1
|
Anwar MM, Sah R, Shrestha S, Ozaki A, Roy N, Fathah Z, Rodriguez-Morales AJ. Disengaging the COVID-19 Clutch as a Discerning Eye Over the Inflammatory Circuit During SARS-CoV-2 Infection. Inflammation 2022; 45:1875-1894. [PMID: 35639261 PMCID: PMC9153229 DOI: 10.1007/s10753-022-01674-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the cytokine release syndrome (CRS) and leads to multiorgan dysfunction. Mitochondrial dynamics are fundamental to protect against environmental insults, but they are highly susceptible to viral infections. Defective mitochondria are potential sources of reactive oxygen species (ROS). Infection with SARS-CoV-2 damages mitochondria, alters autophagy, reduces nitric oxide (NO), and increases both nicotinamide adenine dinucleotide phosphate oxidases (NOX) and ROS. Patients with coronavirus disease 2019 (COVID-19) exhibited activated toll-like receptors (TLRs) and the Nucleotide-binding and oligomerization domain (NOD-), leucine-rich repeat (LRR-), pyrin domain-containing protein 3 (NLRP3) inflammasome. The activation of TLRs and NLRP3 by SARS-CoV-2 induces interleukin 6 (IL-6), IL-1β, IL-18, and lactate dehydrogenase (LDH). Herein, we outline the inflammatory circuit of COVID-19 and what occurs behind the scene, the interplay of NOX/ROS and their role in hypoxia and thrombosis, and the important role of ROS scavengers to reduce COVID-19-related inflammation.
Collapse
Affiliation(s)
- Mohammed Moustapha Anwar
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.
| | - Ranjit Sah
- Tribhuvan University Institute of Medicine, Kathmandu, Nepal
| | - Sunil Shrestha
- Department of Pharmaceutical and Health Service Research, Nepal Health Research and Innovation Foundation, Lalitpur, Nepal
| | - Akihiko Ozaki
- Department of Breast Surgery, Jyoban Hospital of Tokiwa Foundation, Iwaki, Japan
- Medical Governance Research Institute, Tokyo, Japan
| | - Namrata Roy
- SRM University, SRM Nagar, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Zareena Fathah
- Kings College London, London, UK
- College of Medicine and Health Sciences, United Arab University, Abu Dhabi, United Arab Emirates
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de Las Americas, Pereira, Risaralda, Colombia.
- Institución Universitaria Visión de Las Americas, Pereira, Risaralda, Colombia.
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru.
- School of Medicine, Universidad Privada Franz Tamayo (UNIFRANZ), Cochabamba, Bolivia.
| |
Collapse
|
2
|
Farella I, Panza R, Capozza M, Laforgia N. Lecithinized superoxide dismutase in the past and in the present: Any role in the actual pandemia of COVID-19? Biomed Pharmacother 2021; 141:111922. [PMID: 34323703 PMCID: PMC8277551 DOI: 10.1016/j.biopha.2021.111922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 01/08/2023] Open
Abstract
The Coronavirus disease 19 (Covid-19) pandemic is devastating the public health: it is urgent to find a viable therapy to reduce the multiorgan damage of the disease. A validated therapeutic protocol is still missing. The most severe forms of the disease are related to an exaggerated inflammatory response. The pivotal role of reactive oxygen species (ROS) in the amplification of inflammation makes the antioxidants a potential therapy, but clinical trials are needed. The lecitinized superoxide dismutase (PC-SOD) could represent a possibility because of bioaviability, safety, and its modulatory effect on the innate immune response in reducing the harmful consequences of oxidative stress. In this review we summarize the evidence on lecitinized superoxide dismutase in animal and human studies, to highlight the rationale for using the PC-SOD to treat COVID-19.
Collapse
Affiliation(s)
- Ilaria Farella
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and 6 Human Oncology, "Aldo Moro" University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy.
| | - Raffaella Panza
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and 6 Human Oncology, "Aldo Moro" University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy.
