1
|
Antony GS, Ramkumar M, Sujiritha B, Vikash N, Rajaram K, Ganesan P, Ayyadurai N, Kamini NR. Leiotrametes flavida MTCC 12927 laccase: scaleup, purification and its application for dye decolourisation enhanced by water-soluble mediator TEMPOL. 3 Biotech 2025; 15:106. [PMID: 40181804 PMCID: PMC11961788 DOI: 10.1007/s13205-025-04259-9] [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: 09/11/2024] [Accepted: 03/02/2025] [Indexed: 04/05/2025] Open
Abstract
Laccase production from an isolated white rot fungus identified as Leiotrametes flavida MTCC 12927 was optimised in flasks, scaled up in 5 and 10 L working volume fermenters and purified to homogeneity. Purified laccase was tested for its decolourisation potential on 3 dyes: Basic Red 46 (azo class), Acid Blue 9 (triarylmethane class) and Reactive Blue 21 (phthalocyanine class). TEMPOL (4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl), a less expensive water-soluble hydroxy derivative of mediator TEMPO (2,2,6,6-tetramethylpiperidin-1-yl) oxidanyl), was tested as a mediator which had not been reported before to our knowledge along with well-known mediators hydroxybenzotriazole (HBT) and syringaldehyde (SYD). Laccase in the presence of HBT could decolourise all the 3 dyes. Basic Red 46 and Acid Blue 9 were completely decolourised in 4 and 8 h, respectively. Reactive Blue 21 showed a decolourisation of 70.3% compared to 50.3% with TEMPOL and 20% with the enzyme alone. Laccase + TEMPOL combination was significantly better than HBT in decolourising azo dye Basic Red 46 completely within 30 min compared to 4 h for HBT. Furthermore, laccase TEMPOL and HBT combinations were also tested in azo dye-containing tannery effluent. TEMPOL showed a higher decolourisation rate of 36% compared to 22% for the latter. Chemical oxygen demand (COD) analysis of treated effluent samples showed a COD reduction of 23.2% for laccase + TEMPOL treatment and 19.2% for laccase + HBT compared to 7.8% with laccase alone. TEMPOL showed potential as a low-cost, water-soluble mediator for azo dye decolourisation. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04259-9.
Collapse
Affiliation(s)
- George Sebastian Antony
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| | - Mannankatti Ramkumar
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| | - Baskaran Sujiritha
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| | - Negi Vikash
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| | - Krishnasamy Rajaram
- Centre for Analysis, Testing, Evaluation & Reporting Services (CATERS), Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| | - Ponesakki Ganesan
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| | - Nirakulam Ayyadurai
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| | - Numbi Ramudu Kamini
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research–Central Leather Research Institute (CSIR-CLRI), Chennai, Tamil Nadu 600020 India
| |
Collapse
|
2
|
Ale T, Ale T, Baker KJ, Zuniga KM, Hutcheson J, Lavik E. Delivery of Tempol from Polyurethane Nanocapsules to Address Oxidative Stress Post-Injury. Bioconjug Chem 2025; 36:146-151. [PMID: 39921627 DOI: 10.1021/acs.bioconjchem.4c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025]
Abstract
Traumatic brain injuries (TBIs) result in significant morbidity and mortality due to the cascade of secondary injuries involving oxidative stress and neuroinflammation. The development of effective therapeutic strategies to mitigate these effects is critical. This study explores the fabrication and characterization of polyurethane nanocapsules for the sustained delivery of Tempol, a potent antioxidant. The nanocapsules were designed to extend the release of Tempol over a 30-day period, addressing the prolonged oxidative stress observed post-TBI. Tempol-loaded polyurethane nanocapsules were synthesized using interfacial polymerization and nanoemulsion techniques. Two generations of nanocapsules were produced, differing in Tempol loading and PEGylation levels. The first generation, with lower Tempol loading, exhibited an average size of 159.8 ± 12.61 nm and a Z-average diameter of 771.9 ± 87.95 nm. The second generation, with higher Tempol loading, showed an average size of 141.4 ± 6.13 nm and a Z-average diameter of 560.7 ± 171.1 nm. The zeta potentials were -18.9 ± 5.02 mV and -11.9 ± 3.54 mV for the first and second generations, respectively. Both generations demonstrated the presence of urethane linkages, confirmed by Fourier Transform Infrared Spectroscopy (FTIR). Loading studies revealed Tempol concentrations of 61.94 ± 3.04 μg/mg for the first generation and 77.61 ± 3.04 μg/mg for the second generation nanocapsules. Release profiles indicated an initial burst followed by a sustained, nearly linear release over 30 days. The higher PEGylation in the second generation nanocapsules is advantageous for intravenous administration, potentially enhancing their therapeutic efficacy in TBI treatment. This study demonstrates the feasibility of using polyurethane nanocapsules for the prolonged delivery of Tempol, offering a promising approach to manage oxidative stress and improve outcomes in TBI patients. Future work will include testing these nanocapsules in vivo to determine their potential at modulating recovery from TBI.
