. Liver tissues were removed for morphological analysis and determination by western blot analysis. Taurine significantly reduced the elevation in the levels of LPS-induced aspartate transaminase and alanine transaminase and decreased the concentrations of LPS-induced inflammatory factors including tumor necrosis factor-α and interleukin-6. Taurine also increased the activity of SOD in serum and the expression of heme oxygenase-1 protein in liver tissue. Taurine pretreatment also reduced the elevated expression levels of LPS-induced cyclooxygenase-2, nuclear factor κB and extracellular regulated protein kinase. The results from the present study demonstrated that taurine alleviates LPS-induced liver injury. The beneficial role of taurine may be associated with its reduction of pro-inflammatory response and oxidative stress.
Collapse
Affiliation(s)
- Yueyan Liu
- Department of Physiology, School of Clinical Medicine, West Anhui Health Vocational College, Lu'an, Anhui 237005, P.R. China
| | - Feng Li
- Department of Physiology, School of Clinical Medicine, West Anhui Health Vocational College, Lu'an, Anhui 237005, P.R. China
| | - Li Zhang
- Department of Anatomy, School of Clinical Medicine, West Anhui Health Vocational College, Lu'an, Anhui 237005, P.R. China
| | - Jianfeng Wu
- Department of Physiology, School of Clinical Medicine, West Anhui Health Vocational College, Lu'an, Anhui 237005, P.R. China
| | - Yanmei Wang
- Department of Anatomy, School of Clinical Medicine, West Anhui Health Vocational College, Lu'an, Anhui 237005, P.R. China
| | - Hong Yu
- Department of Physiology, School of Clinical Medicine, West Anhui Health Vocational College, Lu'an, Anhui 237005, P.R. China
| |
Collapse
|
23
|
Espinoza JA, González PA, Kalergis AM. Modulation of Antiviral Immunity by Heme Oxygenase-1. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:487-493. [PMID: 28082120 DOI: 10.1016/j.ajpath.2016.11.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/05/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022]
Abstract
Heme oxygenase-1 (HO-1) is a stress-inducible, anti-inflammatory, and cytoprotective enzyme expressed in most cell types in the organism. Under several stress stimuli, HO-1 expression and activity is up-regulated to catalyze the rate-limiting enzymatic step of heme degradation into carbon monoxide, free iron, and biliverdin. Besides its effects on cell metabolism, HO-1 is also capable of modulating host innate and adaptive immune responses in response to sepsis, transplantation, and autoimmunity, and preventing oxidative damage associated with inflammation. In addition, recent studies have reported that HO-1 can exert a significant antiviral activity against a wide variety of viruses, including HIV, hepatitis C virus, hepatitis B virus, enterovirus 71, influenza virus, respiratory syncytial virus, dengue virus, and Ebola virus, among others. Herein, we address the current understanding of the functional significance of HO-1 against a variety of viruses and its potential as a therapeutic strategy to prevent and control viral infections. Furthermore, we review the most important features of the immunoregulatory functions for this enzyme.
Collapse
Affiliation(s)
- Janyra A Espinoza
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM, Combined Research Unit 1064, Nantes University Hospital Nantes, Institute for Transplantation-Urology-Nephrology, Université de Nantes, Faculty of Medicine, Nantes, France; Department of Endocrinology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
24
|
Trotter LA, Patel D, Dubin S, Guerra C, McCloud V, Lockwood P, Messer R, Wataha JC, Lewis JB. Violet/blue light activates Nrf2 signaling and modulates the inflammatory response of THP-1 monocytes. Photochem Photobiol Sci 2017; 16:883-889. [DOI: 10.1039/c6pp00299d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Several studies suggest that light in the UVA range (320–400 nm) activates signaling pathways that are anti-inflammatory and antioxidative.
Collapse
Affiliation(s)
| | - D. Patel
- Augusta University
- Dept. of Oral Biology
- Augusta
- USA
- University of Kentucky
| | - S. Dubin
- Western University of Health Sciences
- College of Dental Medicine
- Pomona
- USA
| | - C. Guerra
- Western University of Health Sciences
- College of Dental Medicine
- Pomona
- USA
| | - V. McCloud
- Augusta University
- Dept. of Oral Biology
- Augusta
- USA
| | - P. Lockwood
- Augusta University
- Dept. of Oral Biology
- Augusta
- USA
| | - R. Messer
- Augusta University
- Dept. of Oral Biology
- Augusta
- USA
| | - J. C. Wataha
- Augusta University
- Dept. of Oral Biology
- Augusta
- USA
- University of Washington
| | - J. B. Lewis
- Augusta University
- Dept. of Oral Biology
- Augusta
- USA
- Western University of Health Sciences
| |
Collapse
|
25
|
Carbon Monoxide Inhibits Porcine Reproductive and Respiratory Syndrome Virus Replication by the Cyclic GMP/Protein Kinase G and NF-κB Signaling Pathway. J Virol 2016; 91:JVI.01866-16. [PMID: 27795439 DOI: 10.1128/jvi.01866-16] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) causes significant economic losses to the pork industry worldwide each year. Our previous research demonstrated that heme oxygenase-1 (HO-1) can suppress PRRSV replication via an unknown molecular mechanism. In this study, inhibition of PRRSV replication was demonstrated to be mediated by carbon monoxide (CO), a downstream metabolite of HO-1. Using several approaches, we demonstrate that CO significantly inhibited PRRSV replication in both a PRRSV permissive cell line, MARC-145, and the predominant cell type targeted during in vivo PRRSV infection, porcine alveolar macrophages (PAMs). Our results showed that CO inhibited intercellular spread of PRRSV; however, it did not affect PRRSV entry into host cells. Furthermore, CO was found to suppress PRRSV replication via the activation of the cyclic GMP/protein kinase G (cGMP/PKG) signaling pathway. CO significantly inhibits PRRSV-induced NF-κB activation, a required step for PRRSV replication. Moreover, CO significantly reduced PRRSV-induced proinflammatory cytokine mRNA levels. In conclusion, the present study demonstrates that CO exerts its anti-PRRSV effect by activating the cellular cGMP/PKG signaling pathway and by negatively regulating cellular NF-κB signaling. These findings not only provide new insights into the molecular mechanism of HO-1 inhibition of PRRSV replication but also suggest potential new control measures for future PRRSV outbreaks. IMPORTANCE PRRSV causes great economic losses each year to the swine industry worldwide. Carbon monoxide (CO), a metabolite of HO-1, has been shown to have antimicrobial and antiviral activities in infected cells. Our previous research demonstrated that HO-1 can suppress PRRSV replication. Here we show that endogenous CO produced through HO-1 catalysis mediates the antiviral effect of HO-1. CO inhibits PRRSV replication by activating the cellular cGMP/PKG signaling pathway and by negatively regulating cellular NF-κB signaling. These findings not only provide new insights into the molecular mechanism of HO-1 inhibition of PRRSV replication but also suggest potential new control measures for future PRRSV outbreaks.
