1
|
Belenichev I, Popazova O, Bukhtiyarova N, Savchenko D, Oksenych V, Kamyshnyi O. Modulating Nitric Oxide: Implications for Cytotoxicity and Cytoprotection. Antioxidants (Basel) 2024; 13:504. [PMID: 38790609 PMCID: PMC11118938 DOI: 10.3390/antiox13050504] [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: 03/26/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
Despite the significant progress in the fields of biology, physiology, molecular medicine, and pharmacology; the designation of the properties of nitrogen monoxide in the regulation of life-supporting functions of the organism; and numerous works devoted to this molecule, there are still many open questions in this field. It is widely accepted that nitric oxide (•NO) is a unique molecule that, despite its extremely simple structure, has a wide range of functions in the body, including the cardiovascular system, the central nervous system (CNS), reproduction, the endocrine system, respiration, digestion, etc. Here, we systematize the properties of •NO, contributing in conditions of physiological norms, as well as in various pathological processes, to the mechanisms of cytoprotection and cytodestruction. Current experimental and clinical studies are contradictory in describing the role of •NO in the pathogenesis of many diseases of the cardiovascular system and CNS. We describe the mechanisms of cytoprotective action of •NO associated with the regulation of the expression of antiapoptotic and chaperone proteins and the regulation of mitochondrial function. The most prominent mechanisms of cytodestruction-the initiation of nitrosative and oxidative stresses, the production of reactive oxygen and nitrogen species, and participation in apoptosis and mitosis. The role of •NO in the formation of endothelial and mitochondrial dysfunction is also considered. Moreover, we focus on the various ways of pharmacological modulation in the nitroxidergic system that allow for a decrease in the cytodestructive mechanisms of •NO and increase cytoprotective ones.
Collapse
Affiliation(s)
- Igor Belenichev
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Olena Popazova
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Nina Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Dmytro Savchenko
- Department of Pharmacy and Industrial Drug Technology, Bogomolets National Medical University, 01601 Kyiv, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil State Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
2
|
Bala A. Regulatory role of peroxynitrite in advanced glycation end products mediated diabetic cardiovascular complications. World J Diabetes 2024; 15:572-574. [PMID: 38591082 PMCID: PMC10999052 DOI: 10.4239/wjd.v15.i3.572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 03/15/2024] Open
Abstract
The Advanced Glycation End Products (AGE) binding with its receptor can increase reactive oxygen species (ROS) generation through specific signaling mediators. The effect of superoxide (O2-) and O2- mediated ROS and reactive nitrogen species depends on their concentration and location of formation. Nitric oxide (NO) has anti-inflammatory and anticoagulant properties and a vasodilation effect, but NO can be deactivated by reacting with O2-. This reaction between NO and O2- produces the potent oxidant ONOO-. Therefore, ONOO-'s regulatory role in AGEs in diabetic cardiovascular complications must considered as a regulator of cardiovascular complications in diabetes.
Collapse
Affiliation(s)
- Asis Bala
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology, Guwahati 781035, Assam, India
| |
Collapse
|
3
|
Zhou J, Zhou S, Fan P, Li X, Ying Y, Ping J, Pan Y. Implantable Electrochemical Microsensors for In Vivo Monitoring of Animal Physiological Information. NANO-MICRO LETTERS 2023; 16:49. [PMID: 38087121 PMCID: PMC10716106 DOI: 10.1007/s40820-023-01274-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/24/2023] [Indexed: 10/11/2024]
Abstract
In vivo monitoring of animal physiological information plays a crucial role in promptly alerting humans to potential diseases in animals and aiding in the exploration of mechanisms underlying human diseases. Currently, implantable electrochemical microsensors have emerged as a prominent area of research. These microsensors not only fulfill the technical requirements for monitoring animal physiological information but also offer an ideal platform for integration. They have been extensively studied for their ability to monitor animal physiological information in a minimally invasive manner, characterized by their bloodless, painless features, and exceptional performance. The development of implantable electrochemical microsensors for in vivo monitoring of animal physiological information has witnessed significant scientific and technological advancements through dedicated efforts. This review commenced with a comprehensive discussion of the construction of microsensors, including the materials utilized and the methods employed for fabrication. Following this, we proceeded to explore the various implantation technologies employed for electrochemical microsensors. In addition, a comprehensive overview was provided of the various applications of implantable electrochemical microsensors, specifically in the monitoring of diseases and the investigation of disease mechanisms. Lastly, a concise conclusion was conducted on the recent advancements and significant obstacles pertaining to the practical implementation of implantable electrochemical microsensors.
Collapse
Affiliation(s)
- Jin Zhou
- Laboratory of Agricultural Information Intelligent Sensing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Shenghan Zhou
- Laboratory of Agricultural Information Intelligent Sensing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Peidi Fan
- Laboratory of Agricultural Information Intelligent Sensing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Xunjia Li
- Laboratory of Agricultural Information Intelligent Sensing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, People's Republic of China
| | - Yibin Ying
- Laboratory of Agricultural Information Intelligent Sensing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, People's Republic of China
| | - Jianfeng Ping
- Laboratory of Agricultural Information Intelligent Sensing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, People's Republic of China.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, People's Republic of China.
| | - Yuxiang Pan
- Laboratory of Agricultural Information Intelligent Sensing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, People's Republic of China.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, People's Republic of China.
| |
Collapse
|
4
|
Nedel W, Deutschendorf C, Portela LVC. Sepsis-induced mitochondrial dysfunction: A narrative review. World J Crit Care Med 2023; 12:139-152. [PMID: 37397587 PMCID: PMC10308342 DOI: 10.5492/wjccm.v12.i3.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/08/2023] [Accepted: 04/14/2023] [Indexed: 06/08/2023] Open
Abstract
Sepsis represents a deranged and exaggerated systemic inflammatory response to infection and is associated with vascular and metabolic abnormalities that trigger systemic organic dysfunction. Mitochondrial function has been shown to be severely impaired during the early phase of critical illness, with a reduction in biogenesis, increased generation of reactive oxygen species and a decrease in adenosine triphosphate synthesis of up to 50%. Mitochondrial dysfunction can be assessed using mitochondrial DNA concentration and respirometry assays, particularly in peripheral mononuclear cells. Isolation of monocytes and lymphocytes seems to be the most promising strategy for measuring mitochondrial activity in clinical settings because of the ease of collection, sample processing, and clinical relevance of the association between metabolic alterations and deficient immune responses in mononuclear cells. Studies have reported alterations in these variables in patients with sepsis compared with healthy controls and non-septic patients. However, few studies have explored the association between mitochondrial dysfunction in immune mononuclear cells and unfavorable clinical outcomes. An improvement in mitochondrial parameters in sepsis could theoretically serve as a biomarker of clinical recovery and response to oxygen and vasopressor therapies as well as reveal unexplored pathophysiological mechanistic targets. These features highlight the need for further studies on mitochondrial metabolism in immune cells as a feasible tool to evaluate patients in intensive care settings. The evaluation of mitochondrial metabolism is a promising tool for the evaluation and management of critically ill patients, especially those with sepsis. In this article, we explore the pathophysiological aspects, main methods of measurement, and the main studies in this field.
Collapse
Affiliation(s)
- Wagner Nedel
- Intensive Care Unit, Grupo Hospitalar Conceição, Porto Alegre 91350200, Brazil
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Brazilian Research in Intensive Care Network-BRICNet, São Paulo 04039-002, Brazil
| | - Caroline Deutschendorf
- Infection Control Committee, Hospital de Clínicas de Porto Alegre, Porto Alegre 90410-000, Brazil
| | - Luis Valmor Cruz Portela
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| |
Collapse
|
5
|
Dilken O, Ince C, Kapucu A, Heeman PM, Ergin B. Furosemide exacerbated the impairment of renal function, oxygenation and medullary damage in a rat model of renal ischemia/reperfusion induced AKI. Intensive Care Med Exp 2023; 11:25. [PMID: 37121963 PMCID: PMC10149155 DOI: 10.1186/s40635-023-00509-3] [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: 09/06/2022] [Accepted: 03/17/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND Perioperative acute kidney injury (AKI) caused by ischemia-reperfusion (IR) is a significant contributor to mortality and morbidity after major surgery. Furosemide is commonly used in postoperative patients to promote diuresis and reduce tissue edema. However, the effects of furosemide on renal microcirculation, oxygenation and function are poorly understood during perioperative period following ischemic insult. Herein, we investigated the effects of furosemide in rats subjected IR insult. METHODS 24 Wistar albino rats were divided into 4 groups, with 6 in each; Sham-operated Control (C), Control + Furosemide (C + F), ischemia/reperfusion (IR), and IR + F. After induction of anesthesia (BL), supra-aortic occlusion was applied to IR and IR + F groups for 45 min followed by ongoing reperfusion for 15 min (T1) and 2 h (T2). Furosemide infusion was initiated simultaneously in the intervention groups after ischemia. Renal blood flow (RBF), vascular resistance (RVR), oxygen delivery (DO2ren) and consumption (VO2ren), sodium reabsorption (TNa+), oxygen utilization efficiency (VO2/TNa+), cortical (CμO2) and medullary (MμO2) microvascular oxygen pressures, urine output (UO) and creatinine clearance (Ccr) were measured. Biomarkers of inflammation, oxidative and nitrosative stress were measured and kidneys were harvested for histological analysis. RESULTS IR significantly decreased RBF, mainly by increasing RVR, which was exacerbated in the IR + F group at T2 (2198 ± 879 vs 4233 ± 2636 dyne/s/cm5, p = 0.07). CμO2 (61.6 ± 6.8 vs 86 ± 6.6 mmHg) and MμO2 (51.1 ± 4.1 vs 68.7 ± 4.9 mmHg, p < 0.05) were both reduced after IR and did not improve by furosemide. Moreover, VO2/TNa+ increased in the IR + F group at T2 with respect to the IR group (IR: 3.3 ± 2 vs IR + F: 8.2 ± 10 p = 0.07) suggesting a possible deterioration of oxygen utilization. Ccr did not change, but plasma creatinine increased significantly in IR + F groups. Histopathology revealed widespread damage both in the cortex and medulla in IR, IR + F and C + F groups. CONCLUSION Renal microvascular oxygenation, renal function, renal vascular resistance, oxygen utilization and damage were not improved by furosemide administration after IR insult. Our study suggests that furosemide may cause additional structural and functional impairment to the kidney following ischemic injury and should be used with caution.
Collapse
Affiliation(s)
- Olcay Dilken
- Laboratory of Translational Intensive Care, Department of Intensive Care Adult, Erasmus MC, University Medical Center Rotterdam, Erasmus University, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Intensive Care, Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Turkey
| | - Can Ince
- Laboratory of Translational Intensive Care, Department of Intensive Care Adult, Erasmus MC, University Medical Center Rotterdam, Erasmus University, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Aysegul Kapucu
- Department of Zoology, Faculty of Science, University of Istanbul, Istanbul, Turkey
| | - Paul M Heeman
- Department of Medical Technical Innovation & Development (MIO), Amsterdam University Medical Centre (UMC) Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Bülent Ergin
- Laboratory of Translational Intensive Care, Department of Intensive Care Adult, Erasmus MC, University Medical Center Rotterdam, Erasmus University, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
6
|
Sakaguchi T, Sumiyama F, Kotsuka M, Hatta M, Yoshida T, Hayashi M, Kaibori M, Sekimoto M. Levosimendan Increases Survival in a D-Galactosamine and Lipopolysaccharide Rat Model. Biomedicines 2022; 10:biomedicines10123161. [PMID: 36551917 PMCID: PMC9775427 DOI: 10.3390/biomedicines10123161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Levosimendan, a calcium sensitizer, has an organ protective profile through the inhibition of inflammatory mediators and cytokines in critical conditions, such as heart failure, ischemia-reperfusion injury, and sepsis. The survival effect of levosimendan for acute liver failure has not been examined yet. Male Sprague-Dawley rats were examined in the D-galactosamine hydrochloride and lipopolysaccharide (GalN/LPS) model. Levosimendan was injected intraperitoneally before GalN/LPS treatment. Survival was monitored for 7 days. For biochemical analyses, liver and blood samples were collected from the rats at 1 and 8 h after GaIN/LPS treatment. The pretreatment of levosimendan at 4 mg/kg significantly increased survival in GalN/LPS rats. In the liver specimen, levosimendan significantly inhibited the activation of nuclear factor-κB (NF-κB) at 1 h, and significantly decreased the mRNA expression of inflammatory mediators, including inducible nitric oxide synthase and tumor necrosis factor-α (TNF-α), at 8 h. In serum, levosimendan decreased the levels of nitrite, a metabolite of nitric oxide, and TNF-α protein, as well as aspartate aminotransferase and alanine aminotransferase. These results indicated that Levosimendan ameliorated liver dysfunction and survival in acute liver failure model rats through the suppression of NF-κB activation.