| | - Manuela Capozza
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and 6 Human Oncology, "Aldo Moro" University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Nicola Laforgia
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and 6 Human Oncology, "Aldo Moro" University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| |
Collapse
|
3
|
Vaneev AN, Kost OA, Eremeev NL, Beznos OV, Alova AV, Gorelkin PV, Erofeev AS, Chesnokova NB, Kabanov AV, Klyachko NL. Superoxide Dismutase 1 Nanoparticles (Nano-SOD1) as a Potential Drug for the Treatment of Inflammatory Eye Diseases. Biomedicines 2021; 9:396. [PMID: 33917028 PMCID: PMC8067682 DOI: 10.3390/biomedicines9040396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory eye diseases remain the most common clinical problem in ophthalmology. The secondary processes associated with inflammation, such as overproduction of reactive oxygen species (ROS) and exhaustion of the endogenous antioxidant system, frequently lead to tissue degeneration, vision blurring, and even blindness. Antioxidant enzymes, such as copper-zinc superoxide dismutase (SOD1), could serve as potent scavengers of ROS. However, their delivery into the eye compartments represents a major challenge due to the limited ocular penetration. This work presents a new therapeutic modality specifically formulated for the eye on the basis of multilayer polyion complex nanoparticles of SOD1 (Nano-SOD1), which is characterized by appropriate storage stability and pronounced therapeutic effect without side reactions such as eye irritation; acute, chronic, and reproductive toxicity; allergenicity; immunogenicity; mutagenicity even at high doses. The ability of Nano-SOD1 to reduce inflammatory processes in the eye was examined in vivo in rabbits with a model immunogenic uveitis-the inflammation of the inner vascular tract of the eye. It was shown during preclinical studies that topical instillations of Nano-SOD1 were much more effective compared to the free enzyme in decreasing uveitis manifestations. In particular, we noted statistically significant differences in such inflammatory signs in the eye as corneal and conjunctival edema, iris hyperemia, and fibrin clots. Moreover, Nano-SOD1 penetrates into interior eye structures more effectively than SOD itself and retains enzyme activity in the eye for a much longer period of time, decreasing inflammation and restoring antioxidant activity in the eye. Thus, the presented Nano-SOD1 can be considered as a potentially useful therapeutic agent for the treatment of ocular inflammatory disorders.
Collapse
Affiliation(s)
- Alexander N. Vaneev
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.N.V.); (O.A.K.); (N.L.E.); (A.S.E.); (A.V.K.)
- Research Laboratory of Biophysics, National University of Science and Technology “MISIS”, 119991 Moscow, Russia;
| | - Olga A. Kost
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.N.V.); (O.A.K.); (N.L.E.); (A.S.E.); (A.V.K.)
| | - Nikolay L. Eremeev
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.N.V.); (O.A.K.); (N.L.E.); (A.S.E.); (A.V.K.)
| | - Olga V. Beznos
- Helmholtz National Medical Research Center of Eye Diseases, 105062 Moscow, Russia; (O.V.B.); (N.B.C.)
| | - Anna V. Alova
- School of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Peter V. Gorelkin
- Research Laboratory of Biophysics, National University of Science and Technology “MISIS”, 119991 Moscow, Russia;
| | - Alexander S. Erofeev
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.N.V.); (O.A.K.); (N.L.E.); (A.S.E.); (A.V.K.)
- Research Laboratory of Biophysics, National University of Science and Technology “MISIS”, 119991 Moscow, Russia;
| | - Natalia B. Chesnokova
- Helmholtz National Medical Research Center of Eye Diseases, 105062 Moscow, Russia; (O.V.B.); (N.B.C.)
| | - Alexander V. Kabanov
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.N.V.); (O.A.K.); (N.L.E.); (A.S.E.); (A.V.K.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalia L. Klyachko
- School of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.N.V.); (O.A.K.); (N.L.E.); (A.S.E.); (A.V.K.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Research Institute “Nanotechnology and Nanomaterials”, G.R. Derzhavin Tambov State University, 392000 Tambov, Russia
| |
Collapse
|
4
|
Binevski PV, Balabushevich NG, Uvarova VI, Vikulina AS, Volodkin D. Bio-friendly encapsulation of superoxide dismutase into vaterite CaCO3 crystals. Enzyme activity, release mechanism, and perspectives for ophthalmology. Colloids Surf B Biointerfaces 2019; 181:437-449. [DOI: 10.1016/j.colsurfb.2019.05.077] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 12/22/2022]
|
5
|
Superoxide Dismutase 1 Nanozyme for Treatment of Eye Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:5194239. [PMID: 26697135 PMCID: PMC4678082 DOI: 10.1155/2016/5194239] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 08/06/2015] [Accepted: 08/12/2015] [Indexed: 01/16/2023]
Abstract
Use of antioxidants to mitigate oxidative stress during ocular inflammatory diseases has shown therapeutic potential. This work examines a nanoscale therapeutic modality for the eye on the base of antioxidant enzyme, superoxide dismutase 1 (SOD1), termed "nanozyme." The nanozyme is produced by electrostatic coupling of the SOD1 with a cationic block copolymer, poly(L-lysine)-poly(ethyleneglycol), followed by covalent cross-linking of the complexes with 3,3'-dithiobis(sulfosuccinimidylpropionate) sodium salt. The ability of SOD1 nanozyme as well as the native SOD1 to reduce inflammatory processes in the eye was examined in vivo in rabbits with immunogenic uveitis. Results suggested that topical instillations of both enzyme forms demonstrated anti-inflammatory activity; however, the nanozyme was much more effective compared to the free enzyme in decreasing uveitis manifestations. In particular, we noted statistically significant differences in such inflammatory signs in the eye as the intensities of corneal and iris edema, hyperemia of conjunctiva, lens opacity, fibrin clots, and the protein content in aqueous humor. Clinical findings were confirmed by histological data. Thus, SOD1-containing nanozyme is potentially useful therapeutic agent for the treatment of ocular inflammatory disorders.