Collapse
|
26
|
Wang S, Dougherty EJ, Danner RL. PPARγ signaling and emerging opportunities for improved therapeutics. Pharmacol Res 2016; 111:76-85. [PMID: 27268145 DOI: 10.1016/j.phrs.2016.02.028] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 01/23/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated nuclear receptor that regulates glucose and lipid metabolism, endothelial function and inflammation. Rosiglitazone (RGZ) and other thiazolidinedione (TZD) synthetic ligands of PPARγ are insulin sensitizers that have been used for the treatment of type 2 diabetes. However, undesirable side effects including weight gain, fluid retention, bone loss, congestive heart failure, and a possible increased risk of myocardial infarction and bladder cancer, have limited the use of TZDs. Therefore, there is a need to better understand PPARγ signaling and to develop safer and more effective PPARγ-directed therapeutics. In addition to PPARγ itself, many PPARγ ligands including TZDs bind to and activate G protein-coupled receptor 40 (GPR40), also known as free fatty acid receptor 1. GPR40 signaling activates stress kinase pathways that ultimately regulate downstream PPARγ responses. Recent studies in human endothelial cells have demonstrated that RGZ activation of GPR40 is essential to the optimal propagation of PPARγ genomic signaling. RGZ/GPR40/p38 MAPK signaling induces and activates PPARγ co-activator-1α, and recruits E1A binding protein p300 to the promoters of target genes, markedly enhancing PPARγ-dependent transcription. Therefore in endothelium, GPR40 and PPARγ function as an integrated signaling pathway. However, GPR40 can also activate ERK1/2, a proinflammatory kinase that directly phosphorylates and inactivates PPARγ. Thus the role of GPR40 in PPARγ signaling may have important implications for drug development. Ligands that strongly activate PPARγ, but do not bind to or activate GPR40 may be safer than currently approved PPARγ agonists. Alternatively, biased GPR40 agonists might be sought that activate both p38 MAPK and PPARγ, but not ERK1/2, avoiding its harmful effects on PPARγ signaling, insulin resistance and inflammation. Such next generation drugs might be useful in treating not only type 2 diabetes, but also diverse chronic and acute forms of vascular inflammation such as atherosclerosis and septic shock.
Collapse
Affiliation(s)
- Shuibang Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Edward J Dougherty
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Sodhi K, Hilgefort J, Banks G, Gilliam C, Stevens S, Ansinelli HA, Getty M, Abraham NG, Shapiro JI, Khitan Z. Uric Acid-Induced Adipocyte Dysfunction Is Attenuated by HO-1 Upregulation: Potential Role of Antioxidant Therapy to Target Obesity. Stem Cells Int 2015; 2016:8197325. [PMID: 26681956 PMCID: PMC4670680 DOI: 10.1155/2016/8197325] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/11/2015] [Indexed: 01/11/2023] Open
Abstract
Increased uric acid levels have been implicated in the pathogenesis of metabolic syndrome. To examine the mechanisms by which this occurs, we hypothesized that an increase in heme oxygenase 1, a potent antioxidant gene, will decrease uric acid levels and adipocyte dysfunction via suppression of ROS and xanthine oxidase (XO) levels. We examined the effect of uric acid on adipogenesis in human mesenchymal stem cells (MSCs) in the presence and absence of cobalt protoporphyrin (CoPP), an HO-1 inducer, and tin mesoporphyrin (SnMP), an HO activity inhibitor. Uric acid increased adipogenesis by increasing NADPH oxidase expression and elevation in the adipogenesis markers C/EBPα, PPARγ, and Mest, while decreasing small lipid droplets and Wnt10b levels. We treated MSCs with fructose, a fuel source that increases uric acid levels. Our results showed that fructose increased XO expression as compared to the control and concomitant treatment with CoPP significantly decreased XO expression and uric acid levels. These beneficial effects of CoPP were reversed by SnMP, supporting a role for HO activity in mediating these effects. These findings demonstrate that increased levels of HO-1 appear crucial in modulating the phenotype of adipocytes exposed to uric acid and in downregulating XO and NADPH oxidase levels.
Collapse
Affiliation(s)
- Komal Sodhi
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Jordan Hilgefort
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - George Banks
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Chelsea Gilliam
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Sarah Stevens
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Hayden A. Ansinelli
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Morghan Getty
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Nader G. Abraham
- Departments of Pharmacology and Medicine, New York Medical College, Valhalla, NY 10595, USA
| | - Joseph I. Shapiro
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Zeid Khitan
- Departments of Internal Medicine and Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| |
Collapse
|
28
|
Carbon monoxide decreases interleukin-1β levels in the lung through the induction of pyrin. Cell Mol Immunol 2015; 14:349-359. [PMID: 26435068 PMCID: PMC5380940 DOI: 10.1038/cmi.2015.79] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 07/24/2015] [Accepted: 07/24/2015] [Indexed: 12/20/2022] Open
Abstract
Carbon monoxide (CO) can act as an anti-inflammatory effector in mouse models of lung injury and disease, through the downregulation of pro-inflammatory cytokines production, though the underlying mechanisms remain unclear. The nucleotide-binding oligomerization domain-, leucine-rich region-, and pyrin domain-containing-3 (NLRP3) inflammasome is a protein complex that regulates the maturation and secretion of pro-inflammatory cytokines, including interleukin-1β (IL-1β). In this report, we show that the CO-releasing molecule (CORM-2) can stimulate the expression of pyrin, a negative regulator of the NLRP3 inflammasome. CORM-2 increased the transcription of pyrin in the human leukemic cell line (THP-1) in the absence and presence of lipopolysaccharide (LPS). In THP-1 cells, CORM-2 treatment dose-dependently reduced the activation of caspase-1 and the secretion of IL-1β, and increased the levels of IL-10, in response to LPS and adenosine 5′-triphosphate (ATP), an NLRP3 inflammasome activation model. Genetic interference of IL-10 by small interfering RNA (siRNA) reduced the effectiveness of CORM-2 in inhibiting IL-1β production and in inducing pyrin expression. Genetic interference of pyrin by siRNA increased IL-1β production in response to LPS and ATP, and reversed CORM-2-dependent inhibition of caspase-1 activation. CO inhalation (250 ppm) in vivo increased the expression of pyrin and IL-10 in lung and spleen, and decreased the levels of IL-1β induced by LPS. Consistent with the induction of pyrin and IL-10, and the downregulation of lung IL-1β production, CO provided protection in a model of acute lung injury induced by intranasal LPS administration. These results provide a novel mechanism underlying the anti-inflammatory effects of CO, involving the IL-10-dependent upregulation of pyrin expression.
