72 results on '"D'Agnillo F"'
Search Results
2. A RED CELL SUBSTITUTE BASED ON CROSSLINKED HEMOGLOBIN-SUPEROXIDE DISMUTASE-CATALASE
- Author
-
D'Agnillo, F. and Chang, T.M.S.
- Published
- 1997
3. Two future generations of blood substitutes based on polyhemoglobin–SOD–catalase and nanoencapsulation
- Author
-
Chang, T.M.S., D’Agnillo, F., Yu, W.P., and Razack, S.
- Published
- 2000
- Full Text
- View/download PDF
4. Hemoglobin-based oxygen carriers: from mechanisms of toxicity and clearance to rational drug design
- Author
-
Buehler, P W, D'Agnillo, F, Schaer, D J, University of Zurich, and Schaer, D J
- Subjects
1313 Molecular Medicine ,1312 Molecular Biology ,610 Medicine & health ,10029 Clinic and Policlinic for Internal Medicine - Published
- 2010
5. Sequestration of extracellular hemoglobin within a haptoglobin complex decreases its hypertensive and oxidative effects in dogs and guinea pigs
- Author
-
Boretti, Felicitas S, Buehler, P W, D'Agnillo, F, Kluge, K, Glaus, Tony M, Butt, O I, Jia, Y, Goede, J, Pereira, C P, Maggiorini, M, Schoedon, G, Alayash, A I, Schaer, D J, and University of Zurich
- Subjects
10253 Department of Small Animals ,10032 Clinic for Oncology and Hematology ,610 Medicine & health ,2700 General Medicine ,10029 Clinic and Policlinic for Internal Medicine - Published
- 2009
6. Oxyhemoglobin and Apoptosis
- Author
-
Alayash Ai and D'Agnillo F
- Subjects
Apoptosis ,business.industry ,Cancer research ,Medicine ,business - Published
- 2000
- Full Text
- View/download PDF
7. Sequestration of extracellular hemoglobin within a haptoglobin complex decreases its hypertensive and oxidative effects in dogs and guinea pigs
- Author
-
Boretti, Felicitas S; https://orcid.org/0000-0001-6793-8464, Buehler, P W, D'Agnillo, F, Kluge, K, Glaus, Tony M; https://orcid.org/0000-0003-2456-9536, Butt, O I, Jia, Y, Goede, J, Pereira, C P, Maggiorini, M, Schoedon, G, Alayash, A I, Schaer, D J, Boretti, Felicitas S; https://orcid.org/0000-0001-6793-8464, Buehler, P W, D'Agnillo, F, Kluge, K, Glaus, Tony M; https://orcid.org/0000-0003-2456-9536, Butt, O I, Jia, Y, Goede, J, Pereira, C P, Maggiorini, M, Schoedon, G, Alayash, A I, and Schaer, D J
- Abstract
Release of hemoglobin (Hb) into the circulation is a central pathophysiologic event that contributes to morbidity and mortality in chronic hemolytic anemias and severe malaria. These toxicities arise from Hb-mediated vasoactivity, possibly due to NO scavenging and localized tissue oxidative processes. Currently, there is no established treatment that targets circulating extracellular Hb. Here, we assessed the role of haptoglobin (Hp), the primary scavenger of Hb in the circulation, in limiting the toxicity of cell-free Hb infusion. Using a canine model, we found that glucocorticoid stimulation of endogenous Hp synthesis prevented Hb-induced hemodynamic responses. Furthermore, guinea pigs administered exogenous Hp displayed decreased Hb-induced hypertension and oxidative toxicity to extravascular environments, such as the proximal tubules of the kidney. The ability of Hp to both attenuate hypertensive responses during Hb exposure and prevent peroxidative toxicity in extravascular compartments was dependent on Hb-Hp complex formation, which likely acts through sequestration of Hb rather than modulation of its NO- and O2-binding characteristics. Our data therefore suggest that therapies involving supplementation of endogenous Hb scavengers may be able to treat complications of acute and chronic hemolysis, as well as counter the adverse effects associated with Hb-based oxygen therapeutics.
- Published
- 2009
8. Absence of Hemoprotein-Associated Free Radical Events Following Oxidant Challenge of Crosslinked HemoglobinâSuperoxide Dismutase Catalase
- Author
-
D'Agnillo, F, primary
- Published
- 1998
- Full Text
- View/download PDF
9. Reduction of Hydroxyl Radical Generation in a Rat Hindlimb Model of Ischemia-Reperfusion Injury Using Crosslinked Hemoglobin-Superoxide Dismutase-Catalase
- Author
-
D'agnillo, F., primary and Chang, T. M. S., additional
- Published
- 1997
- Full Text
- View/download PDF
10. Crosslinked Hemoglobin-Superoxide Dismutase-Catalase Scavenges Free Radicals in a Rat Model of Intestinal Ischemia-Reperfusion Injury
- Author
-
Razack, Saleem, primary, D'agnillo, F., additional, and Chang, T. M. S., additional
- Published
- 1997
- Full Text
- View/download PDF
11. Cross-Linked Hemoglobin-Superoxide Dismutase-Catalase Scavenges Oxygen-Derived Free Radicals and Prevents Methemoglobin Formation and Iron Release
- Author
-
D'agnillo, F., primary and Chang, Thomas M.S., additional
- Published
- 1993
- Full Text
- View/download PDF
12. Site-specific modifications and toxicity of blood substitutes - The case of diaspirin cross-linked hemoglobin
- Author
-
D'Agnillo, F. and Alayash, A.I.
- Published
- 2000
- Full Text
- View/download PDF
13. Cross-Linked Hemoglobin-Superoxide Dismutase-Catalase Scavenges Oxygen-Derived Free Radicals and Prevents Methemoglobin Formation and Iron Release
- Author
-
D'agnillo, F. and Chang, Thomas
- Abstract
In this study, we prepared PolyHb-SOD-catalase (intermolecularly cross-linked hemoglobin, superoxide dismutase (SOD), and catalase). We found that PolyHb-SOD-catalase is effective in scavenging oxygen-derived free radicals. In the xanthine/xanthine oxidase system, the initial rate of cytochrome c reduction was 2.13 ± 0.26 nmoles cyt. c/min for PolyHb alone. PolyHb-SOD-catalase reduced this to 0.56 ± 0.08 nmoles cyt. c/min because of its ability to eliminate superoxide (O2). Addition of PolyHb to 200 μM of hydrogen peroxide (H2O2), changed the H2O2 level slightly to 192 ± 0.4 μM. Addition of PolyHb-SOD-catalase, on the other hand, lower the level to 41 ± 0.3 μM. Results also show that both effects were dependent on the concentration of SOD-catalase cross-linked with hemoglobin. Oxidative challenge with H2O2 resulted in minimal changes in the absorbance spectra of PolyHb-SOD-catalase. With PolyHb, there were spectral changes reflecting the formation of methemoglobin and heme degradation. Furthermore, the amount of iron released, after incubation with 250 μM H2O2, was 6.8 ± 1.8 μg/dl for PolyHb-SOD-catalase and 76.6 ± 1.0 μg/dl for PolyHb. These results show that cross-linked SOD-catalase prevents oxidative reactions involving the hemoglobin component of PolyHb-SOD-catalase.
- Published
- 1993
- Full Text
- View/download PDF
14. Absence of Hemoprotein-Associated Free Radical Events Following Oxidant Challenge of Crosslinked Hemoglobin-Superoxide Dismutase Catalase
- Author
-
D'Agnillo, F. and Chanc, T. M. S.
- Published
- 1998
- Full Text
- View/download PDF
15. Hemoglobin-based oxygen carriers: from mechanisms of toxicity and clearance to rational drug design
- Author
-
Buehler, P W, D'Agnillo, F, and Schaer, D J
- Subjects
3. Good health
16. Sequestration of extracellular hemoglobin within a haptoglobin complex decreases its hypertensive and oxidative effects in dogs and guinea pigs
- Author
-
Boretti, Felicitas S, Buehler, P W, D'Agnillo, F, Kluge, K, Glaus, Tony M, Butt, O I, Jia, Y, Goede, J, Pereira, C P, Maggiorini, M, Schoedon, G, Alayash, A I, and Schaer, D J
- Subjects
3. Good health
17. Ultraviolet light and riboflavin accelerates red blood cell dysfunction in vitro and in a guinea pig transfusion model.