Collapse
Affiliation(s)
- Tatsuma Sakaguchi
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
| | - Fusao Sumiyama
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
| | - Masaya Kotsuka
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
| | - Masahiko Hatta
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
| | - Terufumi Yoshida
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
| | - Mikio Hayashi
- Department of Physiology, Kansai Medical University, Hirakata 573-1010, Japan
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
- Correspondence: ; Tel.: +81-72-804-0101 (ext. 56130); Fax: +81-72-804-0170
| | - Mitsugu Sekimoto
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
| |
Collapse
|
7
|
Forceville X, Van Antwerpen P, Annane D, Vincent JL. Selenocompounds and Sepsis-Redox Bypass Hypothesis: Part B-Selenocompounds in the Management of Early Sepsis. Antioxid Redox Signal 2022; 37:998-1029. [PMID: 35287478 DOI: 10.1089/ars.2020.8062] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Endothelial barrier damage, which is in part caused by excess production of reactive oxygen, halogen and nitrogen species (ROHNS), especially peroxynitrite (ONOO-), is a major event in early sepsis and, with leukocyte hyperactivation, part of the generalized dysregulated immune response to infection, which may even become a complex maladaptive state. Selenoenzymes have major antioxidant functions. Their synthesis is related to the need to limit deleterious oxidant redox cycling by small selenocompounds, which may be of therapeutic cytotoxic interest. Plasma selenoprotein-P is crucial for selenium transport from the liver to the tissues and for antioxidant endothelial protection, especially against ONOO-. Above micromolar concentrations, sodium selenite (Na2SeO3) becomes cytotoxic, with a lower cytotoxicity threshold in activated cells, which has led to cancer research. Recent Advances: Plasma selenium (<2% of total body selenium) is mainly contained in selenoprotein-P, and concentrations decrease rapidly in the early phase of sepsis, because of increased selenoprotein-P binding and downregulation of hepatic synthesis and excretion. At low concentrations, Na2SeO3 acts as a selenium donor, favoring selenoprotein-P synthesis in physiology, but probably not in the acute phase of sepsis. Critical Issues: The cytotoxic effects of Na2SeO3 against hyperactivated leukocytes, especially the most immature forms that liberate ROHNS, may be beneficial, but they may also be harmful for activated endothelial cells. Endothelial protection against ROHNS by selenoprotein-P may reduce Na2SeO3 toxicity, which is increased in sepsis. Future Direction: The combination of selenoprotein-P for endothelial protection and the cytotoxic effects of Na2SeO3 against hyperactivated leukocytes may be a promising intervention for early sepsis. Antioxid. Redox Signal. 37, 998-1029.
Collapse
Affiliation(s)
- Xavier Forceville
- Medico-surgical Intensive Care Unit, Great Hospital of East Francilien - Meaux site, Meaux, France.,Clinical Investigation Centre (CIC Inserm1414) CHU de Rennes - Université de Rennes 1, Rennes, France
| | - Pierre Van Antwerpen
- Pharmacognosy, Bioanalysis and Drug Discovery and Analytical Platform of the Faculty of Pharmacy, Univesité libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Djillali Annane
- Service de Réanimation Médicale, Hôpital Raymond Poincaré (APHP), Garches, France.,U1173 Lab. of Inflammation & Infection, (Fédération Hospitalo-Universitaire) FHU SEPSIS, Université Paris Saclay-campus (Université de Versailles Saint-Quentin-en-Yvelines) UVSQ, Versailles, France
| | - Jean Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
8
|
Ma Q, Xu S, Zhai Z, Wang K, Liu X, Xiao H, Zhuo S, Liu Y. Recent Progress of Small‐Molecule Ratiometric Fluorescent Probes for Peroxynitrite in Biological Systems. Chemistry 2022; 28:e202200828. [DOI: 10.1002/chem.202200828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Qingqing Ma
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Shanlin Xu
- Department of Oncology, Zibo Central Hospital Zibo 255036 P. R. China
| | - Zhaodong Zhai
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Kai Wang
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Xueli Liu
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Haibin Xiao
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Shuping Zhuo
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Yuying Liu
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| |
Collapse
|
9
|
Santos SS, Brunialti MKC, Rodrigues LDOCP, Liberatore AMA, Koh IHJ, Martins V, Soriano FG, Szabo C, Salomão R. Effects of the PARP Inhibitor Olaparib on the Response of Human Peripheral Blood Leukocytes to Bacterial Challenge or Oxidative Stress. Biomolecules 2022; 12:biom12060788. [PMID: 35740913 PMCID: PMC9221060 DOI: 10.3390/biom12060788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022] Open
Abstract
Prior studies demonstrate the activation of poly-(ADP-ribose) polymerase 1 (PARP1) in various pathophysiological conditions, including sepsis. We have assessed the effect of olaparib, a clinically used PARP1 inhibitor, on the responses of human peripheral blood leukocytes (PBMCs) obtained from healthy volunteers in response to challenging with live bacteria, bacterial lipopolysaccharide (LPS), or oxidative stress (hydrogen peroxide, H2O2). The viability of PBMCs exposed to olaparib or to the earlier generation PARP inhibitor PJ-34 (0.1–1000 µM) was monitored using Annexin V and 7-aminoactinomycin D. To evaluate the effects of olaparib on the expression of PARP1 and its effects on protein PARylation, PBMCs were stimulated with Staphylococcus aureus with or without olaparib (1–10 μM). Changes in cellular levels of nicotinamide adenine dinucleotide (NAD+) and adenosine triphosphate (ATP), as well as changes in mitochondrial membrane potential (MMP), were measured in PBMCs exposed to H2O2. Bacterial killing was evaluated in PBMCs and polymorphonuclear leukocytes (PMNs) incubated with S. aureus. Cytokine production was measured in supernatants using a cytometric bead array. Reactive oxygen species (ROS), nitric oxide (NO) production, and phagocytic activity of monocytes and neutrophils were measured in whole blood. For ROS and NO production, samples were incubated with heat-killed S. aureus; phagocytic activity was assessed using killed Escherichia coli conjugated to FITC. Olaparib (0.1–100 µM) did not adversely affect lymphocyte viability. Olaparib also did not interfere with PARP1 expression but inhibits S. aureus-induced protein PARylation. In cells challenged with H2O2, olaparib prevented NAD+ and ATP depletion and attenuated mitochondrial membrane depolarization. LPS-induced production of TNF-α, MIP-1α, and IL-10 by PBMCs was also reduced by olaparib. Monocytes and neutrophils displayed significant increases in the production of ROS and NO after stimulation with S. aureus and phagocytic (E. coli) and microbicidal activity, and these responses were not suppressed by olaparib. We conclude that, at clinically relevant concentrations, olaparib exerts cytoprotective effects and modulates inflammatory cytokine production without exerting adverse effects on the cells’ ability to phagocytose or eradicate pathogens. The current data support the concept of repurposing olaparib as a potential experimental therapy for septic shock.
Collapse
Affiliation(s)
- Sidneia Sousa Santos
- Department of Medicine, Division of Infectious Diseases, Escola Paulista de Medicina, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04023, Brazil; (S.S.S.); (M.K.C.B.); (L.d.O.C.P.R.)
| | - Milena Karina Coló Brunialti
- Department of Medicine, Division of Infectious Diseases, Escola Paulista de Medicina, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04023, Brazil; (S.S.S.); (M.K.C.B.); (L.d.O.C.P.R.)
| | - Larissa de Oliveira Cavalcanti Peres Rodrigues
- Department of Medicine, Division of Infectious Diseases, Escola Paulista de Medicina, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04023, Brazil; (S.S.S.); (M.K.C.B.); (L.d.O.C.P.R.)
| | - Ana Maria Alvim Liberatore
- Discipline of Operative Technique and Experimental Surgery, Department of Surgery, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04023, Brazil; (A.M.A.L.); (I.H.J.K.)
| | - Ivan Hong Jun Koh
- Discipline of Operative Technique and Experimental Surgery, Department of Surgery, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04023, Brazil; (A.M.A.L.); (I.H.J.K.)
| | - Vanessa Martins
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland;
| | - Francisco Garcia Soriano
- Laboratory of Medical Research, Faculty of Medicine, University of São Paulo, São Paulo 05403, Brazil;
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland;
- Correspondence: (C.S.); (R.S.)
| | - Reinaldo Salomão
- Department of Medicine, Division of Infectious Diseases, Escola Paulista de Medicina, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04023, Brazil; (S.S.S.); (M.K.C.B.); (L.d.O.C.P.R.)
- Correspondence: (C.S.); (R.S.)
| |
Collapse
|
10
|
Raia L, Zafrani L. Endothelial Activation and Microcirculatory Disorders in Sepsis. Front Med (Lausanne) 2022; 9:907992. [PMID: 35721048 PMCID: PMC9204048 DOI: 10.3389/fmed.2022.907992] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 11/19/2022] Open
Abstract
The vascular endothelium is crucial for the maintenance of vascular homeostasis. Moreover, in sepsis, endothelial cells can acquire new properties and actively participate in the host's response. If endothelial activation is mostly necessary and efficient in eliminating a pathogen, an exaggerated and maladaptive reaction leads to severe microcirculatory damage. The microcirculatory disorders in sepsis are well known to be associated with poor outcome. Better recognition of microcirculatory alteration is therefore essential to identify patients with the worse outcomes and to guide therapeutic interventions. In this review, we will discuss the main features of endothelial activation and dysfunction in sepsis, its assessment at the bedside, and the main advances in microcirculatory resuscitation.
Collapse
Affiliation(s)
- Lisa Raia
- Medical Intensive Care Unit, Hôpital Saint-Louis, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Lara Zafrani
- Medical Intensive Care Unit, Hôpital Saint-Louis, Assistance Publique des Hôpitaux de Paris, Paris, France
- INSERM UMR 976, University of Paris Cité, Paris, France
- *Correspondence: Lara Zafrani
| |
Collapse
|
11
|
Choromańska B, Myśliwiec P, Kozłowski T, Łuba M, Wojskowicz P, Dadan J, Myśliwiec H, Choromańska K, Makarewicz K, Zalewska A, Maciejczyk M. Cross-Talk Between Nitrosative Stress, Inflammation and Hypoxia-Inducible Factor in Patients with Adrenal Masses. J Inflamm Res 2021; 14:6317-6330. [PMID: 34876829 PMCID: PMC8643214 DOI: 10.2147/jir.s337910] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/17/2021] [Indexed: 12/23/2022] Open
Abstract
Background Adrenal masses are the most common of all human tumors. The role of nitrosative stress and inflammation in cancer development has already been demonstrated. However, it is not known whether they are involved in the pathogenesis of adrenal tumors. The aim of the study was to investigate a cross-talk between nitrosative stress, inflammation and hypoxia-inducible factor (HIF-1α) in 75 patients with different types of adrenal masses (non-functional incidentaloma, pheochromocytoma and Cushing's/Conn's adenoma). Methods The plasma concentrations of total nitric oxide (NO), S-nitrosothiols, peroxynitrite nitrotyrosine and the activity of serum myeloperoxidase (MPO) were measured spectrophotometrically, whereas concentrations of interleukin 1 beta (IL-1β), tumor necrosis factor α (TNF-α) and hypoxia-inducible factor 1 alpha (HIF-1α) were measured using commercial ELISA kits. The control group consisted of 50 healthy people matched by age and sex to the study group. The number of subjects was determined a priori based on our previous experiment (power of the test = 0.9; α = 0.05). Results We found significantly higher nitrosative stress (↑nitric oxide, ↑peroxynitrite, ↑S-nitrosothiols and ↑nitrotyrosine) in the plasma of patients with adrenal tumors, which was accompanied by increased inflammatory (↑myeloperoxidase, ↑interleukin 1 beta and ↑tumor necrosis factor α) and hypoxia (HIF-1α) biomarkers. Peroxynitrite and nitrotyrosine were positively correlated with aldosterone level. Nitrosative stress was also associated with inflammation and HIF-1α. Interestingly, plasma nitrotyrosine and serum MPO differentiated patients with adrenal tumor from healthy individuals with high sensitivity and specificity. Moreover, using multivariate regression analysis, we showed that ONOO- and IL-1β depended on cortisol level, while ONOO-, nitrotyrosine and HIF-1α were associated with aldosterone. Unfortunately, none of the assessed biomarkers differentiated between tumor types studied, suggesting that the severity of nitrosative damage and inflammation are similar in patients with incidentaloma, pheochromocytoma, and Cushing's or Conn's adenoma. Conclusion Adrenal tumors are associated with increased protein nitration/S-nitrosylation and inflammation.