Collapse
|
29
|
Jung SS, Moon JS, Xu JF, Ifedigbo E, Ryter SW, Choi AMK, Nakahira K. Carbon monoxide negatively regulates NLRP3 inflammasome activation in macrophages. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1058-67. [PMID: 25770182 DOI: 10.1152/ajplung.00400.2014] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/08/2015] [Indexed: 01/25/2023] Open
Abstract
Inflammasomes are cytosolic protein complexes that promote the cleavage of caspase-1, which leads to the maturation and secretion of proinflammatory cytokines, including interleukin-1β (IL-1β) and IL-18. Among the known inflammasomes, the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3)-dependent inflammasome is critically involved in the pathogenesis of various acute or chronic inflammatory diseases. Carbon monoxide (CO), a gaseous molecule physiologically produced in cells and tissues during heme catabolism, can act as an anti-inflammatory molecule and a potent negative regulator of Toll-like receptor signaling pathways. To date, the role of CO in inflammasome-mediated immune responses has not been fully investigated. Here, we demonstrated that CO inhibited caspase-1 activation and the secretion of IL-1β and IL-18 in response to lipopolysaccharide (LPS) and ATP treatment in bone marrow-derived macrophages. CO also inhibited IL-18 secretion in response to LPS and nigericin treatment, another NLRP3 inflammasome activation model. In contrast, CO did not suppress IL-18 secretion in response to LPS and poly(dA:dT), an absent in melanoma 2 (AIM2)-mediated inflammasome model. LPS and ATP stimulation induced the formation of complexes between NLRP3 and apoptosis-associated speck-like protein, or NLRP3 and caspase-1. CO treatment inhibited these molecular interactions that were induced by LPS and ATP. Furthermore, CO inhibited mitochondrial ROS generation and the decrease of mitochondrial membrane potential induced by LPS and ATP in macrophages. We also observed that the inhibitory effect of CO on the translocation of mitochondrial DNA into the cytosol was associated with suppression of cytokine secretion. Our results suggest that CO negatively regulates NLRP3 inflammasome activation by preventing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Sung-Soo Jung
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, Department of Internal Medicine, Chungnam National University Medical School Daejeon, Republic of Korea
| | - Jong-Seok Moon
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, New York; and Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, New York
| | - Jin-Fu Xu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Emeka Ifedigbo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stefan W Ryter
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, New York; and Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, New York
| | - Augustine M K Choi
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, New York; and Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, New York
| | - Kiichi Nakahira
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, New York; and Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
30
|
Itoh T, Koketsu M, Yokota N, Touho S, Ando M, Tsukamasa Y. Reduced scytonemin isolated from Nostoc commune suppresses LPS/IFNγ-induced NO production in murine macrophage RAW264 cells by inducing hemeoxygenase-1 expression via the Nrf2/ARE pathway. Food Chem Toxicol 2014; 69:330-8. [DOI: 10.1016/j.fct.2014.04.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/09/2014] [Accepted: 04/10/2014] [Indexed: 11/17/2022]
|
31
|
Lee S, Lee SJ, Coronata AA, Fredenburgh LE, Chung SW, Perrella MA, Nakahira K, Ryter SW, Choi AMK. Carbon monoxide confers protection in sepsis by enhancing beclin 1-dependent autophagy and phagocytosis. Antioxid Redox Signal 2014; 20:432-42. [PMID: 23971531 PMCID: PMC3894711 DOI: 10.1089/ars.2013.5368] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIMS Sepsis, a systemic inflammatory response to infection, represents the leading cause of death in critically ill patients. However, the pathogenesis of sepsis remains incompletely understood. Carbon monoxide (CO), when administered at low physiologic doses, can modulate cell proliferation, apoptosis, and inflammation in pre-clinical tissue injury models, though its mechanism of action in sepsis remains unclear. RESULTS CO (250 ppm) inhalation increased the survival of C57BL/6J mice injured by cecal ligation and puncture (CLP) through the induction of autophagy, the down-regulation of pro-inflammatory cytokines, and by decreasing the levels of bacteria in blood and vital organs, such as the lung and liver. Mice deficient in the autophagic protein, Beclin 1 (Becn1(+/-)) were more susceptible to CLP-induced sepsis, and unresponsive to CO therapy, relative to their corresponding wild-type (Becn1(+/+)) littermate mice. In contrast, mice deficient in autophagic protein microtubule-associated protein-1 light chain 3B (LC3B) (Map1lc3b(-/-)) and their corresponding wild-type (Map1lc3b(+/+)) mice showed no differences in survival or response to CO, during CLP-induced sepsis. CO enhanced bacterial phagocytosis in Becn1(+/+) but not Becn1(+/-) mice in vivo and in corresponding cultured macrophages. CO also enhanced Beclin 1-dependent induction of macrophage protein signaling lymphocyte-activation molecule, a regulator of phagocytosis. INNOVATION Our findings demonstrate a novel protective effect of CO in sepsis, dependent on autophagy protein Beclin 1, in a murine model of CLP-induced polymicrobial sepsis. CONCLUSION CO increases the survival of mice injured by CLP through systemic enhancement of autophagy and phagocytosis. Taken together, we suggest that CO gas may represent a novel therapy for patients with sepsis.
