Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Proc Natl Acad Sci U S A ; 117(27): 15554-15564, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32561649

ABSTRACT

The serum haptoglobin protein (Hp) scavenges toxic hemoglobin (Hb) leaked into the bloodstream from erythrocytes. In humans, there are two frequently occurring allelic forms of Hp, resulting in three genotypes: Homozygous Hp 1-1 and Hp 2-2, and heterozygous Hp 2-1. The Hp genetic polymorphism has an intriguing effect on the quaternary structure of Hp. The simplest form, Hp 1-1, forms dimers consisting of two α1ß units, connected by disulfide bridges. Hp 2-1 forms mixtures of linear (α1)2(α2)n-2(ß)n oligomers (n > 1) while Hp 2-2 occurs in cyclic (α2)n(ß)n oligomers (n > 2). Different Hp genotypes bind Hb with different affinities, with Hp 2-2 being the weakest binder. This behavior has a significant influence on Hp's antioxidant capacity, with potentially distinctive personalized clinical consequences. Although Hp has been studied extensively in the past, the finest molecular details of the observed differences in interactions between Hp and Hb are not yet fully understood. Here, we determined the full proteoform profiles and proteoform assemblies of all three most common genetic Hp variants. We combined several state-of-the-art analytical methods, including various forms of chromatography, mass photometry, and different tiers of mass spectrometry, to reveal how the tens to hundreds distinct proteoforms and their assemblies influence Hp's capacity for Hb binding. We extend the current knowledge by showing that Hb binding does not just depend on the donor's genotype, but is also affected by variations in Hp oligomerization, glycosylation, and proteolytic processing of the Hp α-chain.


Subject(s)
Haptoglobins/genetics , Hemoglobins/metabolism , Alleles , Antioxidants/chemistry , Antioxidants/isolation & purification , Antioxidants/metabolism , Glycosylation , Haptoglobins/chemistry , Haptoglobins/isolation & purification , Haptoglobins/metabolism , Hemoglobins/toxicity , Humans , Mass Spectrometry , Models, Molecular , Molecular Structure , Polymorphism, Genetic , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , Structure-Activity Relationship
2.
Am J Physiol Renal Physiol ; 317(4): F922-F929, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31364379

ABSTRACT

Acute kidney injury is a common complication of severe sepsis and contributes to high mortality. The molecular mechanisms of acute kidney injury during sepsis are not fully understood. Because hemoproteins, including myoglobin and hemoglobin, are known to mediate kidney injury during rhabdomyolysis, we hypothesized that cell-free hemoglobin (CFH) would exacerbate acute kidney injury during sepsis. Sepsis was induced in mice by intraperitoneal injection of cecal slurry (CS). To mimic elevated levels of CFH observed during human sepsis, mice also received a retroorbital injection of CFH or dextrose control. Four groups of mice were analyzed: sham treated (sham), CFH alone, CS alone, and CS + CFH. The addition of CFH to CS reduced 48-h survival compared with CS alone (67% vs. 97%, P = 0.001) and increased the severity of illness. After 24 and 48 h, CS + CFH mice had a reduced glomerular filtration rate from baseline, whereas sham, CFH, and CS mice maintained baseline glomerular filtration rate. Biomarkers of acute kidney injury, neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1), were markedly elevated in CS+CFH compared with CS (8-fold for NGAL and 2.4-fold for KIM-1, P < 0.002 for each) after 48 h. Histological examination showed a trend toward increased tubular injury in CS + CFH-exposed kidneys compared with CS-exposed kidneys. However, there were similar levels of renal oxidative injury and apoptosis in the CS + CFH group compared with the CS group. Kidney levels of multiple proinflammatory cytokines were similar between CS and CS + CFH groups. Human renal tubule cells (HK-2) exposed to CFH demonstrated increased cytotoxicity. Together, these results show that CFH exacerbates acute kidney injury in a mouse model of experimental sepsis, potentially through increased renal tubular injury.


Subject(s)
Acute Kidney Injury/pathology , Hemoglobins/toxicity , Sepsis/pathology , Acute Kidney Injury/etiology , Acute Kidney Injury/physiopathology , Animals , Cell Line , Cell Survival/drug effects , Cell-Free System , Cytokines/metabolism , Female , Glomerular Filtration Rate , Hepatitis A Virus Cellular Receptor 1/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Lipocalin-2/metabolism , Male , Mice , Mice, Inbred C57BL , Sepsis/complications , Survival Analysis
3.
J Neurochem ; 145(6): 464-473, 2018 06.
Article in English | MEDLINE | ID: mdl-29500821