Collapse
|
6
|
Broeyer FJF, Osanto S, Suzuki J, de Jongh F, van Slooten H, Tanis BC, Bruning T, Bax JJ, Ritsema van Eck HJ, de Kam ML, Cohen AF, Mituzhima Y, Burggraaf J. Evaluation of lecithinized human recombinant super oxide dismutase as cardioprotectant in anthracycline-treated breast cancer patients. Br J Clin Pharmacol 2015; 78:950-60. [PMID: 24844787 DOI: 10.1111/bcp.12429] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/15/2014] [Indexed: 12/19/2022] Open
Abstract
AIM Anthracycline-induced cardiotoxicity is (partly) mediated by free radical overload. A randomized study was performed in breast cancer patients to investigate whether free radical scavenger super oxide dismutase (SOD) protects against anthracycline-induced cardiotoxicity as measured by changes in echo, electrocardiography and an array of biomarkers. METHOD AND RESULTS Eighty female, chemotherapy-naïve breast cancer patients (median age 49, range 24-67 years) scheduled for four or five courses of adjuvant 3 weekly doxorubicin plus cyclophosphamide (AC) chemotherapy, were randomly assigned to receive 80 mg PC-SOD (human recombinant SOD bound to lecithin) or placebo, administered intravenously (i.v.) immediately prior to each AC course. The primary end point was protection against cardiac damage evaluated using echocardiography, QT assessments and a set of biochemical markers for myocardial function, oxidative stress and inflammation. Assessments were performed before and during each course of chemotherapy, and at 1, 4 and 9 months after completion of the chemotherapy regimen. In all patients cardiac effects such as increases in NT-proBNP concentration and prolongation of the QTc interval were noticed. There were no differences between the PC-SOD and placebo-treated patients in systolic or diastolic cardiac function or for any other of the biomarkers used to assess the cardiac effects of anthracyclines. CONCLUSION PC-SOD at a dose of 80 mg i.v. is not cardioprotective in patients with breast carcinoma treated with anthracyclines.
Collapse
|
7
|
Lee H, Ko EH, Lai M, Wei N, Balroop J, Kashem Z, Zhang M. Delineating the relationships among the formation of reactive oxygen species, cell membrane instability and innate autoimmunity in intestinal reperfusion injury. Mol Immunol 2013; 58:151-9. [PMID: 24365749 DOI: 10.1016/j.molimm.2013.11.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/15/2013] [Accepted: 11/20/2013] [Indexed: 01/07/2023]
Abstract
Acute intestinal ischemia is a medical emergency with a high mortality rate, attesting to the need for a better understanding of its pathogenesis and the development of effective therapies. The goal of this study was to delineate the relationships among intracellular and extracellular events in intestinal ischemia/reperfusion (I/R) injury, particularly the formation of reactive oxygen species (ROS), cell membrane instability associated with lipid peroxidation and the innate autoimmune response mediated by natural IgM and complement. A murine model of natural IgM-mediated intestinal I/R was used. Mice overexpressing anti-oxidant enzyme SOD1 were found to have significantly reduced intestinal tissue damage and complete blockage of IgM-mediated complement activation compared with WT controls. To determine if cell membrane instability was an event intermediate between ROS formation and natural IgM-mediated innate autoimmune response, the cell membrane stabilizer (trehalose) was administered to WT mice prior to the induction of intestinal ischemia. Treatment with trehalose significantly protected animals from I/R injury and inhibited IgM-mediated complement activation although it did not prevent membrane lipid peroxidation. These data indicate that in normal mice subjected to I/R injury, intracellular ROS formation is an event upstream of the lipid peroxidation which results in cell membrane instability. The membrane instability leads to an innate autoimmune response by natural IgM and complement. Trehalose, a nontoxic disaccharide tolerated well by animals and humans, has promise as a protective agent for patients with medical conditions related to acute intestinal ischemia.
Collapse
Affiliation(s)
- Haekyung Lee
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Eun Hee Ko
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Mark Lai
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States; Department of Biomedical Sciences, Long Island University, Brookville, NY 11548, United States
| | - Na Wei
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States; Department of Biomedical Sciences, Long Island University, Brookville, NY 11548, United States
| | - Javi Balroop
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States; Department of Chemical and Biomolecular Engineering, NYU-Polytechnic Institute, Brooklyn, NY 11201, United States
| | - Zerin Kashem
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Ming Zhang
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States; Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, United States.
| |
Collapse
|
8
|
Charlagorla P, Liu J, Patel M, Rushbrook JI, Zhang M. Loss of plasma membrane integrity, complement response and formation of reactive oxygen species during early myocardial ischemia/reperfusion. Mol Immunol 2013; 56:507-12. [PMID: 23911407 DOI: 10.1016/j.molimm.2013.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 05/01/2013] [Indexed: 11/25/2022]
Abstract
Loss of plasma membrane integrity (LPMI) is a hallmark of necrotic cell death. The involvement of complement and ROS in the development of LPMI during the early stages of murine myocardial ischemia-reperfusion injury was investigated. LPMI developed within 1 h of reperfusion to a level that was sustained through 24 h. C3 deposition became significant at 3-h reperfusion and thus contributed little to LPMI prior to this time. SOD1 transgenic mice had significantly less LPMI compared with WT mice at 1 h of reperfusion but not at later time points. Catalase transgenic mice were not protected from LPMI at 1-h reperfusion compared with WT mice, but had 69% less LPMI at 3-h reperfusion. This protection was transient. At 24-h reperfusion the LPMI of catalase transgenic mice was identical to that of WT mice. The delayed benefits of over-expressed catalase compared with SOD1 are consistent with its antioxidant action downstream of SOD1. The onset of LPMI occurs within 1 h of reperfusion at a level that is maintained through 24 h. ROS contribute significantly to LPMI during the first 3 h of reperfusion, while complement deposition, which becomes significant after 3-h reperfusion, may contribute thereafter.