Collapse
Affiliation(s)
- Barbara Choromańska
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Myśliwiec
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Tomasz Kozłowski
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Magdalena Łuba
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Wojskowicz
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Jacek Dadan
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Hanna Myśliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, Bialystok, Poland
| | | | | | - Anna Zalewska
- Experimental Dentistry Laboratory, Medical University of Bialystok, Bialystok, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
12
|
Santos SS, Brunialti MKC, Soriano FG, Szabo C, Salomão R. Repurposing of Clinically Approved Poly-(ADP-Ribose) Polymerase Inhibitors for the Therapy of Sepsis. Shock 2021; 56:901-909. [PMID: 34115723 DOI: 10.1097/shk.0000000000001820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Sepsis' pathogenesis involves multiple mechanisms that lead to a dysregulation of the host's response. Significant efforts have been made in search of interventions that can reverse this situation and increase patient survival. Poly (ADP-polymerase) (PARP) is a constitutive nuclear and mitochondrial enzyme, which functions as a co-activator and co-repressor of gene transcription, thus regulating the production of inflammatory mediators. Several studies have already demonstrated an overactivation of PARP1 in various human pathophysiological conditions and that its inhibition has benefits in regulating intracellular processes. The PARP inhibitor olaparib, originally developed for cancer therapy, paved the way for the expansion of its clinical use for nononcological indications. In this review we discuss sepsis as one of the possible indications for the use of olaparib and other clinically approved PARP inhibitors as modulators of the inflammatory response and cellular dysfunction. The benefit of olaparib and other clinically approved PARP inhibitors has already been demonstrated in several experimental models of human diseases, such as neurodegeneration and neuroinflammation, acute hepatitis, skeletal muscle disorders, aging and acute ischemic stroke, protecting, for example, from the deterioration of the blood-brain barrier, restoring the cellular levels of NAD+, improving mitochondrial function and biogenesis and, among other effects, reducing oxidative stress and pro-inflammatory mediators, such as TNF-α, IL1-β, IL-6, and VCAM1. These data demonstrated that repositioning of clinically approved PARP inhibitors may be effective in protecting against hemodynamic dysfunction, metabolic dysfunction, and multiple organ failure in patients with sepsis. Age and gender affect the response to PARP inhibitors, the mechanisms underlying the lack of many protective effects in females and aged animals should be further investigated and be cautiously considered in designing clinical trials.
Collapse
Affiliation(s)
- Sidnéia Sousa Santos
- Division of Infectious Diseasses, Paulista School of Medicine, Federal University of Sao Paulo, Brazil
| | | | - Francisco Garcia Soriano
- Laboratory of Medical Research, Faculty of Medicine of the University of São Paulo-USP, São Paulo, Brazil
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinaldo Salomão
- Division of Infectious Diseasses, Paulista School of Medicine, Federal University of Sao Paulo, Brazil
| |
Collapse
|
13
|
Joffre J, Hellman J. Oxidative Stress and Endothelial Dysfunction in Sepsis and Acute Inflammation. Antioxid Redox Signal 2021; 35:1291-1307. [PMID: 33637016 DOI: 10.1089/ars.2021.0027] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Under homeostatic conditions, the endothelium dynamically regulates vascular barrier function, coagulation pathways, leukocyte adhesion, and vasomotor tone. During sepsis and acute inflammation, endothelial cells (ECs) undergo multiple phenotypic and functional modifications that are initially adaptive but eventually become harmful, leading to microvascular dysfunction and multiorgan failure. Critical Issues and Recent Advances: Sepsis unbalances the redox homeostasis toward a pro-oxidant state, characterized by an excess production of reactive oxygen species and reactive nitrogen species, mitochondrial dysfunction, and a breakdown of antioxidant systems. In return, oxidative stress (OS) alters multiple EC functions and promotes a proinflammatory, procoagulant, and proadhesive phenotype. The OS also induces glycocalyx deterioration, cell death, increased permeability, and impaired vasoreactivity. Thus, during sepsis, the ECs are both a significant source and one of the main targets of OS. Future Directions: This review aims at covering the current understanding of the role of OS in the endothelial adaptive or maladaptive multifaceted response to sepsis and to outline the therapeutic potential and issues of targeting OS and endothelial dysfunction during sepsis and septic shock. One of the many challenges in the management of sepsis is now based on the detection and correction of these anomalies of endothelial function.
Collapse
Affiliation(s)
- Jérémie Joffre
- Department of Anesthesia and Perioperative Care, University of California, San Francisco School of Medicine, San Francisco, California, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco School of Medicine, San Francisco, California, USA
| |
Collapse
|
14
|
Guarneiri LL, Paton CM, Cooper JA. Pecan-enriched diets decrease postprandial lipid peroxidation and increase total antioxidant capacity in adults at-risk for cardiovascular disease. Nutr Res 2021; 93:69-78. [PMID: 34428717 DOI: 10.1016/j.nutres.2021.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022]
Abstract
Pecans are a rich source of antioxidants, but the effect of regular consumption on post-meal responses is unknown. The objective of this study was to examine the impact of daily pecan consumption for 8 weeks on fasting and postprandial lipid peroxidation, total antioxidant capacity (TAC), and tocopherols in adults at higher risk for cardiovascular disease (CVD) (hypercholesterolemia or elevated adiposity). We hypothesized that daily pecan consumption would result in increased fasting γ-tocopherol, increased fasting and postprandial TAC, and decreased fasting and postprandial lipid peroxidation. This was a randomized, parallel, controlled trial with 3 treatments: two pecan groups and a nut free control (n = 16). The ADD group (n = 15) consumed pecans as part of a free-living diet, and the SUB group (n = 16) substituted the pecans for isocaloric foods from their habitual diet. At the pre- and post-intervention, a high saturated fat breakfast shake was consumed with postprandial blood draws over 2h. In the ADD and SUB groups, postprandial lipid peroxidation was suppressed (iAUC: 0.9 ± 1.3 to -2.9 ± 2.0 and 4.5 ± 1.7 to 0.7 ± 1.1 µM/2h, respectively; P <0.05) and TAC was elevated (iAUC: -240.8 ± 110.2 to 130.9 ± 131.7 and -227.6 ± 131.2 to 208.7 ± 145.7 µM Trolox Equivalents/2h, respectively; P <0.01) from pre- to post-intervention. Furthermore, there was an increase in γ-tocopherol from pre- to post-intervention within the ADD (1.4 ± 0.1 to 1.8 ± 0.1 µg/mL; P <0.001) and SUB groups (1.8 ± 0.2 to 2.1 ± 0.2 µg/mL; P <0.05). There were no changes in any variable within the control group. These findings suggest that daily pecan consumption protects against oxidative stress that occurs following a high-fat meal in adults at risk for CVD.
Collapse
Affiliation(s)
- Liana L Guarneiri
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia
| | - Chad M Paton
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia; Department of Food Science and Technology, University of Georgia, Athens, Georgia
| | - Jamie A Cooper
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia.
| |
Collapse
|
15
|
Forceville X, Van Antwerpen P, Preiser JC. Selenocompounds and Sepsis: Redox Bypass Hypothesis for Early Diagnosis and Treatment: Part A-Early Acute Phase of Sepsis: An Extraordinary Redox Situation (Leukocyte/Endothelium Interaction Leading to Endothelial Damage). Antioxid Redox Signal 2021; 35:113-138. [PMID: 33567962 DOI: 10.1089/ars.2020.8063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Sepsis is a health disaster. In sepsis, an initial, beneficial local immune response against infection evolves rapidly into a generalized, dysregulated response or a state of chaos, leading to multiple organ failure. Use of life-sustaining supportive therapies creates an unnatural condition, enabling the complex cascades of the sepsis response to develop in patients who would otherwise die. Multiple attempts to control sepsis at an early stage have been unsuccessful. Recent Advances: Major events in early sepsis include activation and binding of leukocytes and endothelial cells in the microcirculation, damage of the endothelial surface layer (ESL), and a decrease in the plasma concentration of the antioxidant enzyme, selenoprotein-P. These events induce an increase in intracellular redox potential and lymphocyte apoptosis, whereas apoptosis is delayed in monocytes and neutrophils. They also induce endothelial mitochondrial and cell damage. Critical Issues: Neutrophil production increases dramatically, and aggressive immature forms are released. Leukocyte cross talk with other leukocytes and with damaged endothelial cells amplifies the inflammatory response. The release of large quantities of reactive oxygen, halogen, and nitrogen species as a result of the leukocyte respiratory burst, endothelial mitochondrial damage, and ischemia/reperfusion processes, along with the marked decrease in selenoprotein-P concentrations, leads to peroxynitrite damage of the ESL, reducing flow and damaging the endothelial barrier. Future Directions: Endothelial barrier damage by activated leukocytes is a time-sensitive event in sepsis, occurring within hours and representing the first step toward organ failure and death. Reducing or stopping this event is necessary before irreversible damage occurs.
Collapse
Affiliation(s)
- Xavier Forceville
- Medico-Surgical Intensive Care Unit, Great Hospital of East Francilien-Meaux Site, Hôpital Saint Faron, Meaux, France.,Clinical Investigation Center (CIC Inserm 1414), CHU de Rennes, Université de Rennes 1, Rennes, France
| | - Pierre Van Antwerpen
- Pharmacognosy, Bioanalysis and Drug Discovery and Analytical Platform of the Faculty of Pharmacy, Université libre de Bruxelles (ULB), Bruxelles, Belgium
| | | |
Collapse
|
16
|
Macrophage-produced peroxynitrite induces antibiotic tolerance and supersedes intrinsic mechanisms of persister formation. Infect Immun 2021; 89:e0028621. [PMID: 34097475 DOI: 10.1128/iai.00286-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus is a leading human pathogen that frequently causes chronic and relapsing infections. Antibiotic tolerant persister cells contribute to frequent antibiotic failure in patients. Macrophages represent an important niche during S. aureus bacteremia and recent work has identified a role for oxidative burst in the formation of antibiotic tolerant S. aureus. We find that host-derived peroxynitrite, the reaction product of superoxide and nitric oxide, is the main mediator of antibiotic tolerance in macrophages. Using a collection of S. aureus clinical isolates, we find that, despite significant variation in persister formation in pure culture, all strains were similarly enriched for antibiotic tolerance following internalization by activated macrophages. Our findings suggest that host interaction strongly induces antibiotic tolerance and may negate bacterial mechanisms of persister formation, established in pure culture. These findings emphasize the importance of studying antibiotic tolerance in the context of bacterial interaction with the host suggest that modulation of the host response may represent a viable therapeutic strategy to sensitize S. aureus to antibiotics.
Collapse
|
17
|
Huang S, Zhang L, Dai L, Wang Y, Tian Y. Nonenzymatic Electrochemical Sensor with Ratiometric Signal Output for Selective Determination of Superoxide Anion in Rat Brain. Anal Chem 2021; 93:5570-5576. [PMID: 33757286 DOI: 10.1021/acs.analchem.1c00151] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
There is still an urgent need to develop reliable analytical methods of O2•- in vivo for deeply elucidating the roles of O2•- playing in the brain. Herein, a nonenzymatic electrochemical sensor with ratiometric signal output was developed for an in vivo analysis of O2•- in the rat brain. Diphenylphosphonate-2-naphthol ester (ND) was designed and synthesized as a specific recognition molecule for the selective determination of O2•-. An anodic peak ascribed to the oxidation of 2-naphthol was generated via the nucleophilic substitution between ND and O2•- and was increased with the increasing concentration of O2•-. Meanwhile, the inner reference of methylene blue (MB) was co-assembled at the electrode surface to enhance the determination accuracy of O2•-. The anodic peak current ratio between 2-naphthol and MB exhibited a good linear relationship with the concentration of O2•- from 2 to 200 μM. Because of the stable molecule character of ND and its specific reaction with O2•-, the developed electrochemical sensor demonstrated excellent selectivity toward various potential interferences in the brain and good stability even after storage for 7 days. Accordingly, the present electrochemical sensor with high selectivity, high stability, and high accuracy was successfully exploited in monitoring the levels of O2•- in the rat brain and that of the diabetic model followed by cerebral ischemia.