Collapse
Affiliation(s)
- Seonmin Lee
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital , Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lee JH, Jung NH, Lee BH, Kim SH, Jun JH. Suppression of Heme Oxygenase-1 by Prostaglandin E2-Protein Kinase A-A-Kinase Anchoring Protein Signaling Is Central for Augmented Cyclooxygenase-2 Expression in Lipopolysaccharide-Stimulated RAW 264.7 Macrophages. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2013; 5:329-36. [PMID: 24003391 PMCID: PMC3756181 DOI: 10.4168/aair.2013.5.5.329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/13/2012] [Accepted: 12/26/2012] [Indexed: 12/21/2022]
Abstract
PURPOSE Prostaglandin (PG) E2 is an immunomodulatory lipid mediator generated mainly via the cyclooxygenase-2 (COX-2) pathway from arachidonic acid at sites of infection and inflammation. A positive feedback loop of PGE2 on COX-2 expression is critical for homeostasis during toll-like receptor (TLR)-mediated inflammatory processes. The mechanism of PGE2-regulated COX-2 expression remains poorly understood. The low-molecular-weight stress protein heme oxygenase-1 (HO-1) contributes to the anti-inflammatory, anti-oxidant and anti-apoptotic response against environmental stress. METHODS We explored the involvement of HO-1 on PGE2 regulation of LPS-induced COX-2 expression in RAW 264.7 macrophages. RESULTS LPS-induced COX-2 expression in RAW 264.7 macrophages was enhanced by exogenous PGE2 or cyclic AMP (cAMP) analogue and was suppressed by a COX inhibitor (indomethacin), a protein kinase A (PKA) inhibitor (KT5720), and A kinase anchoring protein (AKAP) disruptors (Ht31 and RIAD). This result suggests that the stimulatory effects of endogenous and exogenous PGE2 on COX-2 expression are mediated by a cAMP-PKA-AKAP-dependent pathway. The induction of HO-1 was observed in LPS-stimulated RAW 264.7 macrophages. This induction was suppressed by exogenous PGE2 and enhanced by blockage of the endogenous PGE2 effect by the PKA inhibitor or AKAP disruptors. In addition, HO-1 induction by the HO activator copper protoporphyrin suppressed LPS-induced COX-2 expression, which was restored by the addition of exogenous PGE2. The induction of HO-1 inhibited LPS-induced NF-κB p-65 nuclear expression and translocation. CONCLUSIONS AKAP plays an important role in PGE2 regulation of COX-2 expression, and the suppression of HO-1 by PGE2-cAMP-PKA-AKAP signaling helps potentiate the LPS-induced COX-2 expression through a positive feedback loop in RAW 264.7 macrophages.
Collapse
Affiliation(s)
- Jae-Hyoung Lee
- Department of Internal Medicine, Eulji Hospital, Eulji University School of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
33
|
MP4CO, a pegylated hemoglobin saturated with carbon monoxide, is a modulator of HO-1, inflammation, and vaso-occlusion in transgenic sickle mice. Blood 2013; 122:2757-64. [PMID: 23908468 DOI: 10.1182/blood-2013-02-486282] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Transgenic sickle mice expressing β(S) hemoglobin have activated vascular endothelium in multiple organs that exhibits enhanced expression of NF-ĸB and adhesion molecules and promotes microvascular stasis in sickle, but not normal, mice in response to hypoxia/reoxygenation (H/R), or heme. Induction of heme oxygenase-1 (HO-1) or administration of its products, carbon monoxide (CO) or biliverdin, inhibits microvascular stasis in sickle mice. Infusion of human hemoglobin conjugated with polyethylene glycol and saturated with CO (MP4CO) markedly induced hepatic HO-1 activity and inhibited NF-ĸB activation and H/R-induced microvascular stasis in sickle mice. These effects were mediated by CO; saline or MP4 saturated with O2 (MP4OX) had little to no effect on H/R-induced stasis, though unmodified oxyhemoglobin exacerbated stasis. The HO-1 inhibitor, tin protoporphyrin, blocked MP4CO protection, consistent with HO-1 involvement in the protection afforded by MP4CO. MP4CO also induced nuclear factor-erythroid 2 p45-related factor 2 (Nrf2), an important transcriptional regulator of HO-1 and other antioxidant genes. In a heterozygous (hemoglobin-AS) sickle mouse model, intravenous hemin induced cardiovascular collapse and mortality within 120 minutes, which was significantly reduced by MP4CO, but not MP4OX. These data demonstrate that MP4CO induces cytoprotective Nrf2 and HO-1 and decreases NF-ĸB activation, microvascular stasis, and mortality in transgenic sickle mouse models.
Collapse
|
34
|
Tange S, Zhou Y, Nagakui-Noguchi Y, Imai T, Nakanishi A. Initiation of human astrovirus type 1 infection was blocked by inhibitors of phosphoinositide 3-kinase. Virol J 2013; 10:153. [PMID: 23680019 PMCID: PMC3750554 DOI: 10.1186/1743-422x-10-153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 04/23/2013] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Upon initial contact with a virus, host cells activate a series of cellular signaling cascades that facilitate viral entry and viral propagation within the cell. Little is known about how the human astrovirus (HAstV) exploits signaling cascades to establish an infection in host cells. Recent studies showed that activation of extracellular signal-regulated kinase 1/2 (ERK1/2) is important for HAstV infection, though the involvement of other signaling cascades remains unclear. METHODS A panel of kinase blockers was used to search for cellular signaling pathways important for HAstV1 infection. To determine their impact on the infectious process, we examined viral gene expression, RNA replication, and viral RNA and capsid protein release from host cells. RESULTS Inhibitors of phosphoinositide 3-kinase (PI3K) activation interfered with the infection, independent of their effect on ERK 1/2 activation. Activation of the PI3K signaling cascade occurred at an early phase of the infection, judging from the timeframe of Akt phosphorylation. PI3K inhibition at early times, but not at later times, blocked viral gene expression. However, inhibiting the downstream targets of PI3K activation, Akt and Rac1, did not block infection. Inhibition of protein kinase A (PKA) activation was found to block a later phase of HAstV1 production. CONCLUSIONS Our results reveal a previously unknown, essential role of PI3K in the life cycle of HAstV1. PI3K participates in the early stage of infection, possibly during the viral entry process. Our results also reveal the role of PKA in viral production.