Collapse
Affiliation(s)
- Temitope Ale
- University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Tolulope Ale
- University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Kimberly J Baker
- Defense Health Agency, United States Air Force 59th Medical Wing, Surgical and Technological Advancements for Traumatic Injury in Combat (STATIC), JBSA-Lackland, Texas 78236, United States
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United States
| | - Kameel M Zuniga
- Defense Health Agency, United States Air Force 59th Medical Wing, Surgical and Technological Advancements for Traumatic Injury in Combat (STATIC), JBSA-Lackland, Texas 78236, United States
| | - Jack Hutcheson
- Defense Health Agency, United States Air Force 59th Medical Wing, Surgical and Technological Advancements for Traumatic Injury in Combat (STATIC), JBSA-Lackland, Texas 78236, United States
| | - Erin Lavik
- University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland 20850, United States
| |
Collapse
|
3
|
Huang S, Xu M, Deng X, Da Q, Li M, Huang H, Zhao L, Jing L, Wang H. Anti irradiation nanoparticles shelter immune organ from radio-damage via preventing the IKK/IκB/NF-κB activation. Mol Cancer 2024; 23:234. [PMID: 39425231 PMCID: PMC11490033 DOI: 10.1186/s12943-024-02142-4] [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: 07/23/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Normal tissue and immune organ protection are critical parts of the tumor radiation therapy process. Radiation-induced immune organ damage (RIOD) causes several side reactions by increasing oxidative stress and inflammatory responses, resulting in unsatisfactory curability in tumor radiation therapy. The aim of this study was to develop a novel and efficient anti irradiation nanoparticle and explore its mechanism of protecting splenic tissue from radiation in mice. METHODS Nanoparticles of triphenylphosphine cation NIT radicals (NPs-TPP-NIT) were prepared and used to protect the spleens of mice irradiated with X-rays. Splenic tissue histopathology and hematological parameters were investigated to evaluate the protective effect of NPs-TPP-NIT against X-ray radiation. Proteomics was used to identify differentially expressed proteins related to inflammatory factor regulation. In addition, in vitro and in vivo experiments were performed to assess the impact of NPs-TPP-NIT on radiation therapy. RESULTS NPs-TPP-NIT increased superoxide dismutase, catalase, and glutathione peroxidase activity and decreased malondialdehyde levels and reactive oxygen species generation in the spleens of mice after exposure to 6.0 Gy X-ray radiation. Moreover, NPs-TPP-NIT inhibited cell apoptosis, blocked the activation of cleaved cysteine aspartic acid-specific protease/proteinase, upregulated the expression of Bcl-2, and downregulated that of Bax. We confirmed that NPs-TPP-NIT prevented the IKK/IκB/NF-κB activation induced by ionizing radiation, thereby alleviating radiation-induced splenic inflammatory damage. In addition, when used during radiotherapy for tumors in mice, NPs-TPP-NIT exhibited no significant toxicity and conferred no significant tumor protective effects. CONCLUSIONS NPs-TPP-NIT prevented activation of IKK/IκB/NF-κB signaling, reduced secretion of pro-inflammatory factors, and promoted production of anti-inflammatory factors in the spleen, which exhibited radiation-induced damage repair capability without diminishing the therapeutic effect of radiation therapy. It suggests that NPs-TPP-NIT serve as a potential radioprotective drug to shelter immune organs from radiation-induced damage.