Collapse
Affiliation(s)
- Shiqi Huang
- School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| | - Limin Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| | - Liyi Dai
- School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| | - Yuanyuan Wang
- School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| | - Yang Tian
- School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| |
Collapse
|
18
|
Structure-Dependent Effects of Bisphosphonates on Inflammatory Responses in Cultured Neonatal Mouse Calvaria. Antioxidants (Basel) 2020; 9:antiox9060503. [PMID: 32526922 PMCID: PMC7346192 DOI: 10.3390/antiox9060503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022] Open
Abstract
Bisphosphonates (BPs) are classified into two groups, according to their side chain structures, as nitrogen-containing BPs (NBPs) and non-nitrogen-containing BPs (non-NBPs). In this study, we examined the effects of NBPs and non-NBPs on inflammatory responses, by quantifying the inflammatory mediators, prostaglandin E2 (PGE2) and nitric oxide (NO), in cultured neonatal mouse calvaria. All examined NBPs (pamidronate, alendronate, incadronate, risedronate, zoledronate) stimulated lipopolysaccharide (LPS)-induced PGE2 and NO production by upregulating COX-2 and iNOS mRNA expression, whereas non-NBPs (etidronate, clodronate, tiludronate) suppressed PGE2 and NO production, by downregulating gene expression. Additionally, [4-(methylthio) phenylthio] methane bisphosphonate (MPMBP), a novel non-NBP with an antioxidant methylthio phenylthio group in its side chain, exhibited the most potent anti-inflammatory activity among non-NBPs. Furthermore, results of immunohistochemistry showed that the nuclear translocation of NF-κB/p65 and tyrosine nitration of cytoplasmic protein were stimulated by zoledronate, while MPMBP inhibited these phenomena, by acting as a superoxide anion (O2−) scavenger. These findings indicate that MPMBP can act as an efficacious agent that causes fewer adverse effects in patients with inflammatory bone diseases, including periodontitis and rheumatoid arthritis.
Collapse
|
19
|
Guo B, Bai Y, Ma Y, Liu C, Wang S, Zhao R, Dong J, Ji HL. Preclinical and clinical studies of smoke-inhalation-induced acute lung injury: update on both pathogenesis and innovative therapy. Ther Adv Respir Dis 2019; 13:1753466619847901. [PMID: 31068086 PMCID: PMC6515845 DOI: 10.1177/1753466619847901] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Smoke-inhalation-induced acute lung injury (SI-ALI) is a leading cause of morbidity and mortality in victims of fire tragedies. SI-ALI contributes to an estimated 30% of burn-caused patient deaths, and recently, more attention has been paid to the specific interventions for this devastating respiratory illness. In the last decade, much progress has been made in the understanding of SI-ALI patho-mechanisms and in the development of new therapeutic strategies in both preclinical and clinical studies. This article reviews the recent progress in the treatment of SI-ALI, based on pathophysiology, thermal damage, airway obstruction, the nuclear-factor kappa-B signaling pathway, and oxidative stress. Preclinical therapeutic strategies include use of mesenchymal stem cells, hydrogen sulfide, peroxynitrite decomposition catalysts, and proton-pump inhibitors. Clinical interventions include high-frequency percussive ventilation, perfluorohexane, inhaled anticoagulants, and nebulized epinephrine. The animal model, dose, clinical application, and pharmacology of these medications are summarized. Future directions and further needs for developing innovative therapies are discussed.
Collapse
Affiliation(s)
- Bingxin Guo
- Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang Henan, China
| | - Yichun Bai
- Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang Henan, China
| | - Yana Ma
- Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang Henan, China
| | - Cong Liu
- Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang Henan, China
| | - Song Wang
- Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang Henan, China
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Jiaxing Dong
- Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang Henan, China
| | - Hong-Long Ji
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, TX, USA
| |
Collapse
|
20
|
Fukuda S, Ihara K, Andersen CR, Randolph AC, Nelson CL, Zeng Y, Kim J, DeWitt DS, Rojas JD, Koutrouvelis A, Herndon DN, Prough DS, Enkhbaatar P. Modulation of Peroxynitrite Reduces Norepinephrine Requirements in Ovine MRSA Septic Shock. Shock 2019; 52:e92-e99. [PMID: 30499879 DOI: 10.1097/shk.0000000000001297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular hypo-responsiveness to vasopressors during septic shock is a challenging problem. This study is to test the hypothesis that reactive nitrogen species (RNS), such as peroxynitrite, are major contributing factors to vascular hypo-responsiveness in septic shock. We hypothesized that adjunct therapy with peroxynitrite decomposition catalyst (PDC) would reduce norepinephrine requirements in sepsis resuscitation. Fourteen female Merino sheep were subjected to a "two-hit" injury (smoke inhalation and endobronchial instillation of live methicillin-resistant Staphylococcus aureus [1.6-2.5 × 10 CFUs]). The animals were randomly allocated to control: injured, fluid resuscitated, and titrated norepinephrine, n = 7; or PDC: injured, fluid resuscitated, titrated norepinephrine, and treated with PDC, n = 7. One-hour postinjury, an intravenous injection of PDC (0.1 mg/kg) was followed by a continuous infusion (0.04 mg/kg/h). Titration of norepinephrine started at 0.05 mcg/kg/min based on their mean arterial pressure. All animals were mechanically ventilated and monitored in the conscious state for 24 h. The mean arterial pressure was well maintained in the PDC with significantly less norepinephrine requirement from 7 to 23 h after injury compared with control. Total norepinephrine dose, the highest norepinephrine rate, and time on norepinephrine support were also significantly lower in PDC. Modified sheep organ failure assessment scores at 6 to 18 h postinjury were significantly lower in PDC compared with control. PDC improved survival rate at 24 h (71.4% vs. 28.6%). PDC treatment had no adverse effects. In conclusion, the modulation of RNS may be considered an effective adjunct therapy for septic shock, in the case of hypo-responsiveness to norepinephrine.
Collapse
Affiliation(s)
- Satoshi Fukuda
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
- Shriners Hospital for Children, Galveston, Texas
| | - Koji Ihara
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Clark R Andersen
- Department of Preventive Medicine and Community Health, Office of Biostatistics, University of Texas Medical Branch, Galveston, Texas
| | - Anita C Randolph
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Christina L Nelson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Yaping Zeng
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Jisoo Kim
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Douglas S DeWitt
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Jose D Rojas
- Department of Respiratory Care, School of Health Professions, University of Texas Medical Branch, Galveston, Texas
| | | | | | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
- Shriners Hospital for Children, Galveston, Texas
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
- Shriners Hospital for Children, Galveston, Texas
| |
Collapse
|
21
|
Nakatake R, Hishikawa H, Kotsuka M, Ishizaki M, Matsui K, Nishizawa M, Yoshizawa K, Kaibori M, Okumura T. The Proton Pump Inhibitor Lansoprazole Has Hepatoprotective Effects in In Vitro and In Vivo Rat Models of Acute Liver Injury. Dig Dis Sci 2019; 64:2854-2866. [PMID: 30989463 DOI: 10.1007/s10620-019-05622-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 04/08/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS The proton pump inhibitor lansoprazole (LPZ) is clinically used to reduce gastric acid secretion, but little is known about its possible hepatoprotective effects. This study aimed to investigate the hepatoprotective effects of LPZ and its potential mechanisms using in vitro and in vivo rat models of liver injury. METHODS For the in vitro model of liver injury, primary cultured rat hepatocytes were treated with interleukin-1β in the presence or absence of LPZ. The influence of LPZ on inducible nitric oxide synthase (iNOS) induction and nitric oxide (NO) production and on the associated signaling pathways was analyzed. For the in vivo model, rats were treated with D-galactosamine (GalN) and lipopolysaccharide (LPS). The effects of LPZ on survival and proinflammatory mediator expression (including iNOS and tumor necrosis factor-α) in these rats were examined. RESULTS LPZ inhibited iNOS induction partially through suppression of the nuclear factor-kappa B signaling pathway in hepatocytes, thereby reducing potential liver injury from excessive NO levels. Additionally, LPZ increased survival by 50% and decreased iNOS, tumor necrosis factor-α, and cytokine-induced neutrophil chemoattractant-1 mRNA expression in the livers of GalN/LPS-treated rats. LPZ also inhibited nuclear factor-kappa B activation by GalN/LPS. CONCLUSIONS LPZ inhibits the induction of several inflammatory mediators (including cytokines, chemokines, and NO) partially through suppression of nuclear factor-kappa B, resulting in the prevention of fulminant liver failure. The therapeutic potential of LPZ for liver injuries warrants further investigation.
Collapse
Affiliation(s)
- Richi Nakatake
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Hidehiko Hishikawa
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Masaya Kotsuka
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Morihiko Ishizaki
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Kosuke Matsui
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Mikio Nishizawa
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Katsuhiko Yoshizawa
- Laboratory of Environmental Sciences, Department of Food Sciences and Nutrition, School of Human Environmental Sciences, Mukogawa Women's University, 6-46 Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Tadayoshi Okumura
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.,Research Organization of Science and Technology, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
22
|
Levosimendan pretreatment improves survival of septic rats after partial hepatectomy and suppresses iNOS induction in cytokine-stimulated hepatocytes. Sci Rep 2019; 9:13398. [PMID: 31527618 PMCID: PMC6746814 DOI: 10.1038/s41598-019-48792-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 01/09/2019] [Indexed: 01/01/2023] Open
Abstract
We evaluated the survival effects and biochemical profiles of levosimendan in septic rats after partial hepatectomy and investigated its effects in cultured hepatocytes. Thirty-two rats underwent 70% hepatectomy and were randomised equally into four groups, followed by lipopolysaccharide (LPS) injection (250 µg/kg, i.v.) after 48 h. Levosimendan was given (i.p.) 1 h before LPS injection [group (A) levosimendan 2 mg/kg; (B) 1; (C) 0.5; (D) vehicle]. Survival at 7 days was increased significantly in group A compared with that in group D [A: 63%; B: 38%; C: 13%; D: 0%]. In serum, levosimendan decreased the level of tumour necrosis factor-α, interleukin (IL)-1β, IL-6 and nitric oxide (NO). In remnant livers, levosimendan inhibited inducible nitric oxide synthase (iNOS) gene expression. In primary cultured rat hepatocytes stimulated by IL-1β, levosimendan suppressed NO production by inhibiting iNOS promoter activity and stability of its mRNA.
Collapse
|
23
|
Fujiwara O, Fukuda S, Lopez E, Zeng Y, Niimi Y, DeWitt DS, Herndon DN, Prough DS, Enkhbaatar P. Peroxynitrite decomposition catalyst reduces vasopressin requirement in ovine MRSA sepsis. Intensive Care Med Exp 2019; 7:12. [PMID: 31512009 PMCID: PMC6738358 DOI: 10.1186/s40635-019-0227-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
Background Sepsis is one of the most frequent causes of death in the intensive care unit. Host vascular hypo-responsiveness to vasopressors during septic shock is one of the challenging problems. This study tested the hypothesis that adjunct therapy with peroxynitrite decomposition catalyst (WW-85) would reduce arginine vasopressin (AVP) requirements during sepsis resuscitation, using ovine sepsis model. Methods Thirteen adult female Merino sheep, previously instrumented with multiple vascular catheters, were subjected to “two-hit” (cotton smoke inhalation and intrapulmonary instillation of live methicillin-resistant Staphylococcus aureus; 3.5 × 1011 colony-forming units) injury. Post injury, animals were awakened and randomly allocated to the following groups: (1) AVP: injured, fluid resuscitated, and titrated with AVP, n = 6 or (2) WW-85 + AVP: injured, fluid resuscitated, treated with WW-85, and titrated with AVP, n = 7. One-hour post injury, a bolus intravenous injection of WW-85 (0.1 mg/kg) was followed by a 23-h continuous infusion (0.02 mg/kg/h). Titration of AVP started at a dose of 0.01 unit/min, when mean arterial pressure (MAP) decreased by 10 mmHg from baseline, despite aggressive fluid resuscitation, and the rate was further adjusted to maintain MAP. After the injury, all animals were placed on a mechanical ventilator and monitored in the conscious state for 24 h. Results The injury induced severe hypotension refractory to aggressive fluid resuscitation. High doses of AVP were required to partially attenuate the sepsis-induced hypotension. However, the cumulative AVP requirement was significantly reduced by adjunct treatment with WW-85 at 17–24 h after the injury (p < 0.05). Total AVP dose and the highest AVP rate were significantly lower in the WW-85 + AVP group compared to the AVP group (p = 0.02 and 0.04, respectively). Treatment with WW-85 had no adverse effects. In addition, the in vitro effects of AVP on isolated artery diameter changes were abolished with peroxynitrite co-incubation. Conclusions The modulation of reactive nitrogen species, such as peroxynitrite, may be considered as a novel adjunct treatment option for septic shock associated with vascular hypo-responsiveness to vasopressors.
Collapse
Affiliation(s)
- Osamu Fujiwara
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA
| | - Satoshi Fukuda
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA
| | - Ernesto Lopez
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA
| | - Yaping Zeng
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA
| | - Yosuke Niimi
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA
| | - Douglas S DeWitt
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA
| | - David N Herndon
- Shriners Hospital for Children, Galveston, Texas, USA.,Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555 1102, USA. .,Shriners Hospital for Children, Galveston, Texas, USA.
| |
Collapse
|
24
|
Abstract
Multiple organ dysfunction syndrome (MODS) is one of the most common syndromes of critical illness and the leading cause of mortality among critically ill patients. Multiple organ dysfunction syndrome is the clinical consequence of a dysregulated inflammatory response, triggered by clinically diverse factors with the main pillar of management being invasive organ support. During the last years, the advances in the clarification of the molecular pathways that trigger, mitigate, and determine the outcome of MODS have led to the increasing recognition of MODS as a distinct disease entity with distinct etiology, pathophysiology, and potential future therapeutic interventions. Given the lack of effective treatment for MODS, its early recognition, the early intensive care unit admission, and the initiation of invasive organ support remain the most effective strategies of preventing its progression and improving outcomes.