Collapse
Affiliation(s)
- Shoichiro Tange
- Section of Gene Therapy, Department of Aging Intervention, National Center for Geriatrics and Gerontology, 35, Gengo, Morioka, Obu, Aichi 474-8522, Japan
| | | | | | | | | |
Collapse
|
35
|
Van Phan T, Sul OJ, Ke K, Lee MH, Kim WK, Cho YS, Kim HJ, Kim SY, Chung HT, Choi HS. Carbon monoxide protects against ovariectomy-induced bone loss by inhibiting osteoclastogenesis. Biochem Pharmacol 2013; 85:1145-52. [DOI: 10.1016/j.bcp.2013.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 12/15/2022]
|
36
|
Increased heme-oxygenase 1 expression in mesenchymal stem cell-derived adipocytes decreases differentiation and lipid accumulation via upregulation of the canonical Wnt signaling cascade. Stem Cell Res Ther 2013; 4:28. [PMID: 23497794 PMCID: PMC3706794 DOI: 10.1186/scrt176] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 01/21/2013] [Indexed: 12/12/2022] Open
Abstract
Introduction Heme oxygenase (HO), a major cytoprotective enzyme, attenuates oxidative stress and obesity. The canonical Wnt signaling cascade plays a pivotal role in the regulation of adipogenesis. The present study examined the interplay between HO-1and the Wnt canonical pathway in the modulation of adipogenesis in mesenchymal stem cell (MSC)-derived adipocytes. Methods To verify the role of HO-1 in generating small healthy adipocytes, cobalt protoporphyrin (CoPP), inducer of HO-1, was used during adipocyte differentiation. Lipid accumulation was measured by Oil red O staining and lipid droplet size was measured by BODIPY staining. Results During adipogenesis in vitro, differentiating pre-adipocytes display transient increases in the expression of genes involved in canonical Wnt signaling cascade. Increased levels of HO-1 expression and HO activity resulted in elevated levels of β-catenin, pGSK3β, Wnt10b, Pref-1, and shh along with increased levels of adiponectin (P < 0.05). In addition, induction of HO-1 resulted in a reduction in C/EBPα, PPARγ, Peg-1/Mest, aP2, CD36 expression and lipid accumulation (P < 0.05). Suppression of HO-1 gene by siRNA decreased Wnt10b, pGSK3β and β-catenin expression, and increased lipid accumulation. The canonical Wnt responsive genes, IL-8 and SFRP1, were upregulated by CoPP and their expression was decreased by the concurrent administration of tin mesoporphyrin (SnMP), an inhibitor of HO activity. Furthermore, knockdown of Wnt10b gene expression by using siRNA showed increased lipid accumulation, and this effect was not decreased by concurrent treatment with CoPP. Also our results show that blocking the Wnt 10b antagonist, Dickkopf 1 (Dkk-1), by siRNA decreased lipid accumulation and this effect was further enhanced by concurrent administration of CoPP. Conclusions This is the first study to demonstrate that HO-1 acts upstream of canonical Wnt signaling cascade and decreases lipogenesis and adipocyte differentiation suggesting that the HO-1 mediated increase in Wnt10b can modulate the adipocyte phenotype by regulating the transcriptional factors that play a role in adipogenesis. This is evidenced by a decrease in lipid accumulation and inflammatory cytokine levels, increased adiponectin levels and elevation of the expression of genes of the canonical Wnt signaling cascade.
Collapse
|
37
|
Zhou J, Ludlow LE, Hasang W, Rogerson SJ, Jaworowski A. Opsonization of malaria-infected erythrocytes activates the inflammasome and enhances inflammatory cytokine secretion by human macrophages. Malar J 2012; 11:343. [PMID: 23046548 PMCID: PMC3528456 DOI: 10.1186/1475-2875-11-343] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 10/05/2012] [Indexed: 12/19/2022] Open
Abstract
Background Antibody opsonization of Plasmodium falciparum-infected erythrocytes (IE) plays a crucial role in anti-malarial immunity by promoting clearance of blood-stage infection by monocytes and macrophages. The effects of phagocytosis of opsonized IE on macrophage pro-inflammatory cytokine responses are poorly understood. Methods Phagocytic clearance, cytokine response and intracellular signalling were measured using IFN-γ-primed human monocyte-derived macrophages (MDM) incubated with opsonized and unopsonized trophozoite-stage CS2 IE, a chondroitin sulphate-binding malaria strain. Cytokine secretion was measured by bead array or ELISA, mRNA using quantitative PCR, and activation of NF-κB by Western blot and electrophoretic mobility shift assay. Data were analysed using the Mann–Whitney U test or the Wilcoxon signed rank test as appropriate. Results Unopsonized CS2 IE were not phagocytosed whereas IE opsonized with pooled patient immune serum (PPS) were (Phagocytic index (PI)=18.4, [SE 0.38] n=3). Unopsonized and opsonized IE induced expression of TNF, IL-1β and IL-6 mRNA by MDM and activated NF-κB to a similar extent. Unopsonized IE induced secretion of IL-6 (median= 622 pg/ml [IQR=1,250-240], n=9) but no IL-1β or TNF, whereas PPS-opsonized IE induced secretion of IL-1β (18.6 pg/mL [34.2-14.4]) and TNF (113 pg/ml [421–17.0]) and increased IL-6 secretion (2,195 pg/ml [4,658-1,095]). Opsonized, but not unopsonized, CS2 IE activated caspase-1 cleavage and enzymatic activity in MDM showing that Fc receptor-mediated phagocytosis activates the inflammasome. MDM attached to IgG-coated surfaces however secreted IL-1β in response to unopsonized IE, suggesting that internalization of IE is not absolutely required to activate the inflammasome and stimulate IL-1β secretion. Conclusions It is concluded that IL-6 secretion from MDM in response to CS2 IE does not require phagocytosis, whereas secretion of TNF and IL-1β is dependent on Fcγ receptor-mediated phagocytosis; for IL-1β, this occurs by activation of the inflammasome. The data presented in this paper show that generating antibody responses to blood-stage malaria parasites is potentially beneficial both in reducing parasitaemia via Fcγ receptor-dependent macrophage phagocytosis and in generating a robust pro-inflammatory response.