Collapse
|
9
|
Pioglitazone ameliorates systolic and diastolic cardiac dysfunction in rat model of angiotensin II-induced hypertension. Int J Cardiol 2013; 167:409-15. [DOI: 10.1016/j.ijcard.2012.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 10/16/2011] [Accepted: 01/01/2012] [Indexed: 12/31/2022]
|
10
|
Suzuki J, Broeyer F, Cohen A, Takebe M, Burggraaf J, Mizushima Y. Pharmacokinetics of PC-SOD, a Lecithinized Recombinant Superoxide Dismutase, After Single- and Multiple-Dose Administration to Healthy Japanese and Caucasian Volunteers. J Clin Pharmacol 2013; 48:184-92. [DOI: 10.1177/0091270007309705] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
11
|
Szadujkis-Szadurska K, Grzesk G, Szadujkis-Szadurski L, Gajdus M, Matusiak G. Role of nitric oxide and cGMP in the modulation of vascular contraction induced by angiotensin II and Bay K8644 during ischemia/reperfusion. Exp Ther Med 2012; 5:616-620. [PMID: 23407814 PMCID: PMC3570230 DOI: 10.3892/etm.2012.846] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 11/01/2012] [Indexed: 12/02/2022] Open
Abstract
Vascular smooth muscle tone changes under the influence of numerous contracting and relaxing factors. The purpose of the present study was to determine the modulating effect of ischemia and reperfusion (I/R) on contraction triggered by angiotensin II (ANG II) and Bay K8644 as well as to investigate the importance of nitric oxide (NO) and cGMP in these reactions. Experiments were performed on isolated and perfused Wistar rat tail arteries. The contraction triggered by ANG II and Bay K8644 with the use of intracellular (in calcium-free physiological salt solution; FPSS) and extracellular (in physiological salt solution; PSS) pools of calcium ions after I/R and in the presence of sodium nitroprusside (SNP), 8Br-cGMP, an endothelial NO synthase (NOSe) inhibitor (L-NG-nitroarginine methyl ester; L-NAME) or ODQ [an inhibitor of soluble guanylyl cyclase (GC)] was evaluated. ANG II triggered contraction in FPSS and PSS, but Bay K8644 only in PSS. Ischemia reduced and reperfusion intensified the response of the artery to ANG II, but did not change the action of Bay K8644. SNP and 8Br-cGMP reduced the response of the vessels to ANG II and did not change the modulating effect of ischemia, but reduced the intensifying action of reperfusion on contraction caused by the presence of ANG II. SNP lowered the action of Bay K8644 in PSS. In PSS, L-NAME and ODQ intensified the action of ANG II, eliminating the reducing effect of ischemia on the contraction caused by ANG II, but did not influence the intensifying reaction caused by reperfusion. L-NAME and ODQ did not influence the action of Bay K8644. I/R modulated the contraction of arteries triggered by ANG II, but did not influence the response to Bay K8644. The intra- and extracellular pools of calcium ions mediate the action of ANG II, but Bay K8644 stimulated contraction only with participation of calcium ions flowing into the cell. Control of the vascular smooth muscle tone associated with the action of NO and cGMP is subject to modulation under conditions of I/R.
Collapse
Affiliation(s)
- Katarzyna Szadujkis-Szadurska
- Department of Pharmacology and Therapeutics, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz 85-094, Poland
| | | | | | | | | |
Collapse
|
12
|
Raza SS, Khan MM, Ashafaq M, Ahmad A, Khuwaja G, Khan A, Siddiqui MS, Safhi MM, Islam F. Silymarin protects neurons from oxidative stress associated damages in focal cerebral ischemia: a behavioral, biochemical and immunohistological study in Wistar rats. J Neurol Sci 2011; 309:45-54. [PMID: 21840019 DOI: 10.1016/j.jns.2011.07.035] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 07/15/2011] [Accepted: 07/20/2011] [Indexed: 01/28/2023]
Abstract
Cerebral stroke is the third largest cause of death and the severe leading cause of disability, thus have astronomical financial and social burden worldwide. Accumulated evidence suggests that ROS can be scavenged through utilizing natural antioxidant compounds present in foods and medicinal plants. In this study, we examined whether silymarin, an antioxidant, present in the milk of thistle can prevent or slowdown neuronal injury in focal cerebral ischemia. Male Wistar rats were pre-treated with silymarin (200mg/kg body weight, dissolved in 0.3 % sodium carboxymethyl cellulose, once orally) for 15 days. On day 16, they underwent a transient 2h suture-occlusion of the middle cerebral artery followed by 22 h of reperfusion. Rats were tested for neurobehavioral activity after 22 h reperfusion. Silymarin was found to be successful in upregulating the antioxidant status and lowering the apoptotic responses, and functional recovery returned close to the baseline. This study revealed that silymarin, a naturally occurring flavone from the milk thistle (Silybum marianum), may be helpful in slowing down the progression of neurodegeneration in focal cerebral ischemia. These results suggest that the neuroprotective potential of silymarin is mediated through its anti-oxidative and anti-apoptotic properties.