Collapse
Affiliation(s)
- Nicholas M Gourd
- Department of Intensive Care Medicine, Derriford Hospital, 6634University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom.,Faculty of Medicine and Dentistry, 6634University of Plymouth, Plymouth, United Kingdom
| | - Nikitas Nikitas
- Department of Intensive Care Medicine, Derriford Hospital, 6634University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom
| |
Collapse
|
25
|
Wepler M, Merz T, Wachter U, Vogt J, Calzia E, Scheuerle A, Möller P, Gröger M, Kress S, Fink M, Lukaschewski B, Rumm G, Stahl B, Georgieff M, Huber-Lang M, Torregrossa R, Whiteman M, McCook O, Radermacher P, Hartmann C. The Mitochondria-Targeted H2S-Donor AP39 in a Murine Model of Combined Hemorrhagic Shock and Blunt Chest Trauma. Shock 2019; 52:230-239. [PMID: 29927788 DOI: 10.1097/shk.0000000000001210] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Hemorrhagic shock (HS) accounts for 30% to 40% of trauma-induced mortality, which is due to multi-organ-failure subsequent to systemic hyper-inflammation, triggered by hypoxemia and tissue ischemia. The slow-releasing, mitochondria-targeted H2S donor AP39 exerted beneficial effects in several models of ischemia-reperfusion injury and acute inflammation. Therefore, we tested the effects of AP39-treatment in a murine model of combined blunt chest trauma (TxT) and HS with subsequent resuscitation. METHODS After blast wave-induced TxT or sham procedure, anesthetized and instrumented mice underwent 1 h of hemorrhage followed by 4 h of resuscitation comprising an i.v. bolus injection of 100 or 10 nmol kg AP39 or vehicle, retransfusion of shed blood, fluid resuscitation, and norepinephrine. Lung mechanics and gas exchange were assessed together with hemodynamics, metabolism, and acid-base status. Blood and tissue samples were analyzed for cytokine and chemokine levels, western blot, immunohistochemistry, mitochondrial oxygen consumption (JO2), and histological changes. RESULTS High dose AP39 attenuated systemic inflammation and reduced the expression of inducible nitric oxide synthase (iNOS) and IκBα expression in lung tissue. In the combined trauma group (TxT + HS), animals treated with high dose AP39 presented with the lowest mean arterial pressure and thus highest norepinephrine requirements and higher mortality. Low dose AP39 had no effects on hemodynamics, leading to unchanged norepinephrine requirements and mortality rates. CONCLUSION AP39 is a systemic anti-inflammatory agent. In our model of trauma with HS, there may be a narrow dosing and timing window due to its potent vasodilatory properties, which might result in or contribute to aggravation of circulatory shock-related hypotension.
Collapse
Affiliation(s)
- Martin Wepler
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
- Department of Anesthesiology, University Hospital, Ulm, Germany
| | - Tamara Merz
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Ulrich Wachter
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Josef Vogt
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Enrico Calzia
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | | | - Peter Möller
- Institute of Pathology, University Hospital, Ulm, Germany
| | - Michael Gröger
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Sandra Kress
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Marina Fink
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Britta Lukaschewski
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Grégoire Rumm
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Bettina Stahl
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital, Ulm, Germany
| | | | - Matthew Whiteman
- University of Exeter Medical School, St. Luke's Campus, Exeter, England, UK
| | - Oscar McCook
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Clair Hartmann
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
- Department of Anesthesiology, University Hospital, Ulm, Germany
| |
Collapse
|
26
|
Qu W, Niu C, Zhang X, Chen W, Yu F, Liu H, Zhang X, Wang S. Construction of a novel far-red fluorescence light-up probe for visualizing intracellular peroxynitrite. Talanta 2019; 197:431-435. [DOI: 10.1016/j.talanta.2019.01.065] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/04/2019] [Accepted: 01/16/2019] [Indexed: 10/27/2022]
|
27
|
Salomão R, Ferreira B, Salomão M, Santos S, Azevedo L, Brunialti M. Sepsis: evolving concepts and challenges. Braz J Med Biol Res 2019; 52:e8595. [PMID: 30994733 PMCID: PMC6472937 DOI: 10.1590/1414-431x20198595] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
Sepsis remains a major cause of morbidity and mortality worldwide, with increased burden in low- and middle-resource settings. The role of the inflammatory response in the pathogenesis of the syndrome has supported the modern concept of sepsis. Nevertheless, a definition of sepsis and the criteria for its recognition is a continuous process, which reflects the growing knowledge of its mechanisms and the success and failure of diagnostic and therapeutic interventions. Here we review the evolving concepts of sepsis, from the "systemic inflammatory response syndrome triggered by infection" (Sepsis-1) to "a severe, potentially fatal, organic dysfunction caused by an inadequate or dysregulated host response to infection" (Sepsis-3). We focused in the pathophysiology behind the concept and the criteria for recognition and diagnosis of sepsis. A major challenge in evaluating the host response in sepsis is to characterize what is protective and what is harmful, and we discuss that, at least in part, the apparent dysregulated host response may be an effort to adapt to a hostile environment. The new criteria for recognition and diagnosis of sepsis were derived from robust databases, restricted, however, to developed countries. Since then, the criteria have been supported in different clinical settings and in different economic and epidemiological contexts, but still raise discussion regarding their use for the identification versus the prognostication of the septic patient. Clinicians should not be restricted to definition criteria when evaluating patients with infection and should wisely use the broad array of information obtained by rigorous clinical observation.
Collapse
Affiliation(s)
- R. Salomão
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - B.L. Ferreira
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - M.C. Salomão
- Departamento de Moléstias Infecciosas e Parasitárias Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - S.S. Santos
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L.C.P. Azevedo
- Unidade de Terapia Intensiva do Hospital Sírio Libanês, São Paulo, SP, Brasil
| | - M.K.C. Brunialti
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
28
|
Discovery of 2-Substituted 3-Arylquinoline Derivatives as Potential Anti-Inflammatory Agents Through Inhibition of LPS-Induced Inflammatory Responses in Macrophages. Molecules 2019; 24:molecules24061162. [PMID: 30909606 PMCID: PMC6472047 DOI: 10.3390/molecules24061162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022] Open
Abstract
We describe herein the preparation of certain 2-substituted 3-arylquinoline derivatives and the evaluation of their anti-inflammatory effects in LPS-activated murine J774A.1 macrophage cells. Among these newly synthesized 2-substituted 3-arylquinoline derivatives, 2-(4-methoxy- benzoyl)-3-(3,4,5-trimethoxyphenyl)quinoline (18a) and 2-(4-fluorobenzoyl)-3-(3,4,5-trimethoxy- phenyl)quinoline (18b) are two of the most active compounds which can inhibit the production of NO at non-cytotoxic concentrations. Our results have also indicated that compounds 18a and 18b significantly decrease the secretion of pro-inflammatory cytokines (TNF-á and IL-6), inhibit the expression of iNOS, suppress the phosphorylation of MAPKs, and attenuate the activity of NF-êB by LPS-activated macrophages. Through molecular docking analysis, we found that 18b could fit into the middle of the TNF-á dimer and form hydrophobic interactions with Leu55, Leu57 chain A and B, Tyr59, Val123 chain B and D, Ile 155. These results suggest that both 18a and 18b are potential lead compounds in inhibiting LPS-induced inflammatory responses. Further structural optimization to discover novel anti-inflammatory agents is ongoing.
Collapse
|
29
|
Hemorrhagic blood failure: Oxygen debt, coagulopathy, and endothelial damage. J Trauma Acute Care Surg 2019; 82:S41-S49. [PMID: 28328671 DOI: 10.1097/ta.0000000000001436] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Liu F, Dong H, Tian Y. Real-time monitoring of peroxynitrite (ONOO−) in the rat brain by developing a ratiometric electrochemical biosensor. Analyst 2019; 144:2150-2157. [DOI: 10.1039/c9an00079h] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a reactive oxygen species (ROS), peroxynitrite (ONOO−) generated by nitric oxide (NO) and superoxide anion (O2˙−) plays important roles in physiological and pathological processes in the brain.
Collapse
Affiliation(s)
- Feiyue Liu
- Shanghai State Key Laboratory of Green Chemistry and Chemical Processes
- Department of Chemistry
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| | - Hui Dong
- Shanghai State Key Laboratory of Green Chemistry and Chemical Processes
- Department of Chemistry
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| | - Yang Tian
- Shanghai State Key Laboratory of Green Chemistry and Chemical Processes
- Department of Chemistry
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| |
Collapse
|
31
|
Alcamo AM, Pang D, Bashir DA, Carcillo JA, Nguyen TC, Aneja RK. Role of Damage-Associated Molecular Patterns and Uncontrolled Inflammation in Pediatric Sepsis-Induced Multiple Organ Dysfunction Syndrome. J Pediatr Intensive Care 2018; 8:25-31. [PMID: 31073505 DOI: 10.1055/s-0038-1675639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/19/2018] [Indexed: 01/20/2023] Open
Abstract
The incidence of multiple organ dysfunction syndrome (MODS) in sepsis varies from 17 to 73% and furthermore, increases the risk of death by 60% when controlled for the number of dysfunctional organs. Several MODS phenotypes exist, each unique in presentation and pathophysiology. Common to the phenotypes is the stimulation of the immune response by pathogen-associated molecular patterns (PAMPs), or danger-associated molecular patterns (DAMPs) causing an unremitting inflammation. Two of the MODS phenotypes are discussed in detail, thrombocytopenia-associated multiple organ failure (TAMOF) and the hyperinflammatory phenotype-macrophage activating syndrome (MAS) and hemophagocytic lymphohistiocytosis (HLH). In the end, we will briefly review the role of mitochondrial dysfunction as a significant contributor to the pathogenesis of MODS.
Collapse
Affiliation(s)
- Alicia M Alcamo
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Diana Pang
- Department of Critical Care Medicine, Children's Hospital of the King's Daughters, Norfolk, Virginia, United States
| | - Dalia A Bashir
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, United States.,Michael E. DeBakey Veteran Affairs Medical Center, Center for Translational Research on Inflammatory Diseases, Houston, Texas, United States
| | - Joseph A Carcillo
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Trung C Nguyen
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, United States.,Michael E. DeBakey Veteran Affairs Medical Center, Center for Translational Research on Inflammatory Diseases, Houston, Texas, United States
| | - Rajesh K Aneja
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
32
|
Shen X, Han G, Li S, Song Y, Shen H, Zhai Y, Wang Y, Zhang F, Dong N, Li T, Yao Y, Zhu H. Association between the T6459C point mutation of the mitochondrial MT-CO1 gene and susceptibility to sepsis among Chinese Han people. J Cell Mol Med 2018; 22:5257-5264. [PMID: 30207067 PMCID: PMC6201344 DOI: 10.1111/jcmm.13746] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 05/18/2018] [Indexed: 11/29/2022] Open
Abstract
To search for an association between sepsis and mitochondrial genetic basis, we began our study. In this study, a proband harbouring mitochondrial T6459C mutation with sepsis and his Chinese Han pedigree including 7 members of 3 generations were enrolled. General information, blood parameters and mitochondrial full sequence scanning of all members were performed, and cellular functions, including cellular reactive oxygen species (ROS) levels, mitochondrial membrane potential (MMP), degrees of cell apoptosis and adenosine triphosphate (ATP) concentrations, were measured in members with and without the T6459C mutation. Through mitochondrial full sequence scanning and analysis of all members we found, the maternal members (I-1, II-1, II-2 and II-4) in this Chinese Han pedigree all had the mitochondrial T6459C mutation and were used as the mutation group. The non-maternal members (II-3, III-1 and III-2) did not have this mutation and were used as the non-mutation group. The differences in all indicators, including the blood routine, blood biochemistry and coagulation function tests, between members in these two groups were not significant. Under the non-stimulation condition, the mutation group had higher ROS levels (4210.42 ± 1043.35 vs 3387.78 ± 489.66, P = .028) and apoptosis ratios (P = .004) and lower ATP concentrations (P = .049) and MMP levels (P = .047) than the non-mutation group. After 6 hours of simulated LPS stimulation, the mutation group had significantly increased ROS levels (5759.25 ± 2297.90 vs 3862.00 ± 1519.77, P = .045) compared with the non-mutation group, whereas the mutation group continued to demonstrate higher ROS levels (P = .045) and apoptosis ratios (P = .003) and lower MMP levels (P = .005) and ATP concentrations (P = .010). We speculated that the mtDNA T6459C mutation might be the basis for the genetic susceptibility to sepsis.