Collapse
Affiliation(s)
- Jingling Zhou
- Centre for Virology, Burnet Institute, PO Box 2284, Melbourne, Victoria, 3001, Australia
| | | | | | | | | |
Collapse
|
38
|
Wen Z, Liu Y, Li F, Wen T. Low dose of carbon monoxide intraperitoneal injection provides potent protection against GalN/LPS-induced acute liver injury in mice. J Appl Toxicol 2012; 33:1424-32. [PMID: 23015538 DOI: 10.1002/jat.2806] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 06/24/2012] [Accepted: 06/27/2012] [Indexed: 12/20/2022]
Abstract
Carbon monoxide (CO) is an important effector-signaling molecule involved in various pathophysiological processes. Here we investigated the protective effects of exogenous CO in a murine model of acute liver damage induced by d-galactosamine (GalN) and lipopolysaccharide (LPS). Exogenous CO gas was administered to mice via intraperitoneal injection (first at a dose of 15 ml kg(-1) and then, 6 h later, 8 ml kg(-1)), which caused a significant elevation of blood carboxyhemoglobin levels of up to 12-14% for more than 12 h. GalN/LPS were given to induce acute liver damage in mice 30 min prior to CO exposure. This showed that GalN/LPS induced severe liver injury in mice, whereas CO injection remarkably improved the survival rate of mice and led to attenuated hepatocellular damage. CO exhibited anti-oxidative capabilities by inhibiting hepatic malondialdehyde contents and restoring superoxide dismutase and glutathione, as well as by reducing inducible NOS/NO production. The anti-apoptotic and anti-inflammatory effects of CO were substantial, characterized by a notable inhibition of hepatocyte apoptosis and a reduction of pro-inflammatory cytokines in mice. Our findings thus supported the hypothesis that exogenous CO provides protective effects against acute liver damage in mice, mainly dependent on its anti-oxidative, anti-inflammatory and anti-apoptotic properties.
Collapse
Affiliation(s)
- Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People's Republic of China; Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | | | | | | |
Collapse
|
39
|
Murray TS, Okegbe C, Gao Y, Kazmierczak BI, Motterlini R, Dietrich LEP, Bruscia EM. The carbon monoxide releasing molecule CORM-2 attenuates Pseudomonas aeruginosa biofilm formation. PLoS One 2012; 7:e35499. [PMID: 22563385 PMCID: PMC3338523 DOI: 10.1371/journal.pone.0035499] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/19/2012] [Indexed: 12/16/2022] Open
Abstract
Chronic infections resulting from biofilm formation are difficult to eradicate with current antimicrobial agents and consequently new therapies are needed. This work demonstrates that the carbon monoxide-releasing molecule CORM-2, previously shown to kill planktonic bacteria, also attenuates surface-associated growth of the Gram-negative pathogen Pseudomonas aeruginosa by both preventing biofilm maturation and killing bacteria within the established biofilm. CORM-2 treatment has an additive effect when combined with tobramycin, a drug commonly used to treat P. aeruginosa lung infections. CORM-2 inhibited biofilm formation and planktonic growth of the majority of clinical P. aeruginosa isolates tested, for both mucoid and non-mucoid strains. While CORM-2 treatment increased the production of reactive oxygen species by P. aeruginosa biofilms, this increase did not correlate with bacterial death. These data demonstrate that CO-RMs possess potential novel therapeutic properties against a subset of P. aeruginosa biofilm related infections.
Collapse
Affiliation(s)
- Thomas S. Murray
- Departments of Pediatrics, Yale University School of Medicine New Haven, Connecticut, United States of America
- Laboratory Medicine, Yale University School of Medicine New Haven, Connecticut, United States of America
| | - Chinweike Okegbe
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - Yuan Gao
- Laboratory Medicine, Yale University School of Medicine New Haven, Connecticut, United States of America
| | - Barbara I. Kazmierczak
- Internal Medicine, Yale University School of Medicine New Haven, Connecticut, United States of America
| | | | - Lars E. P. Dietrich
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - Emanuela M. Bruscia
- Departments of Pediatrics, Yale University School of Medicine New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
40
|
TLR4 Activity Is Required in the Resolution of Pulmonary Inflammation and Fibrosis after Acute and Chronic Lung Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:275-92. [DOI: 10.1016/j.ajpath.2011.09.019] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 09/26/2011] [Accepted: 09/30/2011] [Indexed: 01/22/2023]
|
41
|
Vanella L, Kim DH, Sodhi K, Barbagallo I, Burgess AP, Falck JR, Schwartzman ML, Abraham NG. Crosstalk between EET and HO-1 downregulates Bach1 and adipogenic marker expression in mesenchymal stem cell derived adipocytes. Prostaglandins Other Lipid Mediat 2011; 96:54-62. [PMID: 21821145 DOI: 10.1016/j.prostaglandins.2011.07.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/14/2011] [Accepted: 07/19/2011] [Indexed: 01/10/2023]
Abstract
Epoxygenase activity and synthesis of epoxyeicosatrienoic acids (EETs) have emerged as important modulators of obesity and diabetes. We examined the effect of the EET-agonist 12-(3-hexylureido)dodec-8(2) enoic acid on mesenchymal stem cell (MSC) derived adipocytes proliferation and differentiation. MSCs expressed substantial levels of EETs and inhibition of soluble epoxide hydrolase (sEH) increased the level of EETs and decreased adipogenesis. EET agonist treatment increased HO-1 expression by inhibiting a negative regulator of HO-1 expression, Bach-1. EET treatment also increased βcatenin and pACC levels while decreasing PPARγ C/EBPα and fatty acid synthase levels. These changes were manifested by a decrease in the number of large inflammatory adipocytes, TNFα, IFNγ and IL-1α, but an increase in small adipocytes and in adiponectin levels. In summary, EET agonist treatment inhibits adipogenesis and decreases the levels of inflammatory cytokines suggesting the potential action of EETs as intracellular lipid signaling modulators of adipogenesis and adiponectin.