- Author
-
Baek JH, Shin HKH, Xu F, Zhang X, Williams MC, Gao Y, Vostal JG, Buehler PW, Villa C, and D'Agnillo F
- Subjects
- Animals, Guinea Pigs, Erythrocyte Transfusion, Osmotic Fragility drug effects, Humans, Male, Hemoglobins, Riboflavin pharmacology, Ultraviolet Rays, Erythrocytes drug effects, Erythrocytes metabolism, Hemolysis drug effects
- Abstract
Background: Quality assessment of modified or processed red blood cell (RBC) components, such as pathogen-reduced RBCs, using only in vitro testing may not always be predictive of in vivo performance. Mouse or rat in vivo models are limited by a lack of applicability to certain aspects of human RBC biology. Here, we used a guinea pig model to study the effects of riboflavin combined with UV light on the integrity of RBCs in vitro and following transfusion in vivo., Materials and Methods: Guinea pig RBCs were collected from whole blood (WB) treated with varying UV doses (10, 20, 40 or 80 J/mL) in the presence of riboflavin (UVR-RBCs). In vitro tests for UVR-RBCs included hemolysis, osmotic fragility, and cellular morphology by scanning electron microscopy. Guinea pigs transfused with one-day post-treatment UVR-RBCs were evaluated for plasma hemoglobin (Hb), non-transferrin bound iron (NTBI), total iron and Perls-detectable hemosiderin deposition in the spleen and kidney, and renal uptake of Hb., Results: Acute RBC injury was dose dependently accelerated after treatment with UV light in the presence of riboflavin. Aberrant RBC morphology was evident at 20, 40, and 80 J/mL, and membrane lysis with Hb release was prominent at 80 J/mL. Guinea pigs transfused with 40 and 80 J/mL UVR-RBCs showed increased plasma Hb levels, and plasma NTBI was elevated in all UVR-RBC groups (10-80 J/mL). Total iron levels and Perls-hemosiderin staining in spleen and kidney as well as Hb uptake in renal proximal tubules were increased 8 hours post-transfusion with 40 and 80 J/mL UVR-RBCs., Discussion: UVR-RBCs administered to guinea pigs increased markers of intravascular and extravascular hemolysis in a UV dose-dependent manner. This model may allow for the discrimination of RBC injury during testing of extensively processed RBCs intended for transfusion.
- Published
- 2024
- Full Text
- View/download PDF
18. Renal glomerular and tubular responses to glutaraldehyde- polymerized human hemoglobin.
- Author
-
Williams MC, Zhang X, Baek JH, and D'Agnillo F
- Abstract
Hemoglobin-based oxygen carriers (HBOCs) are being developed as oxygen and volume replacement therapeutics, however, their molecular and cellular effects on the vasculature and different organ systems are not fully defined. Using a guinea pig transfusion model, we examined the renal glomerular and tubular responses to PolyHeme, a highly characterized glutaraldehyde-polymerized human hemoglobin with low tetrameric hemoglobin content. PolyHeme-infused animals showed no major changes in glomerular histology or loss of specific markers of glomerular podocytes (Wilms tumor 1 protein, podocin, and podocalyxin) or endothelial cells (ETS-related gene and claudin-5) after 4, 24, and 72 h. Relative to sham controls, PolyHeme-infused animals also showed similar expression and subcellular distribution of N-cadherin and E-cadherin, two key epithelial junctional proteins of proximal and distal tubules, respectively. In terms of heme catabolism and iron-handling responses, PolyHeme induced a moderate but transient expression of heme oxygenase-1 in proximal tubular epithelium and tubulointerstitial macrophages that was accompanied by increased iron deposition in tubular epithelium. Contrary to previous findings with other modified or acellular hemoglobins, the present data show that PolyHeme does not disrupt the junctional integrity of the renal glomerulus and tubular epithelium, and triggers moderate activation of heme catabolic and iron sequestration systems likely as part of a renal adaptive response., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Williams, Zhang, Baek and D’Agnillo.)
- Published
- 2023
- Full Text
- View/download PDF
19. Intranasal or airborne transmission-mediated delivery of an attenuated SARS-CoV-2 protects Syrian hamsters against new variants.
- Author
-
Stauft CB, Selvaraj P, D'Agnillo F, Meseda CA, Liu S, Pedro CL, Sangare K, Lien CZ, Weir JP, Starost MF, and Wang TT
- Subjects
- Respiratory Aerosols and Droplets, SARS-CoV-2, Mesocricetus, Cricetinae, Spike Glycoprotein, Coronavirus genetics, Mice, Animals, Male, Antibodies, Viral, Vaccinia virus, Antibodies, Neutralizing, COVID-19 prevention & control
- Abstract
Detection of secretory antibodies in the airway is highly desirable when evaluating mucosal protection by vaccines against a respiratory virus, such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We show that intranasal delivery of an attenuated SARS-CoV-2 (Nsp1-K164A/H165A) induces both mucosal and systemic IgA and IgG in male Syrian hamsters. Interestingly, either direct intranasal immunization or airborne transmission-mediated delivery of Nsp1-K164A/H165A in Syrian hamsters offers protection against heterologous challenge with variants of concern (VOCs) including Delta, Omicron BA.1, BA.2.12.1 and BA.5. Vaccinated animals show significant reduction in both tissue viral loads and lung inflammation. Similarly attenuated viruses bearing BA.1 and BA.5 spike boost variant-specific neutralizing antibodies in male mice that were first vaccinated with modified vaccinia virus Ankara vectors (MVA) expressing full-length WA1/2020 Spike protein. Together, these results demonstrate that our attenuated virus may be a promising nasal vaccine candidate for boosting mucosal immunity against future SARS-CoV-2 VOCs., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2023
- Full Text
- View/download PDF
20. HIF-1α-Dependent Metabolic Reprogramming, Oxidative Stress, and Bioenergetic Dysfunction in SARS-CoV-2-Infected Hamsters.
- Author
-
Jana S, Heaven MR, Stauft CB, Wang TT, Williams MC, D'Agnillo F, and Alayash AI
- Subjects
- Cricetinae, Energy Metabolism, Glycolysis, Inflammation, Oxygen, Proteomics, SARS-CoV-2, COVID-19 metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Oxidative Stress
- Abstract
The mechanistic interplay between SARS-CoV-2 infection, inflammation, and oxygen homeostasis is not well defined. Here, we show that the hypoxia-inducible factor (HIF-1α) transcriptional pathway is activated, perhaps due to a lack of oxygen or an accumulation of mitochondrial reactive oxygen species (ROS) in the lungs of adult Syrian hamsters infected with SARS-CoV-2. Prominent nuclear localization of HIF-1α and increased expression of HIF-1α target proteins, including glucose transporter 1 (Glut1), lactate dehydrogenase (LDH), and pyruvate dehydrogenase kinase-1 (PDK1), were observed in areas of lung consolidation filled with infiltrating monocytes/macrophages. Upregulation of these HIF-1α target proteins was accompanied by a rise in glycolysis as measured by extracellular acidification rate (ECAR) in lung homogenates. A concomitant reduction in mitochondrial respiration was also observed as indicated by a partial loss of oxygen consumption rates (OCR) in isolated mitochondrial fractions of SARS-CoV-2-infected hamster lungs. Proteomic analysis further revealed specific deficits in the mitochondrial ATP synthase (Atp5a1) within complex V and in the ATP/ADP translocase (Slc25a4). The activation of HIF-1α in inflammatory macrophages may also drive proinflammatory cytokine production and complement activation and oxidative stress in infected lungs. Together, these findings support a role for HIF-1α as a central mediator of the metabolic reprogramming, inflammation, and bioenergetic dysfunction associated with SARS-CoV-2 infection.
- Published
- 2022
- Full Text
- View/download PDF
21. Active and Passive Immunization of Syrian Hamsters with An Attenuated SARS-CoV-2 Protects against New Variants of Concern.
- Author
-
Wang T, Stauft C, Selvaraj P, D'agnillo F, Meseda C, Sangare K, Pedro C, Liu S, Lien C, Weir J, and Starost M
- Abstract
Detection of secretory antibodies in the airway is highly desirable when evaluating mucosal protection by a vaccine against a respiratory virus like the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We show that a single intranasal delivery of an attenuated SARS-CoV-2 (Nsp1-K164A/H165A) induced both mucosal and systemic IgA and IgG in Syrian hamsters. Interestingly, either active or passive immunization of hamsters with Nsp1-K164A/H165A offered protection against heterologous challenge with variants of concern (VOCs) including Delta, Omicron BA.1, and Omicron BA.2.12.1. Among challenged animals, Nsp1-K164A/H165A vaccination specifically reduced viral loads in the respiratory tract and suppressed infection-induced macrophage accumulation and MX1 upregulation in the lung. The absence of variant-specific mucosal and systemic antibodies was associated with breakthrough infections, particularly of the nasal cavity following challenges with Omicron isolates. Together, our study demonstrates that an attenuated nasal vaccine may be developed to boost mucosal immunity against future SARS-CoV-2 VOCs.
- Published
- 2022
- Full Text
- View/download PDF
22. Intranasal delivery of a rationally attenuated SARS-CoV-2 is immunogenic and protective in Syrian hamsters.
- Author
-
Liu S, Stauft CB, Selvaraj P, Chandrasekaran P, D'Agnillo F, Chou CK, Wu WW, Lien CZ, Meseda CA, Pedro CL, Starost MF, Weir JP, and Wang TT
- Subjects
- Cricetinae, Mice, Animals, Humans, Mesocricetus, Antibody Formation, Administration, Intranasal, COVID-19 Vaccines, Lung pathology, Mice, Transgenic, Spike Glycoprotein, Coronavirus genetics, SARS-CoV-2 genetics, COVID-19 prevention & control
- Abstract
Few live attenuated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are in pre-clinical or clinical development. We seek to attenuate SARS-CoV-2 (isolate WA1/2020) by removing the polybasic insert within the spike protein and the open reading frames (ORFs) 6-8, and by introducing mutations that abolish non-structural protein 1 (Nsp1)-mediated toxicity. The derived virus (WA1-ΔPRRA-ΔORF6-8-Nsp1
K164A/H165A ) replicates to 100- to 1000-fold-lower titers than the ancestral virus and induces little lung pathology in both K18-human ACE2 (hACE2) transgenic mice and Syrian hamsters. Immunofluorescence and transcriptomic analyses of infected hamsters confirm that three-pronged genetic modifications attenuate the proinflammatory pathways more than the removal of the polybasic cleavage site alone. Finally, intranasal administration of just 100 PFU of the WA1-ΔPRRA-ΔORF6-8-Nsp1K164A/H165A elicits robust antibody responses in Syrian hamsters and protects against SARS-CoV-2-induced weight loss and pneumonia. As a proof-of-concept study, we demonstrate that live but sufficiently attenuated SARS-CoV-2 vaccines may be attainable by rational design., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
23. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19.