Collapse
Affiliation(s)
- Xiaodong Shen
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Guoxin Han
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Shuoshuo Li
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Yang Song
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Hong Shen
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Yongzhi Zhai
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Yingchan Wang
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Fei Zhang
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Ning Dong
- Trauma Research CenterFirst Hospital Affiliated to the Chinese PLA General HospitalBeijingChina
| | - Tanshi Li
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| | - Yongming Yao
- Trauma Research CenterFirst Hospital Affiliated to the Chinese PLA General HospitalBeijingChina
| | - Haiyan Zhu
- Emergency DepartmentChinese PLA General HospitalBeijingChina
| |
Collapse
|
33
|
Abstract
This pilot study was conducted to profile safety of nebulized racemic epinephrine when used as a therapy for smoke inhalation injury in severely burned children. We enrolled 16 patients who were 7 to 19 years of age ([mean ± SD], 12 ± 4 years) with burns covering more than 30% of the TBSA (55 ± 17%) and smoke inhalation injury, as diagnosed by bronchoscopy at burn center admission. Patients were randomized to receive either standard of care (n = 8), which consisted of nebulized acetylcysteine, nebulized heparin, and nebulized albuterol, or to receive standard of care plus nebulized epinephrine (n = 8). Primary endpoints were death, chest pain, and adverse changes in cardiopulmonary hemodynamics (arrhythmia, arterial blood pressure, electrocardiographic [ST segment] changes, and peak inspiratory pressure). Additional endpoints included total days on ventilator, pulmonary function, and physiological cardiopulmonary measurements at intensive care unit discharge. No adverse events were observed during or after the nebulization of epinephrine, and no deaths were reported that were attributable to the administration of nebulized epinephrine. The groups did not significantly differ with regard to age, sex, burn size, days on ventilator, pulmonary function, or cardiopulmonary fitness. Results of this pilot trial indicate epinephrine to be safe when administered to pediatric burn patients with smoke inhalation injury. Current data warrant future efficacy studies with a greater number of patients.
Collapse
|
34
|
Islam BU, Habib S, Ali SA, Moinuddin, Ali A. Role of Peroxynitrite-Induced Activation of Poly(ADP-Ribose) Polymerase (PARP) in Circulatory Shock and Related Pathological Conditions. Cardiovasc Toxicol 2018; 17:373-383. [PMID: 27990620 DOI: 10.1007/s12012-016-9394-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peroxynitrite is a powerful oxidant, formed from the reaction of nitric oxide and superoxide. It is known to interact and modify different biological molecules such as DNA, lipids and proteins leading to alterations in their structure and functions. These events elicit various cellular responses, including cell signaling, causing oxidative damage and committing cells to apoptosis or necrosis. This review discusses nitrosative stress-induced modification in the DNA molecule that results in the formation of 8-nitroguanine and 8-oxoguanine, and its role in disease conditions. Different approaches of cell death, such as necrosis and apoptosis, are modulated by cellular high-energy species, such as ATP and NAD+. High concentrations of peroxynitrite are known to cause necrosis, whereas low concentrations lead to apoptosis. Any damage to DNA activates cellular DNA repair machinery, like poly(ADP-ribose) polymerase (PARP). PARP-1, an isoform of PARP, is a DNA nick-sensing enzyme that becomes activated upon sensing DNA breakage and triggers the cleavage of NAD+ into nicotinamide and ADP-ribose and polymerizes the latter on nuclear acceptor proteins. Peroxynitrite-induced hyperactivation of PARP causes depletion of NAD+ and ATP culminating cell dysfunction, necrosis or apoptosis. This mechanistic pathway is implicated in the pathogenesis of a variety of diseases, including circulatory shock (which is characterized by cellular hypoxia triggered by systemic altered perfusion and tissue oxygen utilization leading end-organ dysfunction), sepsis and inflammation, injuries of the lung and the intestine. The cytotoxic effects of peroxynitrite centering on the participation of PARP-1 and ADP-ribose in previously stated diseases have also been discussed in this review.
Collapse
Affiliation(s)
- Badar Ul Islam
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Safia Habib
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Syed Amaan Ali
- Kothiwal Dental College and Research Center, Moradabad, UP, India
| | - Moinuddin
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Asif Ali
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India.
| |
Collapse
|
35
|
Ito H, Malgerud E, Asmussen S, Lopez E, Salzman AL, Enkhbaatar P. R-100 improves pulmonary function and systemic fluid balance in sheep with combined smoke-inhalation injury and Pseudomonas aeruginosa sepsis. J Transl Med 2017; 15:266. [PMID: 29282084 PMCID: PMC5745620 DOI: 10.1186/s12967-017-1366-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 12/12/2017] [Indexed: 11/30/2022] Open
Abstract
Background Septic shock is a major cause of death in intensive care units around the world . The aim of the study was to investigate whether the novel drug R-100 (a superoxide degradation catalyst and nitric oxide donor) improves pulmonary function in a sheep model of septic shock caused by Pseudomonas aeruginosa and smoke inhalation. Methods Eleven female sheep were prepared surgically and randomly assigned to a treatment group (n = 5) or a control group (n = 6) after inhalation of cooled cotton smoke and airway instillation of live P. aeruginosa (2.5 × 1011 CFU) by bronchoscope under deep anesthesia and analgesia. The treatment group received an intravenous infusion of a total of 80 mg/kg of R-100 diluted in 500 mL of 5% dextrose. The control group was given 500 mL of 5% dextrose. All animals received intravenous lactated Ringer’s solution to maintain a hematocrit level at baseline ± 3%. Blood gas and hemodynamics were measured at baseline and then analyzed every 3 h during the 24-h study period. Results are expressed as mean ± SEM. Results The treated animals showed significant improvement in their pulmonary gas exchange (PaO2/FiO2 ratio at 24 h: 246 ± 29 vs. 90 ± 40 mmHg control, P < 0.05). Pulmonary arterial pressures were reduced in the treated group (24 h: 26 ± 1 vs. 30 ± 2 cm mmHg control, P < 0.05). The treated animals also had an improved total fluid balance after 24 h (190 ± 45/24 h mL vs. 595 ± 234/24 h mL control, P < 0.05). Conclusions Treatment with R-100 improves pulmonary gas exchange and blood oxygenation, and prevents a fluid imbalance in sheep subjected to smoke inhalation and P. aeruginosa.
Collapse
Affiliation(s)
- Hiroshi Ito
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Erik Malgerud
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Sven Asmussen
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Ernesto Lopez
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | | | - Perenlei Enkhbaatar
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
36
|
Sharma NK, Tashima AK, Brunialti MKC, Ferreira ER, Torquato RJS, Mortara RA, Machado FR, Assuncao M, Rigato O, Salomao R. Proteomic study revealed cellular assembly and lipid metabolism dysregulation in sepsis secondary to community-acquired pneumonia. Sci Rep 2017; 7:15606. [PMID: 29142235 PMCID: PMC5688086 DOI: 10.1038/s41598-017-15755-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/01/2017] [Indexed: 12/21/2022] Open
Abstract
Sepsis is a life-threatening disorder characterized by organ dysfunction and a major cause of mortality worldwide. The major challenge in studying sepsis is its diversity in such factors as age, source of infection and etiology. Recently, genomic and proteomic approaches have improved our understanding of its complex pathogenesis. In the present study, we use quantitative proteomics to evaluate the host proteome response in septic patients secondary to community-acquired pneumonia (CAP). Samples obtained at admission and after 7 days of follow-up were analyzed according to the outcomes of septic patients. The patients' proteome profiles were compared with age- and gender-matched healthy volunteers. Bioinformatic analyses of differentially expressed proteins showed alteration in the cytoskeleton, cellular assembly, movement, lipid metabolism and immune responses in septic patients. Actin and gelsolin changes were assessed in mononuclear cells using immunofluorescence, and a higher expression of gelsolin and depletion of actin were observed in survivor patients. Regarding lipid metabolism, changes in cholesterol, HDL and apolipoproteins were confirmed using enzymatic colorimetric methods in plasma. Transcriptomic studies revealed a massive change in gene expression in sepsis. Our proteomic results stressed important changes in cellular structure and metabolism, which are possible targets for future interventions of sepsis.
Collapse
Affiliation(s)
- Narendra Kumar Sharma
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil
| | - Alexandre Keiji Tashima
- Departamento de Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, 04023-900, Brazil
| | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil
| | - Eden Ramalho Ferreira
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, 04023-062, Brazil
| | - Ricardo Jose Soares Torquato
- Departamento de Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, 04023-900, Brazil
| | - Renato Arruda Mortara
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, 04023-062, Brazil
| | - Flavia Ribeiro Machado
- Intensive Care Unit, Hospital São Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, 04024-002, Brazil
| | - Murillo Assuncao
- Intensive Care Unit, Hospital Israelita Albert Einstein, Sao Paulo, 05652- 900, Brazil
| | - Otelo Rigato
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil
- Intensive Care Unit, Hospital Sirio Libanes, Sao Paulo, 01409-001, Brazil
| | - Reinaldo Salomao
- Division of Infectious Diseases, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de Sao Paulo, Sao Paulo, 04039-032, Brazil.
| |
Collapse
|
37
|
Okuyama T, Nakatake R, Kaibori M, Okumura T, Kon M, Nishizawa M. A sense oligonucleotide to inducible nitric oxide synthase mRNA increases the survival rate of rats in septic shock. Nitric Oxide 2017; 72:32-40. [PMID: 29128398 DOI: 10.1016/j.niox.2017.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 10/04/2017] [Accepted: 11/07/2017] [Indexed: 12/28/2022]
Abstract
Natural antisense transcripts (asRNAs) that do not encode proteins are transcribed from rat, mouse, and human genes, encoding inducible nitric oxide synthase (iNOS), which catalyzes the production of the inflammatory mediator nitric oxide (NO). In septic shock, NO is excessively produced in hepatocytes and macrophages. The iNOS asRNA interacts with and stabilizes iNOS mRNA. We found that single-stranded 'sense' oligonucleotides corresponding to the iNOS mRNA sequence reduced iNOS mRNA levels by interfering with the mRNA-asRNA interactions in rat hepatocytes. The iNOS sense oligonucleotides that were substituted with phosphorothioate bonds and locked nucleic acids efficiently decreased the levels of iNOS mRNA and iNOS protein. In this study, the gene expression patterns in the livers of two endotoxemia model rats with acute liver failure were compared. Next, we optimized the sequence and modification of the iNOS sense oligonucleotides in interleukin 1β-treated rat hepatocytes. When a sense oligonucleotide was simultaneously administered with d-galactosamine and bacterial lipopolysaccharide (LPS) to rats, their survival rate significantly increased compared to the rats administered d-galactosamine and LPS alone. In the livers of the sense oligonucleotide-administered rats, apoptosis in the hepatocytes markedly decreased. These results suggest that natural antisense transcript-targeted regulation technology using iNOS sense oligonucleotides may be used to treat human inflammatory diseases, such as sepsis and septic shock.
Collapse
Affiliation(s)
- Tetsuya Okuyama
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Richi Nakatake
- Department of Surgery, Kansai Medical University, Hirakata, Osaka, Japan
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, Hirakata, Osaka, Japan
| | - Tadayoshi Okumura
- Department of Surgery, Kansai Medical University, Hirakata, Osaka, Japan; Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Masanori Kon
- Department of Surgery, Kansai Medical University, Hirakata, Osaka, Japan
| | - Mikio Nishizawa
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan.
| |
Collapse
|
38
|
Frydrych LM, Fattahi F, He K, Ward PA, Delano MJ. Diabetes and Sepsis: Risk, Recurrence, and Ruination. Front Endocrinol (Lausanne) 2017; 8:271. [PMID: 29163354 PMCID: PMC5670360 DOI: 10.3389/fendo.2017.00271] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/27/2017] [Indexed: 12/16/2022] Open
Abstract
Sepsis develops when an infection surpasses local tissue containment. A series of dysregulated physiological responses are generated, leading to organ dysfunction and a 10% mortality risk. When patients with sepsis demonstrate elevated serum lactates and require vasopressor therapy to maintain adequate blood pressure in the absence of hypovolemia, they are in septic shock with an in-hospital mortality rate >40%. With improvements in intensive care treatment strategies, overall sepsis mortality has diminished to ~20% at 30 days; however, mortality continues to steadily climb after recovery from the acute event. Traditionally, it was thought that the complex interplay between inflammatory and anti-inflammatory responses led to sepsis-induced organ dysfunction and mortality. However, a closer examination of those who die long after sepsis subsides reveals that many initial survivors succumb to recurrent, nosocomial, and secondary infections. The comorbidly challenged, physiologically frail diabetic individuals suffer the highest infection rates. Recent reports suggest that even after clinical "recovery" from sepsis, persistent alterations in innate and adaptive immune responses exists resulting in chronic inflammation, immune suppression, and bacterial persistence. As sepsis-associated immune defects are associated with increased mortality long-term, a potential exists for immune modulatory therapy to improve patient outcomes. We propose that diabetes causes a functional immune deficiency that directly reduces immune cell function. As a result, patients display diminished bactericidal clearance, increased infectious complications, and protracted sepsis mortality. Considering the substantial expansion of the elderly and obese population, global adoption of a Western diet and lifestyle, and multidrug resistant bacterial emergence and persistence, diabetic mortality from sepsis is predicted to rise dramatically over the next two decades. A better understanding of the underlying diabetic-induced immune cell defects that persist following sepsis are crucial to identify potential therapeutic targets to bolster innate and adaptive immune function, prevent infectious complications, and provide more durable diabetic survival.