Collapse
Affiliation(s)
- Luca Vanella
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Protective effects of hemin in an experimental model of ventilator-induced lung injury. Eur J Pharmacol 2011; 661:102-8. [DOI: 10.1016/j.ejphar.2011.04.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 04/06/2011] [Accepted: 04/14/2011] [Indexed: 02/05/2023]
|
43
|
Piantadosi CA, Withers CM, Bartz RR, MacGarvey NC, Fu P, Sweeney TE, Welty-Wolf KE, Suliman HB. Heme oxygenase-1 couples activation of mitochondrial biogenesis to anti-inflammatory cytokine expression. J Biol Chem 2011; 286:16374-85. [PMID: 21454555 PMCID: PMC3091243 DOI: 10.1074/jbc.m110.207738] [Citation(s) in RCA: 207] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 03/16/2011] [Indexed: 12/20/2022] Open
Abstract
The induction of heme oxygenase-1 (HO-1; Hmox1) by inflammation, for instance in sepsis, is associated both with an anti-inflammatory response and with mitochondrial biogenesis. Here, we tested the idea that HO-1, acting through the Nfe2l2 (Nrf2) transcription factor, links anti-inflammatory cytokine expression to activation of mitochondrial biogenesis. HO-1 induction after LPS stimulated anti-inflammatory IL-10 and IL-1 receptor antagonist (IL-1Ra) expression in mouse liver, human HepG2 cells, and mouse J774.1 macrophages but blunted tumor necrosis factor-α expression. This was accompanied by nuclear Nfe2l2 accumulation and led us to identify abundant Nfe2l2 and other mitochondrial biogenesis transcription factor binding sites in the promoter regions of IL10 and IL1Ra compared with pro-inflammatory genes regulated by NF-κΒ. Mechanistically, HO-1, through its CO product, enabled these transcription factors to bind the core IL10 and IL1Ra promoters, which for IL10 included Nfe2l2, nuclear respiratory factor (NRF)-2 (Gabpa), and MEF2, and for IL1Ra, included NRF-1 and MEF2. In cells, Hmox1 or Nfe2l2 RNA silencing prevented IL-10 and IL-1Ra up-regulation, and HO-1 induction failed post-LPS in Nfe2l2-silenced cells and post-sepsis in Nfe2l2(-/-) mice. Nfe2l2(-/-) mice compared with WT mice, showed more liver damage, higher mortality, and ineffective CO rescue in sepsis. Nfe2l2(-/-) mice in sepsis also generated higher hepatic TNF-α mRNA levels, lower NRF-1 and PGC-1α mRNA levels, and no enhancement of anti-inflammatory Il10, Socs3, or bcl-x(L) gene expression. These findings disclose a highly structured transcriptional network that couples mitochondrial biogenesis to counter-inflammation with major implications for immune suppression in sepsis.
Collapse
Affiliation(s)
- Claude A Piantadosi
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Wang Y, Zhang MX, Meng X, Liu FQ, Yu GS, Zhang C, Sun T, Wang XP, Li L, Wang YY, Ding SF, Yang JM, Zhang Y. Atorvastatin suppresses LPS-induced rapid upregulation of Toll-like receptor 4 and its signaling pathway in endothelial cells. Am J Physiol Heart Circ Physiol 2011; 300:H1743-52. [DOI: 10.1152/ajpheart.01335.2008] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In the present study, we tested our hypothesis that atorvastatin exerts its anti-inflammation effect via suppressing LPS-induced rapid upregulation of Toll-like receptor 4 (TLR4) mRNA and its downstream p38, ERK, and NF-κB signaling pathways in human umbilical-vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs). TLR4 mRNA expression and its downstream kinase activities induced by LPS alone or atorvastatin + LPS in endothelial cells were quantified using quantitative real-time PCR and enzyme-linked immunosorbent assay. Preincubation of LPS-stimulated endothelial cells with TLR4 siRNA was conducted to identify the target of the anti-inflammatory effects of atorvastatin. Atorvastatin incubation resulted in the reduction of LPS-induced TLR4 mRNA expression, ERK1/2 and P38 MAPK phosphorylation, and NF-κB binding activity. Pretreatment with MEK/ERK1/2 inhibitor PD98059 attenuated atorvastatin + LPS-induced NF-κB activity but had no effect on P38 MAPK phosphorylation. In contrast, pretreatment with P38 MAPK inhibitor SB203580 resulted in upregulation of atorvastatin + LPS-induced ERK1/2 phosphorylation but had no significant effects on NF-κB activity. On the other hand, blocking NF-κB with SN50 produced no effects on atorvastatin + LPS-induced ERK1/2 and P38 MAPK phosphorylation. Moreover, TLR4 gene silencing produced the same effects as the atorvastatin treatment. In conclusion, atorvastatin downregulated TLR4 mRNA expression by two distinct signaling pathways. First, atorvastatin stabilized Iκ-Bα, which directly inhibited NF-κB activation. Second, atorvastatin inactivated ERK phosphorylation, which indirectly inhibited NF-κB activation. Suppression of p38 MAPK by atorvastatin upregulates ERK but exerts no effect on NF-κB.
Collapse
Affiliation(s)
- Ying Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Ming Xiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Xiao Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Fu Qiang Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Guang Sheng Yu
- Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Tao Sun
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Xu Ping Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Li Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Yuan Yuan Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Shi Fang Ding
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Jian Min Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital
| |
Collapse
|
45
|
Jeong SI, Lee YE, Jang SI. (2S)-2′-methoxykurarinone from Sophora flavescens suppresses cutaneous T cell-attracting chemokine/CCL27 expression induced by interleukin-ß/tumor necrosis factor-α via heme oxygenase-1 in human keratinocytes. J Med Food 2011; 13:1116-24. [PMID: 20883179 DOI: 10.1089/jmf.2009.1333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
One of the CC chemokines, cutaneous T cell-attracting chemokine (CTACK/CCL27), is a skin-specific CC chemokine that is produced constitutively by keratinocytes and is highly up-regulated in inflammatory skin conditions such as atopic dermatitis and contact dermatitis. (2S)-2′-Methoxykurarinone (MOK) from Sophora flavescens has been demonstrated to have antioxidant effects. Heme oxygenase (HO)-1 has recently emerged as an important cytoprotective enzyme against oxidative stress and inflammatory responses in many cell types. This study aimed to define whether and how MOK regulates skin specific CTACK/CCL27 chemokine production in human HaCaT keratinocytes. The level of CTACK/CCL27 and HO-1 expression was measured by reverse transcription-polymerase chain reaction, and signaling was evaluated by western blot analysis. CTACK/CCL27 production was determined by enzyme-linked immunosorbent assay. Pretreatment with MOK suppressed tumor necrosis factor-α (TNF-α)- and interleukin (IL)-1ß-induced CTACK/CCL27 production in human HaCaT keratinocytes. MOK inhibited TNF-α- and IL-1ß-induced nuclear factor κB (NF-κB) activation. Interestingly, pretreatment with MOK significantly suppressed TNF-α- and IL-1ß-induced CTACK/CCL27 production through the induction of HO-1. This suppression was completely abolished by HO-1 small interfering RNA. Furthermore, carbon monoxide, but not other end products of HO-1 activity, also suppressed TNF-α- and IL-1ß-induced CTACK/CCL27 production. These results demonstrate that MOK attenuates TNF-α- and IL-1ß-induced production of CTACK/CCL27 in human HaCaT keratinocytes by inhibiting NF-κB activation and induction of HO-1.