- Author
-
D'Agnillo F, Walters KA, Xiao Y, Sheng ZM, Scherler K, Park J, Gygli S, Rosas LA, Sadtler K, Kalish H, Blatti CA 3rd, Zhu R, Gatzke L, Bushell C, Memoli MJ, O'Day SJ, Fischer TD, Hammond TC, Lee RC, Cash JC, Powers ME, O'Keefe GE, Butnor KJ, Rapkiewicz AV, Travis WD, Layne SP, Kash JC, and Taubenberger JK
- Subjects
- Cellular Senescence, Fibrinolysis, Humans, Lung, SARS-CoV-2, COVID-19
- Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by respiratory distress, multiorgan dysfunction, and, in some cases, death. The pathological mechanisms underlying COVID-19 respiratory distress and the interplay with aggravating risk factors have not been fully defined. Lung autopsy samples from 18 patients with fatal COVID-19, with symptom onset-to-death times ranging from 3 to 47 days, and antemortem plasma samples from 6 of these cases were evaluated using deep sequencing of SARS-CoV-2 RNA, multiplex plasma protein measurements, and pulmonary gene expression and imaging analyses. Prominent histopathological features in this case series included progressive diffuse alveolar damage with excessive thrombosis and late-onset pulmonary tissue and vascular remodeling. Acute damage at the alveolar-capillary barrier was characterized by the loss of surfactant protein expression with injury to alveolar epithelial cells, endothelial cells, respiratory epithelial basal cells, and defective tissue repair processes. Other key findings included impaired clot fibrinolysis with increased concentrations of plasma and lung plasminogen activator inhibitor-1 and modulation of cellular senescence markers, including p21 and sirtuin-1, in both lung epithelial and endothelial cells. Together, these findings further define the molecular pathological features underlying the pulmonary response to SARS-CoV-2 infection and provide important insights into signaling pathways that may be amenable to therapeutic intervention.
- Published
- 2021
- Full Text
- View/download PDF
24. Structural Integrity of the Alveolar-Capillary Barrier in Cynomolgus Monkeys Challenged with Fully Virulent and Toxin-Deficient Strains of Bacillus anthracis.
- Author
-
D'Agnillo F, Zhang X, and Williams MC
- Subjects
- Animals, Antibodies, Bacterial immunology, Antigens, Bacterial pharmacology, Bacterial Toxins pharmacology, Lung drug effects, Macaca fascicularis immunology, Neutrophils immunology, Spores, Bacterial immunology, Spores, Bacterial pathogenicity, Virulence immunology, Anthrax pathology, Bacillus anthracis pathogenicity, Bacteremia pathology, Lung pathology, Respiratory Tract Infections pathology
- Abstract
Inhalational anthrax, a disease caused by inhaling Bacillus anthracis spores, leads to respiratory distress, vascular leakage, high-level bacteremia, and often death within days. Anthrax lethal toxin and edema toxin, which are composed of protective antigen (PA) plus either lethal factor (LF) or edema factor (EF), respectively, play an important yet incompletely defined role in the pulmonary pathophysiology. To better understand their contribution, we examined the structural integrity of the alveolar-capillary barrier in archival formalin-fixed lungs of cynomolgus monkeys challenged with the fully virulent B. anthracis Ames wild-type strain or the isogenic toxin-deficient mutants ΔEF, ΔLF, and ΔPA. Pulmonary spore challenge with the wild-type strain caused high mortality, intra-alveolar hemorrhages, extensive alveolar septal sequestration of bacteria and neutrophils, diffuse destabilization of epithelial and endothelial junctions, increased markers of coagulation and complement activation (including tissue factor and C5a), and multifocal intra-alveolar fibrin deposition. ΔEF challenge was lethal and showed similar alveolar-capillary alterations; however, intra-alveolar hemorrhages, bacterial deposition, and markers of coagulation or complement were absent or markedly lower. In contrast, ΔLF or ΔPA challenges were nonlethal and showed no signs of alveolar bacterial deposition or alveolar-capillary changes. These findings provide evidence that lethal toxin plays a determinative role in bacterial dissemination and alveolar-capillary barrier dysfunction, and edema toxin may significantly exacerbate pulmonary pathologies in a systemic infection., (Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
25. Reversible renal glomerular dysfunction in guinea pigs exposed to glutaraldehyde-polymerized cell-free hemoglobin.
- Author
-
Zhang X, Williams MC, Rentsendorj O, and D'Agnillo F
- Subjects
- Animals, Guinea Pigs, Kidney Glomerulus physiopathology, Male, Proteinuria chemically induced, Proteinuria pathology, Proteinuria physiopathology, Glutaral toxicity, Hemoglobins toxicity, Kidney Glomerulus drug effects, Kidney Glomerulus pathology, Polymerization, Polymers toxicity
- Abstract
Chemically modified hemoglobin (Hb)-based oxygen carriers are promising oxygen replacement therapeutics however their potential renal effects are not fully understood. Using a guinea pig exchange transfusion model, we examined the effects of glutaraldehyde-polymerized bovine hemoglobin (HbG) on the permeability and integrity of the glomerular filtration barrier (GFB), which is comprised of podocytes, fenestrated endothelium, and the glomerular basement membrane. HbG induced marked proteinuria characterized in part by the loss of high molecular weight proteins, including albumin, immunoglobulin, and transferrin, at 4 and 12 h post-infusion that resolved by 72 h. This correlated with HbG-induced GFB alterations based on the reduced expression of specific markers of podocytes (podocin, nephrin, podocalyxin, and Wilms Tumor 1 protein) and endothelial cells (ETS-related gene and claudin-5). Lectin binding studies also demonstrated marked but reversible alterations to the GFB glycocalyx accompanied by increased intraglomerular HbG deposition and 4-HNE protein adduct expression indicative of oxidative damage. Together, these findings indicate that HbG induces reversible glomerular barrier dysfunction in conjunction with transient GFB changes providing new insight into the renal response to chemically modified Hb therapeutics., (Published by Elsevier B.V.)
- Published
- 2018
- Full Text
- View/download PDF
26. Comprehensive Biochemical and Biophysical Characterization of Hemoglobin-Based Oxygen Carrier Therapeutics: All HBOCs Are Not Created Equally.
- Author
-
Meng F, Kassa T, Jana S, Wood F, Zhang X, Jia Y, D'Agnillo F, and Alayash AI
- Subjects
- Animals, Blood Substitutes adverse effects, Blood Substitutes metabolism, Carbon Monoxide metabolism, Cattle, Cell Line, Heme chemistry, Hemoglobins adverse effects, Hemoglobins genetics, Humans, Kinetics, Mice, Oxidation-Reduction, Oxygen metabolism, Protein Engineering, Blood Substitutes chemistry, Blood Substitutes pharmacology, Hemoglobins chemistry, Hemoglobins pharmacology
- Abstract
The development of hemoglobin (Hb)-based oxygen carriers (HBOCs) has been hampered because of safety concerns in humans. Chemical and/or genetic modifications of the Hb introduce varied structural and conformational constraint on the molecule that resulted in proteins with diverse allosteric responses, nitrosative and oxidative side reactions. Here, we present for the first time a comprehensive biochemical and biophysical comparison of human, bovine, and genetically engineered HBOCs that have been tested in humans. We evaluate oxygen equilibrium and ligand binding kinetics under different experimental conditions as well as their autoxidation kinetics, redox reactions, and heme release. We determined the effects of HBOCs on cellular redox states and mitochondrial respiration. Taken together, these experiments provide a better understanding of the relationship between the structure-function and oxidative reactivity of these proteins. One can therefore select independently among these diverse properties to engineer a safe and effective HBOC with improved biochemical/biophysical characteristics.
- Published
- 2018
- Full Text
- View/download PDF
27. Poly(I:C) Induces Human Lung Endothelial Barrier Dysfunction by Disrupting Tight Junction Expression of Claudin-5.
- Author
-
Huang LY, Stuart C, Takeda K, D'Agnillo F, and Golding B
- Subjects
- Adaptor Proteins, Vesicular Transport metabolism, Chemokines metabolism, Endothelial Cells cytology, Humans, Interferon Regulatory Factor-3 metabolism, NF-kappa B metabolism, Permeability drug effects, Poly I-C metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction drug effects, Toll-Like Receptor 3 metabolism, Claudin-5 genetics, Down-Regulation drug effects, Endothelial Cells metabolism, Lung cytology, Poly I-C pharmacology, Tight Junctions drug effects, Tight Junctions metabolism
- Abstract
Viral infections are often accompanied by pulmonary microvascular leakage and vascular endothelial dysfunction via mechanisms that are not completely defined. Here, we investigated the effect of the Toll-like receptor 3 (TLR3) ligand polyinosinic-polycytidylic acid [Poly(I:C)], a synthetic analog of viral double-stranded RNA (dsRNA) commonly used to simulate viral infections, on the barrier function and tight junction integrity of primary human lung microvascular endothelial cells. Poly(I:C) stimulated IL-6, IL-8, TNFα, and IFNβ production in conjunction with the activation of NF-κB and IRF3 confirming the Poly(I:C)-responsiveness of these cells. Poly(I:C) increased endothelial monolayer permeability with a corresponding dose- and time-dependent decrease in the expression of claudin-5, a transmembrane tight junction protein and reduction of CLDN5 mRNA levels. Immunofluorescence experiments revealed disappearance of membrane-associated claudin-5 and co-localization of cytoplasmic claudin-5 with lysosomal-associated membrane protein 1. Chloroquine and Bay11-7082, inhibitors of TLR3 and NF-κB signaling, respectively, protected against the loss of claudin-5. Together, these findings provide new insight on how dsRNA-activated signaling pathways may disrupt vascular endothelial function and contribute to vascular leakage pathologies.