Collapse
Affiliation(s)
- Lynn M. Frydrych
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Fatemeh Fattahi
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Katherine He
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Peter A. Ward
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Matthew J. Delano
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
39
|
Nitrite administration improves sepsis-induced myocardial and mitochondrial dysfunction by modulating stress signal responses. J Anesth 2017; 31:885-894. [PMID: 29063286 DOI: 10.1007/s00540-017-2417-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE A specific therapeutic strategy in sepsis-induced myocardial dysfunction remains to be determined. Nitrite may have cardioprotective effects against sepsis-induced myocardial dysfunction. This study investigated the cardioprotective effects of nitrite on myocardial function, mitochondrial bioenergetics, and its underlying molecular mechanisms in severe septic rats. METHODS Sepsis was induced in male Wistar rats by cecal ligation and puncture (CLP). After CLP, we administered normal saline (NS group) or nitrite (nitrite group) subcutaneously. We administered nitrite at different doses (0.1-10 mg/kg) to ascertain the most effective dose and examined cardiac function in an isolated heart experiment 8 h after CLP. We investigated mitochondrial bioenergetics and molecular mechanisms underlying the administration of nitrite in vitro. RESULTS In isolated heart experiments, the left ventricular developed pressure (96 ± 5 mmHg) at a moderate nitrite dose (1.0 mg/kg) was significantly higher than that in the NS group (75 ± 4 mmHg, P < 0.05). Mitochondrial oxidative phosphorylation in the nitrite group was significantly higher than that in the NS group (P < 0.01). Immunoblotting revealed that nitrite significantly increased the phosphorylation of Akt (P < 0.05) and reduced the nuclear translocation of NF-κB (P < 0.05) compared with the NS group. Nitrite was also shown to improve the rate of survival in severe septic rats (P < 0.001). CONCLUSIONS Our results showed that a moderate nitrite dose improved septic myocardial dysfunction at organ, cellular, and molecular levels via modulation of stress signal responses, which resulted in an improvement in survival.
Collapse
|
40
|
Steven S, Dib M, Roohani S, Kashani F, Münzel T, Daiber A. Time Response of Oxidative/Nitrosative Stress and Inflammation in LPS-Induced Endotoxaemia-A Comparative Study of Mice and Rats. Int J Mol Sci 2017; 18:ijms18102176. [PMID: 29057830 PMCID: PMC5666857 DOI: 10.3390/ijms18102176] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/01/2017] [Accepted: 10/09/2017] [Indexed: 12/22/2022] Open
Abstract
Sepsis is a severe and multifactorial disease with a high mortality rate. It represents a strong inflammatory response to an infection and is associated with vascular inflammation and oxidative/nitrosative stress. Here, we studied the underlying time responses in the widely used lipopolysaccharide (LPS)-induced endotoxaemia model in mice and rats. LPS (10 mg/kg; from Salmonella Typhosa) was intraperitoneally injected into mice and rats. Animals of every species were divided into five groups and sacrificed at specific points in time (0, 3, 6, 9, 12 h). White blood cells (WBC) decreased significantly in both species after 3 h and partially recovered with time, whereas platelet decrease did not recover. Oxidative burst and iNOS-derived nitrosyl-iron hemoglobin (HbNO) increased with time (maxima at 9 or 12 h). Immune cell infiltration (CD68 and F4/80 content) showed an increase with time, which was supported by increased vascular mRNA expression of VCAM-1, P-selectin, IL-6 and TNF-α. We characterized the time responses of vascular inflammation and oxidative/nitrosative stress in LPS-induced endotoxaemic mice and rats. The results of this study will help to interpret and compare data from different animal species in LPS-induced endotoxaemia models for the identification of new drug targets.
Collapse
Affiliation(s)
- Sebastian Steven
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Mobin Dib
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Siyer Roohani
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Fatemeh Kashani
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Thomas Münzel
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Andreas Daiber
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| |
Collapse
|
41
|
Time-Dependent Production of Endothelium-Related Biomarkers is Affected Differently in Hemorrhagic and Septic Shocks. Inflammation 2017; 41:33-41. [PMID: 29019070 DOI: 10.1007/s10753-017-0660-z] [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] [Indexed: 12/29/2022]
Abstract
Shock is associated with inflammation-induced endothelial dysfunction. The aim of this study was to determine time-dependent alteration of blood biomarkers related to endothelial function in hemorrhagic and septic shocks. Hemorrhagic shock was induced by bleeding the animals. A cecal ligation and incision model was used to induce septicemia. Resuscitation was carried out by infusion of lactated Ringer's solution. Resuscitation extended survival time in both shock groups. Blood pressure increased by resuscitation in the hemorrhagic shock but not in the septic shock. While hemorrhage caused a decrease in plasma levels of nitric oxide (NO) and hydrogen sulfide (H2S), asymmetric dimethylarginine (ADMA) and total antioxidant capacity (TAC) levels were increased. Only NO and TAC levels at the late phase were reversed by resuscitation. On the other hand, plasma levels of NO, ADMA, and TAC were increased by septicemia and resuscitation did not alter the septicemia-induced increase. These results indicate that blood biomarkers related to endothelial function were differentially affected by hemorrhage and septicemia. The time scale of biomarker production should be taken into consideration for the diagnostic and therapeutic approaches to these life-threatening diseases.
Collapse
|
42
|
Delport A, Harvey BH, Petzer A, Petzer JP. Methylene blue and its analogues as antidepressant compounds. Metab Brain Dis 2017; 32:1357-1382. [PMID: 28762173 DOI: 10.1007/s11011-017-0081-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022]
Abstract
Methylene Blue (MB) is considered to have diverse medical applications and is a well-described treatment for methemoglobinemias and ifosfamide-induced encephalopathy. In recent years the focus has shifted to MB as an antimalarial agent and as a potential treatment for neurodegenerative disorders such as Alzheimer's disease. Of interest are reports that MB possesses antidepressant and anxiolytic activity in pre-clinical models and has shown promise in clinical trials for schizophrenia and bipolar disorder. MB is a noteworthy inhibitor of monoamine oxidase A (MAO-A), which is a well-established target for antidepressant action. MB is also recognized as a non-selective inhibitor of nitric oxide synthase (NOS) and guanylate cyclase. Dysfunction of the nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) cascade is strongly linked to the neurobiology of mood, anxiety and psychosis, while the inhibition of NOS and/or guanylate cyclase has been associated with an antidepressant response. This action of MB may contribute significantly to its psychotropic activity. However, these disorders are also characterised by mitochondrial dysfunction and redox imbalance. By acting as an alternative electron acceptor/donor MB restores mitochondrial function, improves neuronal energy production and inhibits the formation of superoxide, effects that also may contribute to its therapeutic activity. Using MB in depression co-morbid with neurodegenerative disorders, like Alzheimer's and Parkinson's disease, also represents a particularly relevant strategy. By considering their physicochemical and pharmacokinetic properties, analogues of MB may provide therapeutic potential as novel multi-target strategies in the treatment of depression. In addition, low MAO-A active analogues may provide equal or improved response with a lower risk of adverse effects.
Collapse
Affiliation(s)
- Anzelle Delport
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
- Division of Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Brian H Harvey
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
- Division of Pharmacology, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Anél Petzer
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
- Division of Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Jacobus P Petzer
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
- Division of Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
| |
Collapse
|
43
|
Delano MJ, Ward PA. The immune system's role in sepsis progression, resolution, and long-term outcome. Immunol Rev 2017; 274:330-353. [PMID: 27782333 DOI: 10.1111/imr.12499] [Citation(s) in RCA: 477] [Impact Index Per Article: 68.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis occurs when an infection exceeds local tissue containment and induces a series of dysregulated physiologic responses that result in organ dysfunction. A subset of patients with sepsis progress to septic shock, defined by profound circulatory, cellular, and metabolic abnormalities, and associated with a greater mortality. Historically, sepsis-induced organ dysfunction and lethality were attributed to the complex interplay between the initial inflammatory and later anti-inflammatory responses. With advances in intensive care medicine and goal-directed interventions, early 30-day sepsis mortality has diminished, only to steadily escalate long after "recovery" from acute events. As so many sepsis survivors succumb later to persistent, recurrent, nosocomial, and secondary infections, many investigators have turned their attention to the long-term sepsis-induced alterations in cellular immune function. Sepsis clearly alters the innate and adaptive immune responses for sustained periods of time after clinical recovery, with immune suppression, chronic inflammation, and persistence of bacterial representing such alterations. Understanding that sepsis-associated immune cell defects correlate with long-term mortality, more investigations have centered on the potential for immune modulatory therapy to improve long-term patient outcomes. These efforts are focused on more clearly defining and effectively reversing the persistent immune cell dysfunction associated with long-term sepsis mortality.
Collapse
Affiliation(s)
- Matthew J Delano
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
44
|
Kumanto M, Paukkeri EL, Nieminen R, Moilanen E. Cobalt(II) Chloride Modifies the Phenotype of Macrophage Activation. Basic Clin Pharmacol Toxicol 2017; 121:98-105. [DOI: 10.1111/bcpt.12773] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 02/22/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Mona Kumanto
- The Immunopharmacology Research Group; Faculty of Medicine and Life Sciences; University of Tampere and Tampere University Hospital; Tampere Finland
| | - Erja-Leena Paukkeri
- The Immunopharmacology Research Group; Faculty of Medicine and Life Sciences; University of Tampere and Tampere University Hospital; Tampere Finland
| | - Riina Nieminen
- The Immunopharmacology Research Group; Faculty of Medicine and Life Sciences; University of Tampere and Tampere University Hospital; Tampere Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group; Faculty of Medicine and Life Sciences; University of Tampere and Tampere University Hospital; Tampere Finland
| |
Collapse
|
45
|
Berger NA, Besson VC, Boulares AH, Bürkle A, Chiarugi A, Clark RS, Curtin NJ, Cuzzocrea S, Dawson TM, Dawson VL, Haskó G, Liaudet L, Moroni F, Pacher P, Radermacher P, Salzman AL, Snyder SH, Soriano FG, Strosznajder RP, Sümegi B, Swanson RA, Szabo C. Opportunities for the repurposing of PARP inhibitors for the therapy of non-oncological diseases. Br J Pharmacol 2017; 175:192-222. [PMID: 28213892 DOI: 10.1111/bph.13748] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 12/12/2022] Open
Abstract
The recent clinical availability of the PARP inhibitor olaparib (Lynparza) opens the door for potential therapeutic repurposing for non-oncological indications. Considering (a) the preclinical efficacy data with PARP inhibitors in non-oncological diseases and (b) the risk-benefit ratio of treating patients with a compound that inhibits an enzyme that has physiological roles in the regulation of DNA repair, we have selected indications, where (a) the severity of the disease is high, (b) the available therapeutic options are limited, and (c) the duration of PARP inhibitor administration could be short, to provide first-line options for therapeutic repurposing. These indications are as follows: acute ischaemic stroke; traumatic brain injury; septic shock; acute pancreatitis; and severe asthma and severe acute lung injury. In addition, chronic, devastating diseases, where alternative therapeutic options cannot halt disease development (e.g. Parkinson's disease, progressive multiple sclerosis or severe fibrotic diseases), should also be considered. We present a preclinical and clinical action plan for the repurposing of PARP inhibitors. LINKED ARTICLES This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.