ABSTRACT

Hemopexin (Hpx) binds heme with extraordinary affinity, and after haptoglobin may provide a second line of defense against the toxicity of extracellular hemoglobin (Hb). In this series of experiments, the hypothesis that Hpx protects neurons from Hb neurotoxicity was evaluated in murine primary cultures containing neurons and glial cells. Contrary to hypothesis, Hpx increased neuronal loss due to micromolar concentrations of Hb by 4- to 12-fold, as measured by LDH release assay; conversely, the neurotoxicity of hemin was completely prevented. The endogenous fluorescence of Hpx was quenched by Hb, consistent with transfer of Hb-bound heme to Hpx. This was associated with precipitation of globin chains, as detected by immunostaining and fluorescent Hb labeling. A portion of this precipitate attached firmly to cells and could not be removed by multiple washes. Concomitant treatment with haptoglobin (Hp) prevented globin precipitation and most of the increase in neuronal loss. Hpx weakly attenuated the increase in culture non-heme iron produced by Hb treatment, quantified by ferrozine assay. However, Hb-Hpx toxicity was iron-dependent, and was blocked by deferoxamine and ferrostatin-1. Up-regulation of cell ferritin expression, a primary cell defense against Hb toxicity, was not observed on western blots of culture lysates that had been concomitantly treated with Hpx. These results suggest that Hpx destabilizes Hb in the absence of haptoglobin, leading to globin precipitation and exacerbation of iron-dependent oxidative cell injury. Combined therapy with hemopexin plus haptoglobin may be preferable to hemopexin alone after CNS hemorrhage.


Subject(s)
Haptoglobins/metabolism , Hemoglobins/toxicity , Hemopexin/toxicity , Neurotoxicity Syndromes/physiopathology , Animals , Antidotes/pharmacology , Cyclohexylamines/pharmacology , Deferoxamine/pharmacology , Female , Ferritins/metabolism , Globins/metabolism , Heme Oxygenase-1/metabolism , Hemin/toxicity , Iron/metabolism , Male , Mice , Neuroglia/drug effects , Neurons/drug effects , Nonheme Iron Proteins/metabolism , Phenylenediamines/pharmacology , Pregnancy , Primary Cell Culture
4.
Biochem Biophys Res Commun ; 503(1): 152-156, 2018 09 03.
Article in English | MEDLINE | ID: mdl-29859185

ABSTRACT

Hemorrhage into the brain parenchyma or subarachnoid space is associated with edema and vascular injury that is likely mediated at least in part by the toxicity of hemoglobin. In contrast, extravascular blood appears to be less neurotoxic when localized to the retina or adjacent vitreous, the gel filling the posterior segment of the eye. In this study, the hypothesis that vitreous protects neurons from hemoglobin toxicity was investigated in a primary cortical cell culture model. Consistent with prior observations, hemoglobin exposure for 24 h resulted in death of most neurons without injury to co-cultured glia. Neuronal loss was reduced in a concentration-dependent fashion by bovine vitreous, with complete protection produced by 3% vitreous solutions. This effect was associated with a reduction in malondialdehyde but an increase in cell iron. At low vitreous concentrations, its ascorbate content was sufficient to account for most neuroprotection, as equivalent concentrations of ascorbate alone had a similar effect. However, other vitreous antioxidants provided significant protection when applied at concentrations present in undiluted vitreous, and prevented all neuronal loss when combined in the absence of ascorbate. These results indicate that vitreous is an antioxidant cocktail that robustly protects neurons from hemoglobin toxicity, and may contribute to the relative resistance of retinal neurons to hemorrhagic injury.


Subject(s)
Hemoglobins/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Vitreous Body/metabolism , Animals , Cattle , Cells, Cultured , Cerebral Cortex/metabolism , Hemoglobins/toxicity , Lipid Peroxidation/drug effects , Models, Neurological , Neurons/drug effects , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control
5.
J Neuroinflammation ; 13: 26, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26831741