Collapse
Affiliation(s)
- Syed Shadab Raza
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Szadujkis-Szadurska K, Slupski M, Szadujkis-Szadurski R, Jasinski M, Grześk G, Matusiak G. Modulation of the reaction of vascular smooth muscle cells to angiotensin II induced by catalase and aminotriasol during ischemia-reperfusion. Transplant Proc 2010; 42:1614-7. [PMID: 20620485 DOI: 10.1016/j.transproceed.2010.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Accepted: 04/19/2010] [Indexed: 11/18/2022]
Abstract
BACKGROUND We investigated the influence of catalase and aminotriasol on reactions of the smooth muscle cells induced by angiotensin II (ANG II) after ischemia-reperfusion (I/R). MATERIALS AND METHODS Experiments were performed on perfused male Wistar rat tail arteries. Using classical pharmacometric methods we analyzed the influence of ANG II on vascular contraction, in the presence of catalase and aminotriazole, and after I/R. RESULTS A reduction in maximal response and increased EC(5) value were observed after ischemia, while an increased maximal response and decrease EC(50) value were observed after reperfusion. Catalase decreased and aminotriasol increased maximal responses to ANG II. In the presence of catalase, reduction of the maximal response and increase in EC(50) value were observed after reperfusion. In the presence of aminotriasol, we observed increased maximal response and decreased EC(50) value after I/R. CONCLUSION Ischemia reduced and reperfusion increased the responses of vascular smooth muscle cells to ANG II. Catalase decreased and aminotriasol increased hyperreactivity of arteries to ANG II after reperfusion. These results suggested that antioxidative system modulates reactions induced by ANG II. Reperfusion impairs the balance between antioxidants and the production of reactive oxygen species.
Collapse
|
14
|
Li Y, Zhang J, Li L. Retracted: Comparison of the therapeutic effects of different compositions of muskone in the treatment of experimental myocardial infarct in rats and analgesia in mice. Phytother Res 2010; 22:1219-23. [DOI: 10.1002/ptr.2444] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
15
|
Esposito E, Cuzzocrea S. Role of nitroso radicals as drug targets in circulatory shock. Br J Pharmacol 2009; 157:494-508. [PMID: 19630831 DOI: 10.1111/j.1476-5381.2009.00255.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A vast amount of circumstantial evidence implicates oxygen-derived free radicals (especially, superoxide and hydroxyl radical) and high-energy oxidants [such as peroxynitrite (OONO(-))] as mediators of shock and ischaemia/reperfusion injury. Reactive oxygen species can initiate a wide range of toxic oxidative reactions. These include initiation of lipid peroxidation, direct inhibition of mitochondrial respiratory chain enzymes, inactivation of glyceraldehyde-3 phosphate dehydrogenase, inhibition of membrane sodium/potassium adenosine 5'-triphosphate-ase activity, inactivation of membrane sodium channels and other oxidative modifications of proteins. All these toxicities are likely to play a role in the pathophysiology of shock and ischaemia and reperfusion. Moreover, various studies have clearly shown that treatment with either OONO(-) decomposition catalysts, which selectively inhibit OONO(-), or with superoxide dismutase (SOD) mimetics, which selectively mimic the catalytic activity of the human SOD enzymes, have been shown to prevent in vivo the delayed vascular decompensation and the cellular energetic failure associated with shock and ischaemia/reperfusion injury.
Collapse
|
16
|
Abstract
OBJECTIVE To assess the safety and efficacy of lecithinized superoxide dismutase (PC-SOD) in patients with ulcerative colitis (UC). METHOD PC-SOD was injected once daily at doses of 40 mg (n = 22) and 80 mg (n = 20) for a total treatment period of 4 weeks. Efficacy was assessed by UC-Disease Activity Index (DAI) total score. All side effects were recorded and investigated. RESULTS At 4 weeks, the UC-DAI total score was significantly decreased vs baseline in both the 40 mg and 80 mg groups. It was confirmed that PC-SOD 80 mg was, at least, not significantly superior to PC-SOD 40 mg. Twenty incidences of side effects were noted in 12 (54.55%) of 22 patients in the 40 mg group, while there were three incidences of side effects in two (10.00%) of 20 patients in the 80 mg group. None of these side effects was severe. Thus it was concluded that the test drug is safe when given at daily dosages of 40 mg and 80 mg. CONCLUSION In this pilot study PC-SOD improved UC more rapidly than previously existing drugs. A double blind, placebo-controlled clinical trial of PC-SOD 40 mg/day is required to confirm the efficacy of this agent against UC.