Collapse
Affiliation(s)
- Seung-Il Jeong
- Jeonju Biomaterials Institute, College of Alternative Medicine, Jeonju University, Jeonju, Republic of Korea
| | | | | |
Collapse
|
46
|
Hasegawa U, van der Vlies AJ, Simeoni E, Wandrey C, Hubbell JA. Carbon monoxide-releasing micelles for immunotherapy. J Am Chem Soc 2010; 132:18273-80. [PMID: 21128648 DOI: 10.1021/ja1075025] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
With the discovery of important biological roles of carbon monoxide (CO), the use of this gas as a therapeutic agent has attracted attention. However, the medical application of this gas has been hampered by the complexity of the administration method. To overcome this problem, several transition-metal carbonyl complexes, such as Ru(CO)(3)Cl(glycinate), [Ru(CO)(3)Cl(2)](2), and Fe(η(4)-2-pyrone)(CO)(3), have been used as CO-releasing molecules both in vitro and in vivo. We sought to develop micellar forms of metal carbonyl complexes that would display slowed diffusion in tissues and thus better ability to target distal tissue drainage sites. Specifically, we aimed to develop a new CO-delivery system using a polymeric micelle having a Ru(CO)(3)Cl(amino acidate) structure as a CO-releasing segment. The CO-releasing micelles were prepared from triblock copolymers composed of a hydrophilic poly(ethylene glycol) block, a poly(ornithine acrylamide) block bearing Ru(CO)(3)Cl(ornithinate) moieties, and a hydrophobic poly(n-butylacrylamide) block. The polymers formed spherical micelles in the range of 30-40 nm in hydrodynamic diameter. Further characterization revealed the high CO-loading capacity of the micelles. CO-release studies showed that the micelles were stable in physiological buffer and serum and released CO in response to thiol-containing compounds such as cysteine. The CO release of the micelles was slower than that of Ru(CO)(3)Cl(glycinate). In addition, the CO-releasing micelles efficiently attenuated the lipopolysaccharide-induced NF-κB activation of human monocytes, while Ru(CO)(3)Cl(glycinate) did not show any beneficial effects. Moreover, cell viability assays revealed that the micelles significantly reduced the cytotoxicity of the Ru(CO)(3)Cl(amino acidate) moiety. This novel CO-delivery system based on CO-releasing micelles may be useful for therapeutic applications of CO.
Collapse
Affiliation(s)
- Urara Hasegawa
- Institute of Bioengineering (IBI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | | | | | | | | |
Collapse
|
47
|
Shih RH, Yang CM. Induction of heme oxygenase-1 attenuates lipopolysaccharide-induced cyclooxygenase-2 expression in mouse brain endothelial cells. J Neuroinflammation 2010; 7:86. [PMID: 21118574 PMCID: PMC3002338 DOI: 10.1186/1742-2094-7-86] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 11/30/2010] [Indexed: 12/18/2022] Open
Abstract
Background Prostaglandin E2 (PGE2), an arachidonic acid metabolite converted by cyclooxygenase-2 (COX-2), plays important roles in the regulation of endothelial functions in response to bacterial infection. The enzymatic activity of COX-2 can be down-regulated by heme oxygenase-1 (HO-1) induction. However, the mechanisms underlying HO-1 modulating COX-2 protein expression are not known. Objective The aim of the present study was to investigate whether the up-regulation of HO-1 regulates COX-2 expression induced by lipopolysaccharide (LPS), an endotoxin produced by Gram negative bacteria, in mouse brain endothelial cells (bEnd.3) Methods Cultured bEnd.3 cells were used to investigate LPS-induced COX-2 expression and PGE2 production. Cobalt protoporphyrin IX (CoPP, an HO-1 inducer), infection with a recombinant adenovirus carried with HO-1 gene (Adv-HO-1), or zinc protoporphyrin (ZnPP, an HO-1 inhibitor) was used to stimulate HO-1 induction or inhibit HO-1 activity. The expressions of COX-2 and HO-1 were evaluated by western blotting. PGE2 levels were detected by an enzyme-linked immunoassay. Hemoglobin (a chelator of carbon monoxide, CO, one of metabolites of HO-1) and CO-RM2 (a CO releasing molecule) were used to investigate the mechanisms of HO-1 regulating COX-2 expression. Results We found that LPS-induced COX-2 expression and PGE2 production were mediated through NF-κB (p65) via activation of Toll-like receptor 4 (TLR4). LPS-induced COX-2 expression was inhibited by HO-1 induction by pretreatment with CoPP or infection with Adv-HO-1. This inhibitory effect of HO-1 was reversed by pretreatment with either ZnPP or hemoglobin. Pretreatment with CO-RM2 also inhibited TLR4/MyD88 complex formation, NF-κB (p65) activation, COX-2 expression, and PGE2 production induced by LPS. Conclusions We show here a novel inhibition of HO-1 on LPS-induced COX-2/PGE2 production in bEnd.3. Our results reinforce the emerging role of cerebral endothelium-derived HO-1 as a protector against cerebral vascular inflammation triggered by bacterial infection.
Collapse
Affiliation(s)
- Ruey-Horng Shih
- Department of Physiology and Pharmacology, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | | |
Collapse
|