Collapse
Affiliation(s)
- Nathan A Berger
- Center for Science, Health and Society, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Valerie C Besson
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - A Hamid Boulares
- The Stanley Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Constance, Germany
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, Headache Center - University Hospital, University of Florence, Florence, Italy
| | - Robert S Clark
- Department of Critical Care Medicine and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicola J Curtin
- Newcastle University, Northern Institute for Cancer Research, Medical School, University of Newcastle Upon Tyne, Newcastle Upon Tyne, UK
| | | | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering and Department of Neurology and Department of Pharmacology and Molecular Sciences and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering and Department of Neurology and Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Lucas Liaudet
- Department of Intensive Care Medicine and Burn Center, University Hospital Medical Center, Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Flavio Moroni
- Department of Neuroscience, Università degli Studi di Firenze, Florence, Italy
| | - Pál Pacher
- Laboratory of Physiologic Studies, Section on Oxidative Stress Tissue Injury, NIAAA, NIH, Bethesda, USA
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | | | - Solomon H Snyder
- Department of Neurology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Francisco Garcia Soriano
- Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Robert P Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Balázs Sümegi
- Department of Biochemistry and Medical Chemistry, University of Pécs, Pécs, Hungary
| | - Raymond A Swanson
- Department of Neurology, University of California San Francisco and San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
46
|
Al Drees A, Salah Khalil M, Soliman M. Histological and Immunohistochemical Basis of the Effect of Aminoguanidine on Renal Changes Associated with Hemorrhagic Shock in a Rat Model. Acta Histochem Cytochem 2017; 50:11-19. [PMID: 28386146 PMCID: PMC5374099 DOI: 10.1267/ahc.16025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/01/2016] [Indexed: 12/26/2022] Open
Abstract
Acute kidney failure is the main cause of death among patients with severe trauma due to massive blood loss and hemorrhagic shock (HS). Renal cell injury is caused by tissue ischemia. Renal ischemia initiates a complex and interconnected chain of events resulting in cell injury and renal cell necrosis. Nitric oxide plays a crucial role in renal function and can be inhibited by aminoguanidine (AG). We studied whether AG can ameliorate pathological renal changes associated with HS syndrome in a rat model and explored the AG protection mechanism. Rats were intraperitoneally injected with heparin sodium and mean arterial blood pressure was monitored. Animals were divided into three groups: control (without hemorrhage), with or without intra-arterially injected AG; HS (blood continuously withdrawn or reinfused to maintain an MABP of 35-40 mmHg); and HS with AG. We found that AG decreased plasma concentrations of urea, creatinine, and nitrates; ameliorated histological changes of HS-induced rats; and decreased the expressions of inducible nitrogen oxide synthase (iNOS), proapoptotic protein (BAX), and vitamin D receptors (VDR). AG ameliorated kidney injury by inhibiting iNOS resulting in decreased BAX and VDR expressions. Therefore, a therapeutic strategy targeting AG may provide new insights into kidney injury during severe shock.
Collapse
Affiliation(s)
- Abdulmajeed Al Drees
- Department of Physiology/Department of Medical Education, College of Medicine, King Saud University
| | - Mahmoud Salah Khalil
- College of Medicine, King Saud University
- Department of Histology, Faculty of Medicine, Suez Canal University
| | - Mona Soliman
- Department of Physiology/Department of Medical Education, College of Medicine, King Saud University
| |
Collapse
|
47
|
Expression of genes belonging to the interacting TLR cascades, NADPH-oxidase and mitochondrial oxidative phosphorylation in septic patients. PLoS One 2017; 12:e0172024. [PMID: 28182798 PMCID: PMC5300193 DOI: 10.1371/journal.pone.0172024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/30/2017] [Indexed: 01/05/2023] Open
Abstract
Background and objectives Sepsis is a complex disease that is characterized by activation and inhibition of different cell signaling pathways according to the disease stage. Here, we evaluated genes involved in the TLR signaling pathway, oxidative phosphorylation and oxidative metabolism, aiming to assess their interactions and resulting cell functions and pathways that are disturbed in septic patients. Materials and methods Blood samples were obtained from 16 patients with sepsis secondary to community acquired pneumonia at admission (D0), and after 7 days (D7, N = 10) of therapy. Samples were also collected from 8 healthy volunteers who were matched according to age and gender. Gene expression of 84 genes was performed by real-time polymerase chain reactions. Their expression was considered up- or down-regulated when the fold change was greater than 1.5 compared to the healthy volunteers. A p-value of ≤ 0.05 was considered significant. Results Twenty-two genes were differently expressed in D0 samples; most of them were down-regulated. When gene expression was analyzed according to the outcomes, higher number of altered genes and a higher intensity in the disturbance was observed in non-survivor than in survivor patients. The canonical pathways altered in D0 samples included interferon and iNOS signaling; the role of JAK1, JAK2 and TYK2 in interferon signaling; mitochondrial dysfunction; and superoxide radical degradation pathways. When analyzed according to outcomes, different pathways were disturbed in surviving and non-surviving patients. Mitochondrial dysfunction, oxidative phosphorylation and superoxide radical degradation pathway were among the most altered in non-surviving patients. Conclusion Our data show changes in the expression of genes belonging to the interacting TLR cascades, NADPH-oxidase and oxidative phosphorylation. Importantly, distinct patterns are clearly observed in surviving and non-surviving patients. Interferon signaling, marked by changes in JAK-STAT modulation, had prominent changes in both survivors and non-survivors, whereas the redox imbalance (iNOS signaling, oxidative phosphorylation and superoxide radical degradation) affecting mitochondrial functions was prominent in non-surviving patients.
Collapse
|
48
|
Dumbarton TC, Maxan A, Farah N, Sharawy N, Zhou J, Nantais J, Lehmann C. Tetrahydrobiopterin improves microcirculation in experimental sepsis. Clin Hemorheol Microcirc 2017; 67:15-24. [PMID: 28598830 DOI: 10.3233/ch-160207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Tetrahydrobiopterin (BH4), an endogenous nucleic acid derivative, acts as an important cofactor for several enzymes found within the vascular endothelium, which is deranged in sepsis. OBJECTIVE We hypothesized that BH4 would improve capillary density and decrease inflammation within the intestinal microcirculation of septic rats. METHODS We conducted a randomized, controlled trial using two previously validated models of sepsis in rats: 1) A fecal peritonitis model using a stent perforating the ascending colon, and 2) An endotoxemia model using lipopolysaccharide (LPS) toxin from E. coli. Experimental groups receiving BH4 (60 mg/kg) were compared to otherwise healthy controls and to untreated groups with sepsis-like physiology. RESULTS BH4 decreased leukocyte-endothelial adhesion by 55% and 58% (P < 0.05) in the peritonitis model and endotoxemia models, respectively. In the endotoxemia model but not the peritonitis model, BH4 improved functional capillary density in capillary beds within the intestine (141.3 vs. 106.7 mm/cm2, p < 0.05). Macrohemodynamic parameters were no different between placebo treatment and BH4-treated groups. CONCLUSIONS This study demonstrates that BH4 improves capillary density and inflammation in two separate models of sepsis. BH4 may represent a novel adjunct in the treatment of sepsis and septic shock in clinical practice. Further dose-finding studies and clinical trials are warranted.
Collapse
Affiliation(s)
- Tristan C Dumbarton
- Departments of Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Alexander Maxan
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Nizam Farah
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Nivin Sharawy
- Departments of Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Juan Zhou
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jordan Nantais
- Department of General Surgery, Dalhousie University, Halifax, NS, Canada
| | - Christian Lehmann
- Departments of Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
49
|
Fraga CM, Tomasi CD, Damasio DDC, Vuolo F, Ritter C, Dal-Pizzol F. N-acetylcysteine plus deferoxamine for patients with prolonged hypotension does not decrease acute kidney injury incidence: a double blind, randomized, placebo-controlled trial. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2016; 20:331. [PMID: 27745551 PMCID: PMC5066295 DOI: 10.1186/s13054-016-1504-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/26/2016] [Indexed: 12/30/2022]
Abstract
Background The aim was to test the primary hypothesis that in patients suffering from shock, treatment with N-acetylcysteine (NAC) plus deferoxamine (DFX) decreases the incidence of acute kidney injury (AKI). Methods A double-blind, randomized, placebo-controlled trial was conducted in a general intensive care unit in an academic hospital. Patients were included if they had new-onset hypotension, defined as mean arterial blood pressure <60 mmHg or requirement for vasopressor medication. A loading dose of NAC or placebo of 50 mg/kg in 4 h was administered intravenously. After the loading dose, patients received 100 mg/kg/day for the next 48 h. DFX or placebo was administered once at 1000 mg at a rate of 15/mg/kg/h. The primary outcome was the incidence of AKI. Results A total of 80 patients were enrolled in the study. The incidence of AKI was 67 % in the placebo arm and 65 % in the treatment group (relative risk (RR) 0.89 (0.35–2.2)). Furthermore, NAC plus DFX was effective in decreasing the severity and duration of AKI, and patients in the treatment group had lower serum creatinine levels at discharge. No severe adverse event associated with treatment was reported. The effects of NAC plus DFX could be secondary to the attenuation of early inflammatory response and oxidative damage. Conclusion The administration of NAC plus DFX to critically ill patients who had a new episode of hypotension did not decrease the incidence of AKI. Trial registration Clinicaltrials.gov NCT00870883 (Registered 25 March 2009.)
Collapse
Affiliation(s)
- Cassiana Mazon Fraga
- Pathophysiology Laboratory, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Intensive Care Unit, São José Hospital, Criciúma, SC, Brazil
| | | | - Danusa de Castro Damasio
- Intensive Care Unit, São José Hospital, Criciúma, SC, Brazil.,São José Hospital Research Centre, Criciúma, SC, Brazil
| | - Francieli Vuolo
- Pathophysiology Laboratory, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Cristiane Ritter
- Pathophysiology Laboratory, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Intensive Care Unit, São José Hospital, Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Pathophysiology Laboratory, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil. .,Intensive Care Unit, São José Hospital, Criciúma, SC, Brazil. .,Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Avenida Universitária 1105, 88006-000, Criciúma, SC, Brazil.
| |
Collapse
|
50
|
White NJ, Wang Y, Fu X, Cardenas JC, Martin EJ, Brophy DF, Wade CE, Wang X, St John AE, Lim EB, Stern SA, Ward KR, López JA, Chung D. Post-translational oxidative modification of fibrinogen is associated with coagulopathy after traumatic injury. Free Radic Biol Med 2016; 96:181-9. [PMID: 27105953 PMCID: PMC4912420 DOI: 10.1016/j.freeradbiomed.2016.04.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/13/2016] [Accepted: 04/17/2016] [Indexed: 12/16/2022]
Abstract
Victims of trauma often develop impaired blood clot formation (coagulopathy) that contributes to bleeding and mortality. Fibrin polymerization is one critical component of clot formation that can be impacted by post-translational oxidative modifications of fibrinogen after exposure to oxidants. In vitro evidence suggests that Aα-C domain methionine sulfoxide formation, in particular, can induce conformational changes that prevent lateral aggregation of fibrin protofibrils during polymerization. We used mass spectrometry of plasma from trauma patients to find that fibrinogen Aα-C domain methionine sulfoxide content was selectively-increased in patients with coagulopathy vs. those without coagulopathy. This evidence supports a novel linkage between oxidative stress, coagulopathy, and bleeding after injury.
Collapse
Affiliation(s)
- Nathan J White
- University of Washington Division of Emergency Medicine, Harborview Medical Center, Box 359702, 325 9th Avenue, Seattle, WA 98104, USA; Bloodworks Northwest Research Institute, 1551 Eastlake Avenue E, Seattle, WA 98102, USA.
| | - Yi Wang
- Bloodworks Northwest Research Institute, 1551 Eastlake Avenue E, Seattle, WA 98102, USA
| | - Xiaoyun Fu
- Bloodworks Northwest Research Institute, 1551 Eastlake Avenue E, Seattle, WA 98102, USA
| | - Jessica C Cardenas
- Center for Translational Injury Research and Division of Acute Care Surgery, University of Texas Health Science Center at Houston, 6431 Fannin St. MSB 5.240, Houston, TX 77030, USA
| | - Erika J Martin
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, 410 N 12th Street, P.O. Box 980533, Richmond, VA 23298, USA
| | - Donald F Brophy
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, 410 N 12th Street, P.O. Box 980533, Richmond, VA 23298, USA
| | - Charles E Wade
- Center for Translational Injury Research and Division of Acute Care Surgery, University of Texas Health Science Center at Houston, 6431 Fannin St. MSB 5.240, Houston, TX 77030, USA
| | - Xu Wang
- University of Washington Division of Emergency Medicine, Harborview Medical Center, Box 359702, 325 9th Avenue, Seattle, WA 98104, USA
| | - Alexander E St John
- University of Washington Division of Emergency Medicine, Harborview Medical Center, Box 359702, 325 9th Avenue, Seattle, WA 98104, USA
| | - Esther B Lim
- University of Washington Division of Emergency Medicine, Harborview Medical Center, Box 359702, 325 9th Avenue, Seattle, WA 98104, USA
| | - Susan A Stern
- University of Washington Division of Emergency Medicine, Harborview Medical Center, Box 359702, 325 9th Avenue, Seattle, WA 98104, USA
| | - Kevin R Ward
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Building 10-103A North Campus Research Complex 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - José A López
- Bloodworks Northwest Research Institute, 1551 Eastlake Avenue E, Seattle, WA 98102, USA
| | - Dominic Chung
- Bloodworks Northwest Research Institute, 1551 Eastlake Avenue E, Seattle, WA 98102, USA
| |
Collapse
|