Collapse
Affiliation(s)
- Shigao Huang
- Department of Radiation Oncology, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Min Xu
- Department of Chemistry, School of Pharmacy, The Air Force Medical University, Xi'an, 710032, China
- The Third Stationed Outpatient Department, General Hospital of Central Theater Command, Wuhan, 430070, China
| | - Xiaojun Deng
- Department of Chemistry, School of Pharmacy, The Air Force Medical University, Xi'an, 710032, China
| | - Qingyue Da
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Miaomiao Li
- Department of Chemistry, School of Pharmacy, The Air Force Medical University, Xi'an, 710032, China
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Hao Huang
- Department of Radiation Oncology, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Linlin Jing
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Haibo Wang
- Department of Chemistry, School of Pharmacy, The Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Crosswhite P, Sun Z. TNFα Induces DNA and Histone Hypomethylation and Pulmonary Artery Smooth Muscle Cell Proliferation Partly via Excessive Superoxide Formation. Antioxidants (Basel) 2024; 13:677. [PMID: 38929115 PMCID: PMC11200563 DOI: 10.3390/antiox13060677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Objective: The level of tumor necrosis factor-α (TNF-α) is upregulated during the development of pulmonary vascular remodeling and pulmonary hypertension. A hallmark of pulmonary arterial (PA) remodeling is the excessive proliferation of PA smooth muscle cells (PASMCs). The purpose of this study is to investigate whether TNF-α induces PASMC proliferation and explore the potential mechanisms. Methods: PASMCs were isolated from 8-week-old male Sprague-Dawley rats and treated with 0, 20, or 200 ng/mL TNF-α for 24 or 48 h. After treatment, cell number, superoxide production, histone acetylation, DNA methylation, and histone methylation were assessed. Results: TNF-α treatment increased NADPH oxidase activity, superoxide production, and cell numbers compared to untreated controls. TNF-α-induced PASMC proliferation was rescued by a superoxide dismutase mimetic tempol. TNF-α treatment did not affect histone acetylation at either dose but did significantly decrease DNA methylation. DNA methyltransferase 1 activity was unchanged by TNF-α treatment. Further investigation using QRT-RT-PCR revealed that GADD45-α, a potential mediator of DNA demethylation, was increased after TNF-α treatment. RNAi inhibition of GADD45-α alone increased DNA methylation. TNF-α impaired the epigenetic mechanism leading to DNA hypomethylation, which can be abolished by a superoxide scavenger tempol. TNF-α treatment also decreased H3-K4 methylation. TNF-α-induced PASMC proliferation may involve the H3-K4 demethylase enzyme, lysine-specific demethylase 1 (LSD1). Conclusions: TNF-α-induced PASMC proliferation may be partly associated with excessive superoxide formation and histone and DNA methylation.
Collapse
Affiliation(s)
- Patrick Crosswhite
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Human Physiology, Gonzaga University, Spokane, WA 99205, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| |
Collapse
|
5
|
Ai L, Li R, Wang X, Liu Z, Li Y. Tempol alleviates acute lung injury by affecting glutathione synthesis through Nrf2 and inhibiting ferroptosis in lung epithelial cells. J Biochem Mol Toxicol 2024; 38:e23674. [PMID: 38454815 DOI: 10.1002/jbt.23674] [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: 04/17/2023] [Revised: 09/12/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
As a life-threatening disease, acute lung injury (ALI) may progress to chronic pulmonary fibrosis. For the treatment of lung injury, Tempol is a superoxide dismutase mimetic and intracellular redox agent that can be a potential drug. This study investigated the regulatory mechanism of Tempol in the treatment of ALI. A mouse model of ALI was established, and HE staining was used to examine histomorphology. The CCK-8 assay was used to measure cell viability, and oxidative stress was assessed by corresponding kits. Flow cytometry and dichlorodihydrofluorescein diacetate staining assays were used to detect reactive oxygen species (ROS) levels. Protein expression levels were measured by Western blot analysis and ELISA. Pulmonary vascular permeability was used to measure the lung wet/dry weight ratio. The level of oxidative stress was increased in ALI mice, and the level of ferroptosis was upregulated. Tempol inhibited this effect and alleviated ALI. The administration of Tempol alleviated the pathological changes in ALI, inhibited pulmonary vascular permeability, and improved lung injury in ALI mice. The upregulation of genes essential for glutathione (GSH) metabolism induced by lipopolysaccharide (LPS) was inhibited by Tempol. In addition, nuclear factor-related factor 2 (Nrf2) is activated by Tempol therapy to regulate the de novo synthesis pathway of GSH, thereby alleviating LPS-induced lung epithelial cell damage. The results showed that Tempol alleviated ALI by activating the Nrf2 pathway to inhibit oxidative stress and ferroptosis in lung epithelial cells. In conclusion, this study demonstrates that Tempol alleviates ALI by inhibiting ferroptosis in lung epithelial cells through the effect of Nrf2 on GSH synthesis.