Collapse
Affiliation(s)
- Y Suzuki
- Toho University Sakura Medical CenterSakura, Japan
| | | | - S Okamoto
- Keio University School of MedicineTokyo, Japan
| | - T Hibi
- Keio University School of MedicineTokyo, Japan
| |
Collapse
|
17
|
Wu L, Qiao H, Li Y, Li L. Cardioprotective effects of the combined use of puerarin and Danshensu on acute ischemic myocardial injury in rats. Phytother Res 2007; 21:751-6. [PMID: 17450507 DOI: 10.1002/ptr.2157] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Ischemic heart diseases are the leading cause of death in both developed and developing countries over the past decades. The aim of this study was to investigate the cardioprotective effects of the complex preparation (called Shenge), made of puerarin and Danshensu, on acute ischemic myocardial injury in rats and its underlying mechanisms. The left anterior descending (LAD) coronary artery was occluded to induce myocardial ischemia in hearts of SD rats. Shenge was injected into the tail vein 15 min after occlusion at doses of 0, 30, 60 or 120 mg/kg. Then, the ST elevation was measured at 60, 120 and 240 min after Shenge administration. The infarct size, serum levels of creatine kinase isoenzyme-MB (CK-MB), lactate dehydrogenase (LDH), superoxide dismutase (SOD) and malondialdehyde (MDA), and the ST elevation were measured after the rats were killed. Shenge decreased the ST elevation induced by acute myocardial ischemia, reduced infarct size, serum levels of CK-MB, LDH and MDA and increased the serum activity of SOD in a dose-dependent manner. The combined use of puerarin and Danshensu at a ratio of 1:1 shows the most effective activity. In conclusion, Shenge exerts significant cardioprotective effects against acute ischemic myocardial injury in rats, likely through its antioxidant and antilipid peroxidation properties, and thus may be used as an effective and promising medicine for both prophylaxis and treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Limao Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | | | |
Collapse
|
18
|
Wu L, Qiao H, Li Y, Li L. Protective roles of puerarin and Danshensu on acute ischemic myocardial injury in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2007; 14:652-8. [PMID: 17870452 DOI: 10.1016/j.phymed.2007.07.060] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Ischemic heart diseases have been the leading cause of death in both developed and developing countries over the past decades. The aim of this study was to investigate the cardioprotective effects of the complex preparation (called Shenge), made of puerarin (isolated from Pueraria lobata Ohwi., also called Kudzu) and Danshensu (isolated from the Chinese herb, Salvia miltiorrhiza), on acute ischemic myocardial injury in rats and its underlying mechanisms. The left anterior descending (LAD) coronary artery was occluded to induce myocardial ischemia in the hearts of SD rats. Shenge was injected into the tail vein 15 min after occlusion at doses of 0, 30, 60, or 120 mg/kg body wt. ST elevation was then measured at 60, 120, and 240 min after Shenge administration. The ischemic size, serum levels of creatine kinase isoenzyme-MB (CK-MB), lactate dehydrogenase (LDH), superoxide dismutase (SOD) and malondialdehyde (MDA), and ST elevation were measured after the rats were sacrificed. Shenge decreased ST elevation induced by acute myocardial ischemia, reduced ischemic size, serum levels of CK-MB, LDH and MDA, and increased serum activity of SOD in a dose-dependent manner. The combined use of puerarin and Danshensu at a ratio of 1:1 showed the most effective activity. In conclusion, Shenge exerts significant cardioprotective effects against acute ischemic myocardial injury in rats, likely through its antioxidant and anti-lipid peroxidation properties, and thus may be an effective and promising medicine for both prophylaxis and treatment of ischemic heart disease.
Collapse
Affiliation(s)
- L Wu
- College of Pharmaceutical Sciences, Zhejiang University, Zijingang Campus of Zhejiang University, Hangzhou 310058, China.