- Published
- 2016
- Full Text
- View/download PDF
28. Transcriptional Suppression of Renal Antioxidant Enzyme Systems in Guinea Pigs Exposed to Polymerized Cell-Free Hemoglobin.
- Author
-
Rentsendorj O, Zhang X, Williams MC, Buehler PW, and D'Agnillo F
- Abstract
Hemoglobin-based oxygen carriers (HBOCs) are being developed as oxygen and plasma volume-expanding therapeutics though their potential to promote oxidative tissue injury has raised safety concerns. Using a guinea pig exchange transfusion model, we examined the effects of polymerized bovine hemoglobin (HbG) on the transcriptional regulation, activity, and expression of the renal antioxidant enzymes; superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). HbG infusion downregulated the mRNA levels for genes encoding SOD isoforms 1-3, GPx1, GPx3, GPx4, and CAT. This transcriptional suppression correlated with decreased enzymatic activities for SOD, CAT, and GPx. Immunostaining revealed decreased protein expression of SOD1, CAT, and GPx1 primarily in renal cortical tubules. DNA methylation analyses identified CpG hypermethylation in the gene promoters for SOD1-3, GPx1, GPx3, and GPx4, suggesting an epigenetic-based mechanism underlying the observed gene repression. HbG also induced oxidative stress as evidenced by increased renal lipid peroxidation end-products and 4-HNE immunostaining, which could be the result of the depleted antioxidant defenses and/or serve as a trigger for increased DNA methylation. Together, these findings provide evidence that the renal exposure to HbG suppresses the function of major antioxidant defense systems which may have relevant implications for understanding the safety of hemoglobin-based products.
- Published
- 2016
- Full Text
- View/download PDF
29. 1918 pandemic influenza virus and Streptococcus pneumoniae co-infection results in activation of coagulation and widespread pulmonary thrombosis in mice and humans.
- Author
-
Walters KA, D'Agnillo F, Sheng ZM, Kindrachuk J, Schwartzman LM, Kuestner RE, Chertow DS, Golding BT, Taubenberger JK, and Kash JC
- Subjects
- Animals, Blood Coagulation, Disease Models, Animal, Female, Humans, Immunohistochemistry, Influenza A Virus, H1N1 Subtype, Influenza Pandemic, 1918-1919, Influenza, Human complications, Influenza, Human pathology, Mice, Mice, Inbred BALB C, Oligonucleotide Array Sequence Analysis, Orthomyxoviridae Infections complications, Orthomyxoviridae Infections pathology, Pneumococcal Infections complications, Pneumococcal Infections pathology, Pulmonary Embolism pathology, Reverse Transcriptase Polymerase Chain Reaction, Streptococcus pneumoniae, Coinfection complications, Influenza, Human microbiology, Orthomyxoviridae Infections microbiology, Pneumococcal Infections microbiology, Pulmonary Embolism microbiology
- Abstract
To study bacterial co-infection following 1918 H1N1 influenza virus infection, mice were inoculated with the 1918 influenza virus, followed by Streptococcus pneumoniae (SP) 72 h later. Co-infected mice exhibited markedly more severe disease, shortened survival time and more severe lung pathology, including widespread thrombi. Transcriptional profiling revealed activation of coagulation only in co-infected mice, consistent with the extensive thrombogenesis observed. Immunohistochemistry showed extensive expression of tissue factor (F3) and prominent deposition of neutrophil elastase on endothelial and epithelial cells in co-infected mice. Lung sections of SP-positive 1918 autopsy cases showed extensive thrombi and prominent staining for F3 in alveolar macrophages, monocytes, neutrophils, endothelial and epithelial cells, in contrast to co-infection-positive 2009 pandemic H1N1 autopsy cases. This study reveals that a distinctive feature of 1918 influenza virus and SP co-infection in mice and humans is extensive expression of tissue factor and activation of the extrinsic coagulation pathway leading to widespread pulmonary thrombosis., (Copyright © 2015 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
30. Sodium nitrite potentiates renal oxidative stress and injury in hemoglobin exposed guinea pigs.
- Author
-
Baek JH, Zhang X, Williams MC, Hicks W, Buehler PW, and D'Agnillo F
- Subjects
- Acute Kidney Injury blood, Acute Kidney Injury pathology, Animals, Biomarkers metabolism, Cell Death drug effects, Dose-Response Relationship, Drug, Drug Synergism, Fatty Acid-Binding Proteins metabolism, Guinea Pigs, Heme Oxygenase-1 metabolism, Hemoglobins administration & dosage, Infusions, Intravenous, Kidney metabolism, Kidney pathology, Lipid Peroxidation drug effects, Male, Methemoglobin metabolism, Methemoglobinemia blood, Methemoglobinemia pathology, NF-E2-Related Factor 2 metabolism, Nitrates administration & dosage, Oxidation-Reduction, Time Factors, Acute Kidney Injury chemically induced, Hemoglobins toxicity, Kidney drug effects, Methemoglobinemia chemically induced, Nitrates toxicity, Oxidative Stress drug effects
- Abstract
Methemoglobin-forming drugs, such as sodium nitrite (NaNO2), may exacerbate oxidative toxicity under certain chronic or acute hemolytic settings. In this study, we evaluated markers of renal oxidative stress and injury in guinea pigs exposed to extracellular hemoglobin (Hb) followed by NaNO2 at doses sufficient to simulate clinically relevant acute methemoglobinemia. NaNO2 induced rapid and extensive oxidation of plasma Hb in this model. This was accompanied by increased renal expression of the oxidative response effectors nuclear factor erythroid 2-derived-factor 2 (Nrf-2) and heme oxygenase-1 (HO-1), elevated non-heme iron deposition, lipid peroxidation, interstitial inflammatory cell activation, increased expression of tubular injury markers kidney injury-1 marker (KIM-1) and liver-fatty acid binding protein (L-FABP), podocyte injury, and cell death. Importantly, these indicators of renal oxidative stress and injury were minimal or absent following infusion of Hb or NaNO2 alone. Together, these results suggest that the exposure to NaNO2 in settings associated with increased extracellular Hb may potentiate acute renal toxicity via processes that are independent of NaNO2 induced erythrocyte methemoglobinemia., (Published by Elsevier Ireland Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
31. Extracellular Hb enhances cardiac toxicity in endotoxemic guinea pigs: protective role of haptoglobin.
- Author
-
Baek JH, Zhang X, Williams MC, Schaer DJ, Buehler PW, and D'Agnillo F
- Subjects
- Animals, Apoptosis, DNA Damage, Disease Models, Animal, Endotoxemia metabolism, Guinea Pigs, Heart Diseases metabolism, Heart Diseases pathology, Heme Oxygenase-1 metabolism, Iron metabolism, Lipopolysaccharides, Male, Myocardium metabolism, Myocardium pathology, Phagocytosis, Sepsis etiology, Sepsis metabolism, Endotoxemia complications, Haptoglobins pharmacology, Heart Diseases chemically induced, Heart Diseases prevention & control, Hemoglobins toxicity
- Abstract
Endotoxemia plays a major causative role in the myocardial injury and dysfunction associated with sepsis. Extracellular hemoglobin (Hb) has been shown to enhance the pathophysiology of endotoxemia. In the present study, we examined the myocardial pathophysiology in guinea pigs infused with lipopolysaccharide (LPS), a Gram-negative bacterial endotoxin, and purified Hb. We also examined whether the administration of the Hb scavenger haptoglobin (Hp) could protect against the effects observed. Here, we show that Hb infusion following LPS administration, but not either insult alone, increased myocardial iron deposition, heme oxygenase-1 expression, phagocyte activation and infiltration, as well as oxidative DNA damage and apoptosis assessed by 8-hydroxy-2'-deoxyguanosine (8-OHdG) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) immunostaining, respectively. Co-administration of Hp significantly attenuated the myocardial events induced by the combination of LPS and Hb. These findings may have relevant therapeutic implications for the management of sepsis during concomitant disease or clinical interventions associated with the increased co-exposures to LPS and Hb, such as trauma, surgery or massive blood transfusions.