Collapse
Affiliation(s)
- Li Ai
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ran Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiaona Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhijuan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yongxia Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
6
|
Lim EY, Lee SY, Shin HS, Kim GD. Reactive Oxygen Species and Strategies for Antioxidant Intervention in Acute Respiratory Distress Syndrome. Antioxidants (Basel) 2023; 12:2016. [PMID: 38001869 PMCID: PMC10669909 DOI: 10.3390/antiox12112016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening pulmonary condition characterized by the sudden onset of respiratory failure, pulmonary edema, dysfunction of endothelial and epithelial barriers, and the activation of inflammatory cascades. Despite the increasing number of deaths attributed to ARDS, a comprehensive therapeutic approach for managing patients with ARDS remains elusive. To elucidate the pathological mechanisms underlying ARDS, numerous studies have employed various preclinical models, often utilizing lipopolysaccharide as the ARDS inducer. Accumulating evidence emphasizes the pivotal role of reactive oxygen species (ROS) in the pathophysiology of ARDS. Both preclinical and clinical investigations have asserted the potential of antioxidants in ameliorating ARDS. This review focuses on various sources of ROS, including NADPH oxidase, uncoupled endothelial nitric oxide synthase, cytochrome P450, and xanthine oxidase, and provides a comprehensive overview of their roles in ARDS. Additionally, we discuss the potential of using antioxidants as a strategy for treating ARDS.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
- Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
- Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
| |
Collapse
|
7
|
Lê T, Buscemi L, Lepore M, Mishkovsky M, Hyacinthe JN, Hirt L. Influence of DNP Polarizing Agents on Biochemical Processes: TEMPOL in Transient Ischemic Stroke. ACS Chem Neurosci 2023; 14:3013-3018. [PMID: 37603041 PMCID: PMC10485885 DOI: 10.1021/acschemneuro.3c00137] [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: 02/22/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Hyperpolarization of 13C by dissolution dynamic nuclear polarization (dDNP) boosts the sensitivity of magnetic resonance spectroscopy (MRS), making possible the monitoring in vivo and in real time of the biochemical reactions of exogenously infused 13C-labeled metabolic tracers. The preparation of a hyperpolarized substrate requires the use of free radicals as polarizing agents. Although added at very low doses, these radicals are not biologically inert. Here, we demonstrate that the presence of the nitroxyl radical TEMPOL influences significantly the cerebral metabolic readouts of a hyperpolarized [1-13C] lactate bolus injection in a mouse model of ischemic stroke with reperfusion. Thus, the choice of the polarizing agent in the design of dDNP hyperpolarized MRS experiments is of great importance and should be taken into account to prevent or to consider significant effects that could act as confounding factors.
Collapse
Affiliation(s)
- Thanh
Phong Lê
- Geneva
School of Health Sciences, HES-SO University
of Applied Sciences and Arts Western Switzerland, Avenue de Champel 47, 1206 Geneva, Switzerland
- Laboratory
of Functional and Metabolic Imaging, Institute
of Physics, École Polytechnique Fédérale de Lausanne
(EPFL), Station 6, 1015 Lausanne, Switzerland
| | - Lara Buscemi
- Department
of Clinical Neurosciences, Lausanne University
Hospital (CHUV), Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Mario Lepore
- CIBM
Center for Biomedical Imaging, École
Polytechnique Fédérale de Lausanne (EPFL), Station 6, 1015 Lausanne, Switzerland
| | - Mor Mishkovsky
- Laboratory
of Functional and Metabolic Imaging, Institute
of Physics, École Polytechnique Fédérale de Lausanne
(EPFL), Station 6, 1015 Lausanne, Switzerland
| | - Jean-Noël Hyacinthe
- Geneva
School of Health Sciences, HES-SO University
of Applied Sciences and Arts Western Switzerland, Avenue de Champel 47, 1206 Geneva, Switzerland
- Laboratory
of Functional and Metabolic Imaging, Institute
of Physics, École Polytechnique Fédérale de Lausanne
(EPFL), Station 6, 1015 Lausanne, Switzerland
- Image
Guided Intervention Laboratory, Faculty of Medicine, University of Geneva, HUG, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva 14, Switzerland
| | - Lorenz Hirt
- Department
of Clinical Neurosciences, Lausanne University
Hospital (CHUV), Rue du Bugnon 46, 1011 Lausanne, Switzerland
| |
Collapse
|
8
|