| | | | | | | |
Collapse
|
19
|
Broeyer FJF, van Aken BE, Suzuki J, Kemme MJB, Schoemaker HC, Cohen AF, Mizushima Y, Burggraaf J. The pharmacokinetics and effects of a long-acting preparation of superoxide dismutase (PC-SOD) in man. Br J Clin Pharmacol 2007; 65:22-9. [PMID: 17610527 PMCID: PMC2291268 DOI: 10.1111/j.1365-2125.2007.02965.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AIM To study the pharmacokinetics (PK), safety and tolerability of single rising doses up to 80 mg of superoxide dismutase covalently linked to lecithin (PC-SOD) in healthy White volunteers. METHODS This double-blind, placebo-controlled, four-period cross-over study was performed in eight healthy volunteers (four male/four female). Three doses of PC-SOD (20, 40 and 80 mg) and placebo were administered intravenously in randomized order. Serum and urinary PC-SOD concentrations were measured predose and up to 96 h after dosing. In addition to standard safety measurements, the urinary excretion of N-acetyl-beta-glucosaminidase, alpha-glutathione S-transferase (alpha-GST) and pi-GST was measured to evaluate renal function. The PK of PC-SOD was analysed using noncompartmental and compartmental methods. RESULTS All treatments were well tolerated, and no obvious relationship between adverse events and treatment was observed. No effects of PC-SOD on renal function could be detected. Dose normalized C(max) and AUC were not different between the different dosages, indicating linearity of plasma concentrations with dose. Estimated PC-SOD clearance was 2.54 ml min(-1)[95% confidence interval (CI) 2.07, 2.83]. The terminal half-life was estimated to be 1.54 days (95% CI 0.93, 2.15). SOD activity was elevated above baseline for 19 +/- 6 h after the 80-mg dose. CONCLUSIONS Single intravenous administrations of PC-SOD in doses up to 80 mg were well tolerated in healthy White male and female volunteers. With the doses used, SOD activity was linearly related to the dose; after the 80-mg dose it was present for an appreciable period. These findings suggest that it is worthwhile to investigate PC-SOD in clinical conditions characterized by a high radical overload.
Collapse
Affiliation(s)
- F J F Broeyer
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Yamawaki M, Sasaki N, Shimoyama M, Miake J, Ogino K, Igawa O, Tajima F, Shigemasa C, Hisatome I. Protective effect of edaravone against hypoxia-reoxygenation injury in rabbit cardiomyocytes. Br J Pharmacol 2004; 142:618-26. [PMID: 15148265 PMCID: PMC1574956 DOI: 10.1038/sj.bjp.0705775] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
1 We examined whether edaravone (Eda), a clinically available radical scavenger, directly protects cardiomyocytes from ischemia/reperfusion (I/R) injury, and whether the timing of its application is critical for protection. 2 Cardioprotective effects of edaravone were tested in the modified cell-pelleting model of ischemia and under exogenous oxidative stress (hydrogen peroxide: H2O2) in isolated adult rabbit ventricular cells. Cell death and reactive oxygen species (ROS) generation were detected using propidium iodide (PI) and DCFH-DA, respectively. These parameters were evaluated objectively using flow cytometory. 3 Hypoxia and reoxygenation aggravated the proportion of dead cells from 32.2+/-1.8% (Baseline) to 51.3+/-2.7% (Control). When 15 microm edaravone was applied either throughout the entire experiment (Through) or only at reoxygenation (Reox), cell death was significantly reduced to 39.9+/-1.8% (P<0.01 vs Control) and 43.3+/-2.5% (P<0.05 vs Control), respectively. In contrast, when edaravone was applied 10 min after reoxygenation, its protective effect disappeared. Cardioprotection by edaravone was more remarkable than that afforded by other free radical scavengers, such as ascorbate and superoxide dismutase (SOD). There is a positive correlation between the cardioprotective effect of edaravone and the extent of ROS reduction. 4 Edaravone blunted the H2O2-induced changes in electrical properties, and significantly prolonged the time to contracture induced by H2O2 in single ventricular myocytes. 5 Taken together, edaravone directly protects cardiomyocytes from I/R injury by attenuating ROS production, even when applied at the time of reoxygenation, suggesting that edaravone could be a potent cardioprotective therapeutic agent against hypoxia-reoxygenation injury.
Collapse
Affiliation(s)
- Masahiro Yamawaki
- Department of Cardiovascular Medicine, Tottori University Faculty of Medicine, Japan
| | - Norihito Sasaki
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
- Author for correspondence:
| | - Masaki Shimoyama
- Department of Cardiovascular Medicine, Tottori University Faculty of Medicine, Japan
| | - Junichiro Miake
- Department of Cardiovascular Medicine, Tottori University Faculty of Medicine, Japan
| | - Kazuhide Ogino
- Department of Cardiovascular Medicine, Tottori University Faculty of Medicine, Japan
| | - Osamu Igawa
- Department of Cardiovascular Medicine, Tottori University Faculty of Medicine, Japan
| | - Fumito Tajima
- Department of Cardiovascular Medicine, Tottori University Faculty of Medicine, Japan
| | - Chiaki Shigemasa
- Department of Cardiovascular Medicine, Tottori University Faculty of Medicine, Japan
| | - Ichiro Hisatome
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| |
Collapse
|
21
|
den Hartog GJM, Haenen GRMM, Boven E, van der Vijgh WJF, Bast A. Lecithinized copper,zinc-superoxide dismutase as a protector against doxorubicin-induced cardiotoxicity in mice. Toxicol Appl Pharmacol 2004; 194:180-8. [PMID: 14736498 DOI: 10.1016/j.taap.2003.09.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Production of superoxide radicals from doxorubicin is widely accepted to be the cause of the cardiotoxicity induced by this antitumor agent. Pretreatment with superoxide dismutase could improve the therapeutic application. Aim of the present study was to determine whether lecithinized superoxide dismutase (PC-SOD) can serve as a cardioprotective drug during doxorubicin treatment. The protective potential of PC-SOD on doxorubicin-induced cardiotoxicity was investigated in BALB/c mice. The possible influence of PC-SOD on the antitumor activity of doxorubicin was investigated in vitro as well as in vivo. Mice were treated intravenously with doxorubicin (4 mg x kg(-1)) or doxorubicin and PC-SOD (5000, 20000 or 80000 U x kg(-1)) weekly x 6 and appropriate controls were included. Cardiotoxicity was monitored for 8 weeks by ECG measurement. The influence of PC-SOD on the antitumor activity of doxorubicin was evaluated in three human malignant cell lines. Nude mice bearing OVCAR-3 human ovarian cancer xenografts were treated intravenously with doxorubicin (8 mg x kg(-1)) alone or preceded by PC-SOD 20000 or 80000 U x kg(-1) weekly x 2 and appropriate controls were included. PC-SOD prevented doxorubicin-induced cardiotoxicity already at 5000 U x kg(-1) whereas 20000 and 80000 U x kg(-1) were equally protective. No toxicity was observed in mice treated with PC-SOD. PC-SOD did not interfere with the antiproliferative effects of doxorubicin in vitro. In vivo, PC-SOD had no negative effect on the inhibition of xenograft growth induced by doxorubicin. It can be concluded that PC-SOD protects the heart, but not the tumor against doxorubicin. These data suggest that PC-SOD may be a suitable cardioprotector during doxorubicin treatment.