- Published
- 2014
- Full Text
- View/download PDF
32. Anthrax lethal toxin downregulates claudin-5 expression in human endothelial tight junctions.
- Author
-
D'Agnillo F, Williams MC, Moayeri M, and Warfel JM
- Subjects
- Animals, Antigens, CD metabolism, Cadherins metabolism, Cell Death drug effects, Dose-Response Relationship, Drug, Endothelial Cells drug effects, Endothelial Cells metabolism, Female, Liver drug effects, Liver metabolism, Mice, Mitogen-Activated Protein Kinases metabolism, Proteolysis drug effects, RNA, Messenger genetics, Signal Transduction drug effects, Antigens, Bacterial pharmacology, Antigens, CD genetics, Bacterial Toxins pharmacology, Cadherins genetics, Endothelium, Vascular metabolism, Gene Expression Regulation drug effects, Tight Junctions metabolism
- Abstract
Vascular leakage pathologies such as pleural effusion and hemorrhage are hallmarks of anthrax pathogenesis. We previously reported that anthrax lethal toxin (LT), the major virulence factor of anthrax, reduces barrier function in cultured primary human microvascular endothelial cells. Here, we show that LT-induced barrier dysfunction is accompanied by the reduced expression of the endothelial tight junction (TJ) protein claudin-5 but no change in the expression of other TJ components occludin, ZO-1, ZO-2, or the adherens junction (AJ) protein VE-cadherin. The downregulation of claudin-5 correlated temporally and dose-dependently with the reduction of transendothelial electrical resistance. LT-induced loss of claudin-5 was independent of cell death and preceded the appearance of actin stress fibers and altered AJ morphology. Pharmacological inhibition of MEK-1/2, two kinases that are proteolytically inactivated by LT, showed a similar reduction in claudin-5 expression. We found that LT reduced claudin-5 mRNA levels but did not accelerate the rate of claudin-5 degradation. Mice challenged with LT also showed significant reduction in claudin-5 expression. Together, these findings support a possible role for LT disruption of endothelial TJs in the vascular leakage pathologies of anthrax.
- Published
- 2013
- Full Text
- View/download PDF
33. Hemoglobin-driven pathophysiology is an in vivo consequence of the red blood cell storage lesion that can be attenuated in guinea pigs by haptoglobin therapy.
- Author
-
Baek JH, D'Agnillo F, Vallelian F, Pereira CP, Williams MC, Jia Y, Schaer DJ, and Buehler PW
- Subjects
- Acute Kidney Injury drug therapy, Acute Kidney Injury etiology, Acute Kidney Injury pathology, Acute Kidney Injury physiopathology, Animals, Aorta pathology, Cardio-Renal Syndrome drug therapy, Cardio-Renal Syndrome etiology, Cardio-Renal Syndrome pathology, Cardio-Renal Syndrome physiopathology, Erythrocyte Deformability, Guinea Pigs, Haptoglobins metabolism, Heme Oxygenase-1 analysis, Hemoglobins metabolism, Humans, Kidney metabolism, Liver enzymology, Male, Nitric Oxide metabolism, Osmotic Fragility, Proteomics, Spleen enzymology, Time Factors, Blood Preservation, Haptoglobins therapeutic use, Hemoglobins adverse effects, Hemolysis, Kidney pathology, Transfusion Reaction
- Abstract
Massive transfusion of blood can lead to clinical complications, including multiorgan dysfunction and even death. Such severe clinical outcomes have been associated with longer red blood cell (rbc) storage times. Collectively referred to as the rbc storage lesion, rbc storage results in multiple biochemical changes that impact intracellular processes as well as membrane and cytoskeletal properties, resulting in cellular injury in vitro. However, how the rbc storage lesion triggers pathophysiology in vivo remains poorly defined. In this study, we developed a guinea pig transfusion model with blood stored under standard blood banking conditions for 2 (new), 21 (intermediate), or 28 days (old blood). Transfusion with old but not new blood led to intravascular hemolysis, acute hypertension, vascular injury, and kidney dysfunction associated with pathophysiology driven by hemoglobin (Hb). These adverse effects were dramatically attenuated when the high-affinity Hb scavenger haptoglobin (Hp) was administered at the time of transfusion with old blood. Pathologies observed after transfusion with old blood, together with the favorable response to Hp supplementation, allowed us to define the in vivo consequences of the rbc storage lesion as storage-related posttransfusion hemolysis producing Hb-driven pathophysiology. Hb sequestration by Hp might therefore be a therapeutic modality for enhancing transfusion safety in severely ill or massively transfused patients.
- Published
- 2012
- Full Text
- View/download PDF
34. Anthrax lethal toxin-mediated disruption of endothelial VE-cadherin is attenuated by inhibition of the Rho-associated kinase pathway.
- Author
-
Warfel JM and D'Agnillo F
- Subjects
- Adherens Junctions metabolism, Antigens, CD genetics, Cadherins genetics, Cells, Cultured, Endothelial Cells metabolism, Endothelium, Vascular cytology, Fluorescein-5-isothiocyanate analogs & derivatives, Fluorescein-5-isothiocyanate metabolism, Humans, Protein Kinase Inhibitors pharmacology, RNA, Messenger metabolism, Serum Albumin metabolism, rho-Associated Kinases metabolism, Antigens, Bacterial toxicity, Antigens, CD metabolism, Bacterial Toxins toxicity, Cadherins metabolism, Endothelial Cells drug effects, rho-Associated Kinases antagonists & inhibitors
- Abstract
Systemic anthrax disease is characterized by vascular leakage pathologies. We previously reported that anthrax lethal toxin (LT) induces human endothelial barrier dysfunction in a cell death-independent manner with actin stress fiber formation and disruption of adherens junctions (AJs). In the present study, we further characterize the molecular changes in the AJ complex and investigate whether AJ structure and barrier function can be preserved by modulating key cytoskeletal signaling pathways. Here, we show that LT reduces total VE-cadherin protein and gene expression but the expression of the key linker protein beta-catenin remained unchanged. The changes in VE-cadherin expression correlated temporally with the appearance of actin stress fibers and a two-fold increase in phosphorylation of the stress fiber-associated protein myosin light chain (p-MLC) and cleavage of Rho-associated kinase-1 (ROCK-1). Co-treatment with ROCK inhibitors (H-1152 and Y27632), but not an inhibitor of MLC kinase (ML-7), blocked LT-induced p-MLC enhancement and stress fiber formation. This was accompanied by the restoration of VE-cadherin expression and membrane localization, and attenuation of the LT-induced increase in monolayer permeability to albumin. Together, these findings suggest the ROCK pathway may be a relevant target for countering LT-mediated endothelial barrier dysfunction.
- Published
- 2011
- Full Text
- View/download PDF
35. Sodium nitrite induces acute central nervous system toxicity in guinea pigs exposed to systemic cell-free hemoglobin.
- Author
-
Buehler PW, Butt OI, and D'Agnillo F
- Subjects
- Animals, Central Nervous System metabolism, Central Nervous System Diseases metabolism, Claudins metabolism, Guinea Pigs, Humans, Male, Membrane Proteins metabolism, Occludin, Oxidation-Reduction, Phosphoproteins metabolism, Zonula Occludens-1 Protein, Central Nervous System drug effects, Central Nervous System Diseases chemically induced, Hemoglobins metabolism, Hemolysis, Sodium Nitrite toxicity
- Abstract
Systemic cell-free hemoglobin (Hb) released via hemolysis disrupts vascular homeostasis, in part, through the scavenging of nitric oxide (NO). Sodium nitrite (NaNO(2)) therapy can attenuate the hypertensive effects of Hb. However, the chemical reactivity of NaNO(2) with Hb may enhance heme- or iron-mediated toxicities. Here, we investigate the effect of NaNO(2) on the central nervous system (CNS) in guinea pigs exposed to systemic cell-free Hb. Intravascular infusion of NaNO(2), at doses sufficient to alleviate Hb-mediated blood pressure changes, reduced the expression of occludin, but not zona occludens-1 (ZO-1) or claudin-5, in cerebral tight junctions 4h after Hb infusion. This was accompanied by increased perivascular heme oxygenase-1 expression, neuronal iron deposition, increased astrocyte and microglial activation, and reduced expression of neuron-specific nuclear protein (NeuN). These CNS changes were not observed in animals treated with Hb or NaNO(2) alone. Taken together, these findings suggest that the use of nitrite salts to treat systemic Hb exposure may promote acute CNS toxicity., (Published by Elsevier Inc.)
- Published
- 2011
- Full Text
- View/download PDF
36. Blood-brain barrier disruption and oxidative stress in guinea pig after systemic exposure to modified cell-free hemoglobin.