Jayachandra K, Gowda MDM, Rudresha GV, Manjuprasanna VN, Urs AP, Nandana MB, Bharatha M, Jameel NM, Vishwanath BS. Inhibition of sPLA 2 enzyme activity by cell-permeable antioxidant EUK-8 and downregulation of p38, Akt, and p65 signals induced by sPLA 2 in inflammatory mouse paw edema model. J Cell Biochem 2023; 124:294-307. [PMID: 36585945 DOI: 10.1002/jcb.30366] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/24/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023]
Abstract
The arachidonic acid (AA) metabolic pathway, plays a vital role in the production of eicosanoids by the action of pro-inflammatory secretory phospholipase A2 (PLA2 ). Release of eicosanoids is known to be involved in many inflammatory diseases. Identification of the inhibitory molecules of this AA pathway enzyme along with the regulation of intracellular signaling cascades may be a finer choice to develop as a powerful anti-inflammatory drug. In this regard, we have screened few cell-permeable antioxidant molecules Tempo, Mito-TEMPO, N,N'-Bis(salicylideneamino)ethane-manganese(II) (EUK)-134, and EUK-8 against pro-inflammatory sPLA2 s. Among these, we found EUK-8 is a potent inhibitor with its IC50 value ranges 0.7-2.0 µM for sPLA2 s isolated from different sources. Furthermore, docking studies confirm the strong binding of EUK-8 towards sPLA2 . In vivo effect of EUK-8 was studied in HSF-sPLA2 -induced edema in mouse paw model. In addition to neutralizing the edema, EUK-8 significantly reduces the phosphorylation level of inflammatory proteins such as p38 member of MAPK pathway, Akt, and p65 along with the suppression of pro-inflammatory cytokine (interleukin-6) and chemokine (CXCL1) in edematous tissue. This shows that EUK-8 not only inhibits the sPLA2 activity, it also plays an important role in the regulation of sPLA2 -induced cell signaling cascades. Apart from the sPLA2 inhibition, we also examine the regulatory actions of EUK-8 with other downstream enzymes of AA pathway such as 5-LOX assay in human polymorphonuclear leukocytes (PMNs) and COX-2 expression in carrageenan-λ induced paw edema. Here EUK-8 significantly inhibits 5-LOX enzyme activity and downregulates COX-2 expression. These data indicate that EUK-8 found to be a promising multitargeted inhibitory molecule toward inflammatory pathway. In conclusion, mitochondrial targeted antioxidant EUK-8 is not only the powerful antioxidant, also a potent anti-inflammatory molecule and may be a choice of molecule for pharmacological applications.
Collapse
Affiliation(s)
- Krishnegowda Jayachandra
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - M D Milan Gowda
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - Gotravalli V Rudresha
- Evolutionary Venomics Lab, Centre for Ecological Sciences, Indian Institute of Science, Bengaluru, Karnataka, India
| | | | - Amog P Urs
- Comprehensive Cancer Centre, The Ohio State University, Columbus, Ohio, USA
| | | | - Madeva Bharatha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - Noor Mohamed Jameel
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| |
Collapse
|
9
|
Liu B, Peng Y, Yi D, Machireddy N, Dong D, Ramirez K, Dai J, Vanderpool R, Zhu MM, Dai Z, Zhao YY. Endothelial PHD2 deficiency induces nitrative stress via suppression of caveolin-1 in pulmonary hypertension. Eur Respir J 2022; 60:2102643. [PMID: 35798360 PMCID: PMC9791795 DOI: 10.1183/13993003.02643-2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/24/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Nitrative stress is a characteristic feature of the pathology of human pulmonary arterial hypertension. However, the role of nitrative stress in the pathogenesis of obliterative vascular remodelling and severe pulmonary arterial hypertension remains largely unclear. METHOD Our recently identified novel mouse model (Egln1Tie2Cre, Egln1 encoding prolyl hydroxylase 2 (PHD2)) has obliterative vascular remodelling and right heart failure, making it an excellent model to use in this study to examine the role of nitrative stress in obliterative vascular remodelling. RESULTS Nitrative stress was markedly elevated whereas endothelial caveolin-1 (Cav1) expression was suppressed in the lungs of Egln1Tie2Cre mice. Treatment with a superoxide dismutase mimetic, manganese (III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride or endothelial Nos3 knockdown using endothelial cell-targeted nanoparticle delivery of CRISPR-Cas9/guide RNA plasmid DNA inhibited obliterative pulmonary vascular remodelling and attenuated severe pulmonary hypertension in Egln1Tie2Cre mice. Genetic restoration of Cav1 expression in Egln1Tie2Cre mice normalised nitrative stress, reduced pulmonary hypertension and improved right heart function. CONCLUSION These data suggest that suppression of Cav1 expression secondary to PHD2 deficiency augments nitrative stress through endothelial nitric oxide synthase activation, which contributes to obliterative vascular remodelling and severe pulmonary hypertension. Thus, a reactive oxygen/nitrogen species scavenger might have therapeutic potential for the inhibition of obliterative vascular remodelling and severe pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Bin Liu