Collapse
Affiliation(s)
- Gertjan J M den Hartog
- Department of Pharmacology and Toxicology, University Maastricht, 6200 MD Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
22
|
Masini E, Cuzzocrea S, Mazzon E, Marzocca C, Mannaioni PF, Salvemini D. Protective effects of M40403, a selective superoxide dismutase mimetic, in myocardial ischaemia and reperfusion injury in vivo. Br J Pharmacol 2002; 136:905-17. [PMID: 12110615 PMCID: PMC1573411 DOI: 10.1038/sj.bjp.0704774] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
1. Myocardial injury caused by ischaemia and reperfusion comes from multiple pathogenic events, including endothelial damage, neutrophil extravasation into tissue, mast cell activation, and peroxidation of cell membrane lipids. These events are followed by myocardial cell alterations resulting eventually in cell necrosis. An enhanced formation of reactive oxygen species is widely accepted as a stimulus for tissue destruction and cardiac failure. 2. In this study, we have investigated the cardioprotective effects of M40403 in myocardial ischaemia-reperfusion injury. M40403 is a low molecular weight, synthetic manganese containing superoxide dismutase mimetic (SODm) that selectively removes superoxide anion. Ischaemia was induced in rat hearts in vivo by ligating the left anterior descending coronary artery. Thirty minutes after the induction of ischaemia, the ligature was removed and reperfusion allowed to occur for at least 60 min. M40403 (0.1-1 mg kg(-1)) was given intravenously 15 min before ischaemia. 3. The results obtained in this study showed that M40403 significantly reduced the extent of myocardial damage, mast cell degranulation and the incidence of ventricular arrhythmias. Furthermore, M40403 significantly attenuated, in a dose-dependent manner, neutrophil infiltration in the myocardium as well as the associated induction of lipid peroxidation. Calcium overload seen post-reperfusion of the ischaemic myocardium was also reduced by M40403. 4. Immunohistochemical analysis for nitrotyrosine revealed a positive staining in cardiac tissue taken after reperfusion: this was attenuated by M40403. Moreover reperfused cardiac tissue sections showed positive staining for P-selectin and for anti-intercellular adhesion molecule (ICAM-1) in the vascular endothelial cells. M40403 treatment markedly reduced the intensity and degree of P-selectin and ICAM-1 in these tissues. No staining for nitrotyrosine, P-selectin or ICAM-1 was found in cardiac tissue taken at the end of the ischaemic period. 5. Overall, M40403 treatment reduced the morphological signs of myocardial cell injury and significantly improved survival. 6. Taken together, these results clearly indicate that M40403 treatment exerts a protective effect against ischaemia-reperfusion-induced myocardial injury, supporting a key role for superoxide anion in reperfusion injuries. This suggests that synthetic enzymes of SOD such as M40403, offer a novel therapeutic approach for the treatment of ischaemic heart disease where superoxide anion plays a dominant role.
Collapse
Affiliation(s)
- Emanuela Masini
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Florence, Italy
| | - Salvatore Cuzzocrea
- Institute of Pharmacology, School of Medicine, University of Messina, 98100 Messina, Italy
| | - Emanuela Mazzon
- Department of Biomorphology, School of Medicine, University of Messina, Italy
| | - Cosimo Marzocca
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Florence, Italy
| | - Pier Francesco Mannaioni
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Florence, Italy
| | - Daniela Salvemini
- MetaPhore Pharmaceuticals, 1910 Innerbelt Business Center Drive, St Louis, Missouri, MO 63114, U.S.A
- Author for correspondence:
| |
Collapse
|