- Author
-
Butt OI, Buehler PW, and D'Agnillo F
- Subjects
- Animals, Apoptosis drug effects, Astrocytes drug effects, Astrocytes pathology, Blood Transfusion, Blood-Brain Barrier drug effects, Blood-Brain Barrier enzymology, Caspase 3 metabolism, Cell-Free System drug effects, Enzyme Induction drug effects, Glial Fibrillary Acidic Protein metabolism, Guinea Pigs, Heme metabolism, Heme Oxygenase (Decyclizing) biosynthesis, Immunoglobulin G metabolism, Iron metabolism, Male, Membrane Proteins metabolism, Tight Junctions drug effects, Tight Junctions metabolism, Blood-Brain Barrier pathology, Hemoglobins administration & dosage, Hemoglobins pharmacology, Oxidative Stress drug effects
- Abstract
Systemic exposure to cell-free hemoglobin (Hb) or its breakdown products after hemolysis or with the use of Hb-based oxygen therapeutics may alter the function and integrity of the blood-brain barrier. Using a guinea pig exchange transfusion model, we investigated the effect of a polymerized cell-free Hb (HbG) on the expression of endothelial tight junction proteins (zonula occludens 1, claudin-5, and occludin), astrocyte activation, IgG extravasation, heme oxygenase (HO), iron deposition, oxidative end products (4-hydroxynonenal adducts and 8-hydroxydeoxyguanosine), and apoptosis (cleaved caspase 3). Reduced zonula occludens 1 expression was observed after HbG transfusion as evidenced by Western blot and confocal microscopy. Claudin-5 distribution was altered in small- to medium-sized vessels. However, total expression of claudin-5 and occludin remained unchanged except for a notable increase in occludin 72 hours after HbG transfusion. HbG-transfused animals also showed increased astrocytic glial fibrillary acidic protein expression and IgG extravasation after 72 hours. Increased HO activity and HO-1 expression with prominent enhancement of HO-1 immunoreactivity in CD163-expressing perivascular cells and infiltrating monocytes/macrophages were also observed. Consistent with oxidative stress, HbG increased iron deposition, 4-hydroxynonenal and 8-hydroxydeoxyguanosine immunoreactivity, and cleaved caspase-3 expression. Systemic exposure to an extracellular Hb triggers blood-brain barrier disruption and oxidative stress, which may have important implications for the use of Hb-based therapeutics and may provide indirect insight on the central nervous system vasculopathies associated with excessive hemolysis., (Published by Elsevier Inc.)
- Published
- 2011
- Full Text
- View/download PDF
37. Hemoglobin-based oxygen carriers: From mechanisms of toxicity and clearance to rational drug design.
- Author
-
Buehler PW, D'Agnillo F, and Schaer DJ
- Subjects
- Animals, Biomarkers metabolism, Blood Substitutes adverse effects, Humans, Oxidative Stress drug effects, Blood Substitutes toxicity, Drug Design, Hemoglobins metabolism, Oxygen metabolism
- Abstract
Hemoglobin-based oxygen carriers (HBOCs) have been developed to support blood oxygen transport capacity during hemorrhagic shock, hemolysis and ischemic insult. Existing product candidates have demonstrated considerable efficacy in experimental animal models and in clinical trial subjects; however, severe adverse safety signals that appeared in recent phase II and phase III clinical trials involving certain HBOCs have in part hindered further development and licensing. Emerging insights into hemoglobin (Hb) toxicity as well as physiologic Hb scavengers such as haptoglobin and CD163 that are capable of detoxifying extracellular Hb in vivo suggest that alternative product candidates could be designed. Together with novel animal models and biomarkers tailored to monitor the effects of extracellular Hb, a new generation of HBOCs can be envisioned., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
38. Differential induction of renal heme oxygenase and ferritin in ascorbate and nonascorbate producing species transfused with modified cell-free hemoglobin.
- Author
-
Butt OI, Buehler PW, and D'Agnillo F
- Subjects
- Animals, Blood Component Transfusion, Blotting, Western, Cattle, Creatinine blood, Guinea Pigs, Heme Oxygenase-1 metabolism, Hemoglobins administration & dosage, Hemoglobinuria blood, Iron metabolism, Male, Polymers metabolism, Rats, Rats, Sprague-Dawley, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Ascorbic Acid metabolism, Ferritins metabolism, Heme Oxygenase (Decyclizing) metabolism, Hemoglobins metabolism, Kidney enzymology, Kidney metabolism
- Abstract
Abstract Heme catabolism and iron sequestration systems play an important role in regulating the response to extracellular hemoglobin (Hb). We previously reported that extracellular Hb oxidizes more readily in the circulation of guinea pigs, a nonascorbate (AA)-producing species with similar plasma and tissue antioxidant status to humans, compared to rats, an AA-producing species. To determine whether these two species exhibit differences in heme catabolism and iron sequestration at the level of the kidney, we examined heme oxygenase (HO), H- and L-ferritin expression, nonheme iron deposition, and renal AA content following transfusion with polymerized bovine hemoglobin (HbG). Both species showed similar rates of hemoglobinuria but urinary HbG was significantly more oxidized in guinea pigs. HbG enhanced HO activity in both species but appeared greater and more sustained in guinea pigs. Conversely, rats showed a greater and more rapid induction of H- and L-ferritin as well as greater iron accumulation and AA content. Furthermore, ferrous and ferric iron deposits were detected in rats while only ferric iron was observed in guinea pigs. These findings suggest significant differences in the renal handling of HbG which may be important for understanding how endogenous antioxidant defenses may modulate the renal response to extracellular Hb.
- Published
- 2010
- Full Text
- View/download PDF
39. Toxicological consequences of extracellular hemoglobin: biochemical and physiological perspectives.
- Author
-
Buehler PW and D'Agnillo F
- Subjects
- Animals, Endothelial Cells metabolism, Erythrocytes metabolism, Hemoglobins metabolism, Hemolysis physiology, Humans, Hydrogen Peroxide metabolism, Oxidation-Reduction, Hemoglobins physiology
- Abstract
Under normal physiology, human red blood cells (RBCs) demonstrate a circulating lifespan of approximately 100-120 days with efficient removal of senescent RBCs taking place via the reticuloendothelial system, spleen, and bone marrow phagocytosis. Within this time frame, hemoglobin (Hb) is effectively protected by efficient RBC enzymatic systems designed to allow for interaction between Hb and diffusible ligands while preventing direct contact between Hb and the external environment. Under normal resting conditions, the concentration of extracellular Hb in circulation is therefore minimal and controlled by specific plasma and cellular (monocyte/macrophage) binding proteins (haptoglobin) and receptors (CD163), respectively. However, during pathological conditions leading to hemolysis, extracellular Hb concentrations exceed normal plasma and cellular binding capacities, allowing Hb to become a biologically relevant vasoactive and redox active protein within the circulation and at extravascular sites. Under conditions of genetic, drug-induced, and autoimmune hemolytic anemias, large quantities of Hb are introduced into the circulation and often lead to acute renal failure and vascular dysfunction. Interestingly, the study of chemically modified Hb for use as oxygen therapeutics has allowed for some basic understanding of extracellular Hb toxicity, particularly in the absence of functional clearance mechanisms and in circulatory antioxidant depleted states.
- Published
- 2010
- Full Text
- View/download PDF
40. Anthrax lethal toxin enhances IkappaB kinase activation and differentially regulates pro-inflammatory genes in human endothelium.
- Author
-
Warfel JM and D'Agnillo F
- Subjects
- Anthrax enzymology, Antigens, Bacterial metabolism, Bacterial Toxins metabolism, Cells, Cultured, Enzyme Activation drug effects, Humans, I-kappa B Proteins metabolism, Inflammation enzymology, Transcription Factor AP-1 metabolism, Transcription Factor RelA metabolism, Transcription, Genetic drug effects, Antigens, Bacterial pharmacology, Bacterial Toxins pharmacology, CD40 Antigens biosynthesis, Chemokine CCL2 biosynthesis, Endothelial Cells enzymology, Endothelium, Vascular enzymology, Gene Expression Regulation drug effects, I-kappa B Kinase metabolism
- Abstract
Anthrax lethal toxin (LT) was previously shown to enhance transcriptional activity of NF-kappaB in tumor necrosis factor-alpha-activated primary human endothelial cells. Here we show that this LT-mediated increase in NF-kappaB activation is associated with the enhanced degradation of the inhibitory proteins IkappaBalpha and IkappaBbeta but not IkappaBepsilon. Moreover, this was accompanied by enhanced activation of the IkappaB kinase complex (IKK), which is responsible for targeting IkappaB proteins for degradation. Importantly, LT enhancement of IkappaBalpha degradation was completely blocked by a selective IKKbeta inhibitor, whereas IkappaBbeta degradation was attenuated, suggesting a mechanistic link. Consistent with the above data, LT-cotreated cells show elevated phosphorylation of two IKK substrates, IkappaBalpha and p65, both of which were blocked by incubation with the IKKbeta inhibitor. Consistent with NF-kappaB activation, LT increased transcription of the NF-kappaB regulated gene CD40. Conversely, LT inhibited transcription of another NF-kappaB-regulated gene, CCL2. This inhibition was linked to the LT-mediated suppression of another CCL2-regulating transcription factor, AP-1 (activator protein-1). These data suggest that LT-mediated enhancement of NF-kappaB is IKK-dependent, but importantly, the net effect of LT on the transcription of proinflammatory genes is driven by the cumulative effect of LT on the particular set of transcription factors that regulate a given promoter. Together, these findings provide new mechanistic insight on how LT may disrupt the host response to anthrax.
- Published
- 2009
- Full Text
- View/download PDF
41. Sequestration of extracellular hemoglobin within a haptoglobin complex decreases its hypertensive and oxidative effects in dogs and guinea pigs.