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Yi Peng
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dan Yi
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Narsa Machireddy
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Daoyin Dong
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karina Ramirez
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Jingbo Dai
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rebecca Vanderpool
- College of Medicine Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Maggie M Zhu
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Zhiyu Dai and You-Yang Zhao contributed equally to this article as lead authors and supervised the work
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Dept of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Dept of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Zhiyu Dai and You-Yang Zhao contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
10
|
Systemic Effects of mitoTEMPO upon Lipopolysaccharide Challenge Are Due to Its Antioxidant Part, While Local Effects in the Lung Are Due to Triphenylphosphonium. Antioxidants (Basel) 2022; 11:antiox11020323. [PMID: 35204206 PMCID: PMC8868379 DOI: 10.3390/antiox11020323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/30/2022] [Accepted: 02/03/2022] [Indexed: 01/08/2023] Open
Abstract
Mitochondria-targeted antioxidants (mtAOX) are a promising treatment strategy against reactive oxygen species-induced damage. Reports about harmful effects of mtAOX lead to the question of whether these could be caused by the carrier molecule triphenylphosphonium (TPP). The aim of this study was to investigate the biological effects of the mtAOX mitoTEMPO, and TPP in a rat model of systemic inflammatory response. The inflammatory response was induced by lipopolysaccharide (LPS) injection. We show that mitoTEMPO reduced expression of inducible nitric oxide synthase in the liver, lowered blood levels of tissue damage markers such as liver damage markers (aspartate aminotransferase and alanine aminotransferase), kidney damage markers (urea and creatinine), and the general organ damage marker, lactate dehydrogenase. In contrast, TPP slightly, but not significantly, increased the LPS-induced effects. Surprisingly, both mitoTEMPO and TPP reduced the wet/dry ratio in the lung after 24 h. In the isolated lung, both substances enhanced the increase in pulmonary arterial pressure induced by LPS observed within 3 h after LPS treatments but did not affect edema formation at this time. Our data suggest that beneficial effects of mitoTEMPO in organs are due to its antioxidant moiety (TEMPO), except for the lung where its effects are mediated by TPP.
Collapse
|
11
|
von Knethen A, Heinicke U, Laux V, Parnham MJ, Steinbicker AU, Zacharowski K. Antioxidants as Therapeutic Agents in Acute Respiratory Distress Syndrome (ARDS) Treatment-From Mice to Men. Biomedicines 2022; 10:98. [PMID: 35052778 PMCID: PMC8773193 DOI: 10.3390/biomedicines10010098] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/26/2021] [Accepted: 12/31/2021] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a major cause of patient mortality in intensive care units (ICUs) worldwide. Considering that no causative treatment but only symptomatic care is available, it is obvious that there is a high unmet medical need for a new therapeutic concept. One reason for a missing etiologic therapy strategy is the multifactorial origin of ARDS, which leads to a large heterogeneity of patients. This review summarizes the various kinds of ARDS onset with a special focus on the role of reactive oxygen species (ROS), which are generally linked to ARDS development and progression. Taking a closer look at the data which already have been established in mouse models, this review finally proposes the translation of these results on successful antioxidant use in a personalized approach to the ICU patient as a potential adjuvant to standard ARDS treatment.
Collapse
Affiliation(s)
- Andreas von Knethen
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Ulrike Heinicke
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Volker Laux
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Andrea U Steinbicker
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|