- Author
-
Boretti FS, Buehler PW, D'Agnillo F, Kluge K, Glaus T, Butt OI, Jia Y, Goede J, Pereira CP, Maggiorini M, Schoedon G, Alayash AI, and Schaer DJ
- Subjects
- Animals, Dogs, Female, Glucocorticoids pharmacology, Guinea Pigs, Haptoglobins metabolism, Hemoglobins toxicity, Iron metabolism, Male, Nitric Oxide metabolism, Oxidative Stress, Haptoglobins pharmacology, Hemoglobins metabolism, Hypertension prevention & control
- Abstract
Release of hemoglobin (Hb) into the circulation is a central pathophysiologic event that contributes to morbidity and mortality in chronic hemolytic anemias and severe malaria. These toxicities arise from Hb-mediated vasoactivity, possibly due to NO scavenging and localized tissue oxidative processes. Currently, there is no established treatment that targets circulating extracellular Hb. Here, we assessed the role of haptoglobin (Hp), the primary scavenger of Hb in the circulation, in limiting the toxicity of cell-free Hb infusion. Using a canine model, we found that glucocorticoid stimulation of endogenous Hp synthesis prevented Hb-induced hemodynamic responses. Furthermore, guinea pigs administered exogenous Hp displayed decreased Hb-induced hypertension and oxidative toxicity to extravascular environments, such as the proximal tubules of the kidney. The ability of Hp to both attenuate hypertensive responses during Hb exposure and prevent peroxidative toxicity in extravascular compartments was dependent on Hb-Hp complex formation, which likely acts through sequestration of Hb rather than modulation of its NO- and O2-binding characteristics. Our data therefore suggest that therapies involving supplementation of endogenous Hb scavengers may be able to treat complications of acute and chronic hemolysis, as well as counter the adverse effects associated with Hb-based oxygen therapeutics.
- Published
- 2009
- Full Text
- View/download PDF
42. Acellular haemoglobin attenuates hypoxia-inducible factor-1alpha (HIF-1alpha) and its target genes in haemodiluted rats.
- Author
-
Manalo DJ, Buehler PW, Baek JH, Butt O, D'agnillo F, and Alayash AI
- Subjects
- Animals, Cell Hypoxia, Erythropoietin genetics, Erythropoietin metabolism, Heme Oxygenase (Decyclizing) genetics, Heme Oxygenase (Decyclizing) metabolism, Hemodilution, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Kidney metabolism, Nitric Oxide Synthase genetics, Nitric Oxide Synthase metabolism, Oxygen metabolism, Rats, Transcription, Genetic, Blood Substitutes pharmacology, Hemoglobins pharmacology, Hydroxyethyl Starch Derivatives pharmacology, Hypoxia-Inducible Factor 1, alpha Subunit antagonists & inhibitors
- Abstract
Hb (haemoglobin)-based blood substitutes represent a class of therapeutics designed to correct oxygen deficit under conditions of anaemia and traumatic blood loss. The influences of these agents on HIF-1alpha (hypoxia-inducible factor-1alpha) target genes involved in adaptation to hypoxia have so far not been studied. In the study presented here, rats underwent 80% ET (exchange transfusion) with either HS (hetastarch) or a polymerized Hb OG (Oxyglobin). HS induced dramatic EPO (erythropoietin) gene transcription, reaching a maximum at 4 h post-ET. In contrast, OG suppressed EPO transcription until approx. 24 h post-ET. Large plasma EPO levels that were observed post-ET with HS were significantly blunted in animals transfused with OG. OG, unlike HS, induced a sharp increase in HO-1 (haem oxygenase-1) transcription at 4 h, which declined rapidly within 24 h, whereas modest increases in iNOS [inducible (nitric oxide synthase)] and constitutive NOS [eNOS (endothelial NOS)] were detected over the control. Our results demonstrate for the first time that severe haemodilution-induced erythropoietic responses in kidneys were attenuated by a low-oxygen-affinity cell-free Hb and suggest that tissue-specific oxygen-sensing pathways can be influenced by allosterically modified Hbs.
- Published
- 2008
- Full Text
- View/download PDF
43. Anthrax lethal toxin enhances TNF-induced endothelial VCAM-1 expression via an IFN regulatory factor-1-dependent mechanism.
- Author
-
Warfel JM and D'Agnillo F
- Subjects
- Antigens, Bacterial immunology, Bacterial Toxins immunology, Cell Nucleus metabolism, E-Selectin immunology, E-Selectin metabolism, Endothelial Cells immunology, Endothelial Cells metabolism, Endothelial Cells virology, GATA2 Transcription Factor metabolism, Humans, Intercellular Adhesion Molecule-1 metabolism, NF-kappa B metabolism, Phosphorylation, Protein Kinases metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, STAT1 Transcription Factor metabolism, Vascular Cell Adhesion Molecule-1 genetics, Antigens, Bacterial metabolism, Bacterial Toxins metabolism, Interferon Regulatory Factor-1 metabolism, Tumor Necrosis Factor-alpha metabolism, Vascular Cell Adhesion Molecule-1 metabolism
- Abstract
Impaired host defenses and vascular dysfunction are hallmarks of the late, antibiotic-refractory stages of systemic anthrax infection. Anthrax lethal toxin (LT), a key virulence factor of Bacillus anthracis, was previously shown to enhance VCAM-1 expression on primary human endothelial cells suggesting a causative link between dysregulated adhesion molecule expression and the poor immune response and vasculitis associated with anthrax. In this study, we report that LT amplification of TNF-induced VCAM-1 expression is driven transcriptionally by the cooperative activation of NF-kappaB and IFN regulatory factor-1 (IRF-1). LT enhancement of NF-kappaB phosphorylation and nuclear translocation correlated temporally with a delayed reaccumulation of IkappaBalpha, while increased induction of IRF-1 was linked to STAT1 activation. LT failed to augment TNF-induced ICAM-1 or E-selectin expression, two adhesion molecules regulated by NF-kappaB, but not IRF-1. These results suggest that LT can differentially modulate NF-kappaB target genes and highlight the importance of IRF-1 in VCAM-1 enhancement. Altering the activity of key transcription factors involved in host response to infection may be a critical mechanism by which LT contributes to anthrax pathogenesis.
- Published
- 2008
- Full Text
- View/download PDF
44. Effects of endogenous ascorbate on oxidation, oxygenation, and toxicokinetics of cell-free modified hemoglobin after exchange transfusion in rat and guinea pig.
- Author
-
Buehler PW, D'Agnillo F, Hoffman V, and Alayash AI
- Subjects
- Animals, Blood Pressure drug effects, Cattle, Cell-Free System, Drug Stability, Guinea Pigs, Heart Rate drug effects, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Kidney drug effects, Kidney pathology, Male, Myocardium pathology, Oxidation-Reduction, Rats, Rats, Sprague-Dawley, Ascorbic Acid blood, Blood Substitutes pharmacokinetics, Blood Substitutes pharmacology, Blood Substitutes toxicity, Hemoglobins pharmacokinetics, Hemoglobins pharmacology, Hemoglobins toxicity, Oxygen blood, Plasma Exchange
- Abstract
Chemically modified hemoglobin (Hb) solutions are promising oxygen therapeutics; however, these agents are prone to intravascular oxidation. Using a 50% exchange transfusion (ET) model with bovine polymerized hemoglobin (PolyHbBv), we examined heme oxidation, oxygenation markers, and toxicokinetics in rats, an ascorbic acid (AA)-producing species, and in guinea pigs, a non-AA-producing species. Plasma AA decreased by 50% in guinea pigs after ET, but it was unchanged in rats for the first 20 h post-ET. Both species cleared PolyHbBv from the circulation at similar rates. However, exposure to ferric PolyHbBv over time was 5-fold greater in the guinea pig. Mass spectrometry analysis of plasma revealed oxidative modifications within the tetrameric fraction of PolyHbBv in guinea pig. Oxygen equilibrium curves of PolyHbBv measured in plasma after ET were more left-shifted in guinea pigs compared with rats, consistent with increased ferric PolyHbBv formation. Renal hypoxia-inducible factor (HIF)-1alpha, whose activity strictly depends on the partial pressure of oxygen increased over time, and it correlated inversely with circulating ferrous PolyHbBv in both species. Interestingly, HIF-1alpha activity was greater in guinea pigs compared with rats at 72 h post-ET. Mean arterial pressure increases were also greater in guinea pigs; however, minimal differences in cardiac and renal pathology were observed in either species. The present findings suggest the importance of plasma AA in maintaining the stability of acellular Hb susceptible to oxidation, and they may be relevant to humans, which display a similar plasma/tissue antioxidant status to guinea pig.
- Published
- 2007
- Full Text
- View/download PDF
45. The large clostridial toxins from Clostridium sordellii and C. difficile repress glucocorticoid receptor activity.
- Author
-
Tait AS, Dalton M, Geny B, D'Agnillo F, Popoff MR, and Sternberg EM
- Subjects
- Animals, Artificial Gene Fusion, COS Cells, Chlorocebus aethiops, Dexamethasone antagonists & inhibitors, Dexamethasone pharmacology, Genes, Reporter, Glucocorticoids antagonists & inhibitors, Glucocorticoids pharmacology, Luciferases analysis, Luciferases genetics, Phosphorylation, Tumor Necrosis Factor-alpha biosynthesis, p38 Mitogen-Activated Protein Kinases metabolism, Bacterial Proteins pharmacology, Bacterial Toxins pharmacology, Clostridioides difficile physiology, Clostridium sordellii physiology, Enterotoxins pharmacology, Receptors, Glucocorticoid antagonists & inhibitors
- Abstract
We have previously shown that Bacillus anthracis lethal toxin represses glucocorticoid receptor (GR) transactivation. We now report that repression of GR activity also occurs with the large clostridial toxins produced by Clostridium sordellii and C. difficile. This was demonstrated using a transient transfection assay system for GR transactivation. We also report that C. sordellii lethal toxin inhibited GR function in an ex vivo assay, where toxin reduced the dexamethasone suppression of the proinflammatory cytokine tumor necrosis factor alpha (TNF-alpha). Furthermore, the glucocorticoid antagonist RU-486 in combination with C. sordellii lethal toxin additively prevented glucocorticoid suppression of TNF-alpha. These findings corroborate the fact that GR is a target for the toxin and suggest a physiological role for toxin-associated GR repression in inflammation. Finally, we show that this repression is associated with toxins that inactivate p38 mitogen-activated protein kinase (MAPK).
- Published
- 2007
- Full Text
- View/download PDF
46. First-generation blood substitutes: what have we learned? Biochemical and physiological perspectives.
- Author
-
Alayash AI, D'Agnillo F, and Buehler PW
- Subjects
- Animals, Blood Substitutes chemistry, Blood Substitutes metabolism, Endothelial Cells metabolism, Hemoglobins chemistry, Hemoglobins genetics, Hemoglobins metabolism, Humans, Molecular Structure, Nitric Oxide metabolism, Oxidation-Reduction, Oxidative Stress drug effects, Protein Engineering, Reactive Oxygen Species metabolism, Recombinant Proteins toxicity, Structure-Activity Relationship, Blood Substitutes toxicity, Drug Design, Endothelial Cells drug effects, Hemoglobins toxicity
- Abstract
Chemically modified or recombinant hemoglobin (Hb)-based oxygen carriers (HBOCs) have been developed as oxygen therapeutics or 'blood substitutes' for use in a variety of clinical settings. Oxidative and nitrosative reactions of acellular Hb can limit the effectiveness and compromise the safety of HBOCs. The reactions between Hb and biologically relevant redox active molecules may also perturb redox sensitive signaling pathways. In recent years, systematic in vitro and in vivo structural and functional evaluation of several HBOCs has been carried out and, in some cases, delineated the 'structural' origin of their toxicity. This enables potential protective strategies against Hb-mediated side reactions to be rationally suggested. Here the authors provide an overview of their research experiences, novel insights into the molecular basis of toxicities of these products and some lessons learned.
- Published
- 2007
- Full Text
- View/download PDF
47. Anthrax lethal toxin enhances cytokine-induced VCAM-1 expression on human endothelial cells.
- Author
-
Steele AD, Warfel JM, and D'Agnillo F
- Subjects
- Cell Adhesion, Cells, Cultured, Endothelial Cells cytology, Humans, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinases metabolism, Protein Kinase Inhibitors pharmacology, Antigens, Bacterial pharmacology, Bacterial Toxins pharmacology, Endothelial Cells drug effects, Endothelial Cells metabolism, Gene Expression Regulation drug effects, Tumor Necrosis Factor-alpha pharmacology, Vascular Cell Adhesion Molecule-1 metabolism
- Abstract
Vascular endothelial dysfunction is thought to play a prominent role in systemic anthrax pathogenesis. We examined the effect of anthrax lethal toxin (LTx), a key virulence factor of Bacillus anthracis, on the expression of vascular cell adhesion molecule-1 (VCAM-1) on normal and cytokine-stimulated human lung microvascular endothelial cells. Confluent endothelial monolayers were treated with lethal factor (LF), protective antigen (PA), or both (LTx) in the presence or absence of tumor necrosis factor-alpha (TNFalpha). LTx enhanced cytokine-induced VCAM-1 expression and monocyte adhesion. LTx alone had no effect on VCAM-1 expression. LF, PA or the combination of a catalytically inactive mutant LF and PA failed to enhance cytokine-induced VCAM-1 expression. Treatment with inhibitors of mitogen-activated protein kinase kinases (MEKs) and mitogen-activated protein kinases did not reproduce the VCAM-1 enhancement effect of LTx, a known MEK metalloprotease, suggesting LTx-mediated MEK cleavage may not be a contributing factor.
- Published
- 2005
- Full Text
- View/download PDF
48. Anthrax lethal toxin induces endothelial barrier dysfunction.
- Author
-
Warfel JM, Steele AD, and D'Agnillo F
- Subjects
- Cadherins drug effects, Cadherins metabolism, Cells, Cultured, Dose-Response Relationship, Drug, Electric Impedance, Endothelial Cells pathology, Enzyme-Linked Immunosorbent Assay, Humans, Microscopy, Fluorescence, Antigens, Bacterial toxicity, Bacterial Toxins toxicity, Capillary Permeability drug effects, Endothelial Cells drug effects, Endothelial Cells metabolism
- Abstract
Hemorrhage and pleural effusion are prominent pathological features of systemic anthrax infection. We examined the effect of anthrax lethal toxin (LT), a major virulence factor of Bacillus anthracis, on the barrier function of primary human lung microvascular endothelial cells. We also examined the distribution patterns of cytoskeletal actin and vascular endothelial-cadherin (VE-cadherin), both of which are involved in barrier function regulation. Endothelial monolayers cultured on porous membrane inserts were treated with the LT components lethal factor (LF) and protective antigen (PA) individually, or in combination. LT induced a concentration- and time-dependent decrease in transendothelial electrical resistance that correlated with increased permeability to fluorescently labeled albumin. LT also produced a marked increase in central actin stress fibers and significantly altered VE-cadherin distribution as revealed by immunofluorescence microscopy and cell surface enzyme-linked immunosorbent assay. Treatment with LF, PA, or the combination of an inactive LF mutant and PA did not alter barrier function or the distribution of actin or VE-cadherin. LT-induced barrier dysfunction was not dependent on endothelial apoptosis or necrosis. The present findings support a possible role for LT-induced barrier dysfunction in the vascular permeability changes accompanying systemic anthrax infection.
- Published
- 2005
- Full Text
- View/download PDF
49. Redox active hemoglobin enhances lipopolysaccharide-induced injury to cultured bovine endothelial cells.
- Author
-
D'Agnillo F
- Subjects
- Animals, Aorta cytology, Apoptosis drug effects, Apoptosis physiology, Blood Proteins pharmacology, Caspase 3, Caspases metabolism, Cattle, Cell Cycle physiology, Cells, Cultured, Endothelium, Vascular cytology, Hydrogen Peroxide pharmacology, Oxidants pharmacology, Oxidation-Reduction, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Hemoglobins metabolism, Lipopolysaccharides toxicity
- Abstract
The interaction of cell-free hemoglobin with lipopolysaccharide (LPS) is thought to aggravate the pathophysiology of sepsis and/or septic shock. This study examines the possible modulatory role of cell-free hemoglobin on LPS-induced apoptosis of cultured bovine aortic endothelial cells. Experiments were performed with or without fetal bovine serum, a source of LPS-binding protein and soluble CD14. In the absence of serum, LPS alone or coincubated with purified bovine hemoglobin (BvHb), human hemoglobin (Hb), or alpha-cross-linked Hb (alphaalphaHb) did not induce apoptosis. In the presence of serum, LPS induced significant apoptosis. LPS combined with BvHb, Hb, or alphaalphaHb produced the same extent of apoptosis as LPS alone. To examine whether the H(2)O(2)-driven redox activity of hemoglobin alters LPS-induced apoptosis, glucose oxidase was added to the system to generate a subtoxic flux of H(2)O(2). The combined treatment of LPS, glucose oxidase, and BvHb, Hb, or alphaalphaHb enhanced apoptosis compared with LPS alone. These findings support a possible mechanism whereby the redox cycling of hemoglobin, and not its direct interaction with LPS, contributes to the hemoglobin-mediated enhancement of LPS-related pathophysiology.
- Published
- 2004
- Full Text
- View/download PDF
50. A role for the myoglobin redox cycle in the induction of endothelial cell apoptosis.
- Author
-
D'Agnillo F and Alayash AI
- Subjects
- Animals, Antioxidants pharmacology, Aorta cytology, Ascorbic Acid pharmacology, Buthionine Sulfoximine pharmacology, Caspase 3, Caspases metabolism, Catalase pharmacology, Cattle, Cell Adhesion, Cell Cycle physiology, Cell Nucleus ultrastructure, Cells, Cultured, Endothelium, Vascular metabolism, Enzyme Activation, Extracellular Space metabolism, Glucose Oxidase pharmacology, Glutathione metabolism, Horses, Hydrogen Peroxide metabolism, Iron chemistry, Myoglobin chemistry, Myoglobin pharmacology, Oxidation-Reduction, Apoptosis physiology, Endothelium, Vascular cytology, Myoglobin metabolism
- Abstract
This study investigates the potential role of the ferric/ferryl redox cycle of myoglobin (Mb) in the development of endothelial cell injury. Bovine aortic endothelial cells were incubated with ferric Mb (0.5-100 micro M) in the presence or absence of low steady states of H(2)O(2) (3-4 micro M) generated by glucose oxidase (GOX). The reaction of ferric Mb with H(2)O(2) generated ferryl Mb as monitored spectrophotometrically. Ferryl Mb formation correlated with the induction of apoptosis as indicated by morphological criteria, caspase 3 activation, phosphatidylserine (PS) externalization, and nuclear condensation by Hoechst 33342 staining. The addition of ascorbate or catalase inhibited the formation of ferryl Mb and the onset of apoptosis, whereas apoptosis was enhanced in cells depleted of intracellular glutathione by pretreatment with buthionine sulfoximine. Mb and Mb/GOX suppressed cell cycle progression, but only Mb/GOX produced significant cell loss revealed by the accumulation of sub G1 events. These results suggest a role for the Mb redox cycle in the induction of endothelial cell apoptosis, which may be relevant in the pathophysiology of diseases characterized by the release of Mb from damaged muscle.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.