ABSTRACT

BACKGROUND: Following intracerebral hemorrhage (ICH), red blood cells release massive amounts of toxic heme that causes local brain injury. Hemopexin (Hpx) has the highest binding affinity to heme and participates in its transport, while heme oxygenase 2 (HO2) is the rate-limiting enzyme for the degradation of heme. Microglia are the resident macrophages in the brain; however, the significance and role of HO2 and Hpx on microglial clearance of the toxic heme (iron-protoporphyrin IX) after ICH still remain understudied. Accordingly, we postulated that global deletion of constitutive HO2 or Hpx would lead to worsening of ICH outcomes. METHODS: Intracerebral injection of stroma-free hemoglobin (SFHb) was used in our study to induce ICH. Hpx knockout (Hpx(-/-)) or HO2 knockout (HO2(-/-)) mice were injected with 10 µL of SFHb in the striatum. After injection, behavioral/functional tests were performed, along with anatomical analyses. Iron deposition and neuronal degeneration were depicted by Perls' and Fluoro-Jade B staining, respectively. Immunohistochemistry with anti-ionized calcium-binding adapter protein 1 (Iba1) was used to estimate activated microglial cells around the injured site. RESULTS: This study shows that deleting Hpx or HO2 aggravated SFHb-induced brain injury. Compared to wild-type littermates, larger lesion volumes were observed in Hpx(-/-) and HO2(-/-) mice, which also bear more degenerating neurons in the peri-lesion area 24 h postinjection. Fewer Iba1-positive microglial cells were detected at the peri-lesion area in Hpx(-/-) and HO2(-/-) mice, interestingly, which is associated with markedly increased iron-positive microglial cells. Moreover, the Iba1-positive microglial cells increased from 24 to 72 h postinjection and were accompanied with improved neurologic deficits in Hpx(-/-) and HO2(-/-) mice. These results suggest that Iba1-positive microglial cells could engulf the extracellular SFHb and provide protective effects after ICH. We then treated cultured primary microglial cells with SFHb at low and high concentrations. The results show that microglial cells actively take up the extracellular SFHb. Of interest, we also found that iron overload in microglia significantly reduces the Iba1 expression level and resultantly inhibits microglial phagocytosis. CONCLUSIONS: This study suggests that microglial cells contribute to hemoglobin-heme clearance after ICH; however, the resultant iron overloads in microglia appear to decrease Iba1 expression and to further inhibit microglial phagocytosis.


Subject(s)
Brain Injuries/etiology , Brain Injuries/genetics , Cerebral Hemorrhage/complications , Heme Oxygenase (Decyclizing)/deficiency , Hemopexin/deficiency , Acyl-CoA Dehydrogenase/metabolism , Animals , Arabidopsis Proteins/metabolism , Cells, Cultured , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/mortality , Disease Models, Animal , Fluoresceins/metabolism , Heme Oxygenase (Decyclizing)/genetics , Hemoglobins/toxicity , Hemopexin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Motor Activity/genetics , Nerve Tissue Proteins/metabolism , Neurologic Examination , Phagocytosis/drug effects , Phagocytosis/genetics , Time Factors
6.
Anesth Analg ; 122(4): 1024-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26771264

ABSTRACT

BACKGROUND: It has been increasingly suggested that propofol protects against hypoxic-/ischemic-induced neuronal injury. As evidenced by hemorrhage-induced stroke, hemorrhage into the brain may also cause brain damage. Whether propofol protects against hemorrhage-induced brain damage remains unknown. Therefore, in this study, we investigated the effects of propofol on hemoglobin-induced cytotoxicity in cultured mouse cortical neurons. METHODS: Neurons were prepared from the cortex of embryonic 15-day-old mice. Hemoglobin was used to induce cytotoxicity in the neurons. The neurons were then treated with propofol for 4 hours. Cytotoxicity was determined by lactate dehydrogenase release assay. Caspase-3 activation was examined by Western blot analysis. Finally, the free radical scavenger U83836E was used to examine the potential involvement of oxidative stress in propofol's effects on hemoglobin-induced cytotoxicity. RESULTS: We found that treatment with hemoglobin induced cytotoxicity in the neurons. Propofol enhanced hemoglobin-induced cytotoxicity. Specifically, there was a significant difference in the amount of lactate dehydrogenase release between hemoglobin plus saline (19.84% ± 5.38%) and hemoglobin plus propofol (35.79% ± 4.41%) in mouse cortical neurons (P = 0.00058, Wilcoxon Mann-Whitney U test, n = 8 in the control group or the treatment group). U83836E did not attenuate the enhancing effects of propofol on hemoglobin-induced cytotoxicity in the neurons, and propofol did not significantly affect caspase-3 activation induced by hemoglobin. These data suggested that caspase-3 activation and oxidative stress might not be the underlying mechanisms by which propofol enhanced hemoglobin-induced cytotoxicity. Moreover, these data suggested that the neuroprotective effects of propofol would be dependent on the condition of the brain injury, which will need to be confirmed in future studies. CONCLUSIONS: These results from our current proof-of-concept study should promote more research in vitro and in vivo to develop better anesthesia care for patients with hemorrhagic stroke.


Subject(s)
Cytotoxins/toxicity , Hemoglobins/toxicity , Hypnotics and Sedatives/toxicity , Neurons/drug effects , Propofol/toxicity , Animals , Cells, Cultured , Mice , Neurons/metabolism , Neurons/pathology
7.
J Neuroinflammation ; 11: 106, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24924349

ABSTRACT

BACKGROUND: Translocation of high-mobility group box 1 (HMGB1) from nucleus could trigger inflammation. Extracellular HMGB1 up-regulates inflammatory response in sepsis as a late mediator. However, little was known about its role in subarachnoid hemorrhage-inducible inflammation, especially in the early stage. This study aims to identify whether HMGB1 translocation occurred early after SAH and also to clarify the potential role of HMGB1 in brain injury following SAH. METHODS: Sprague-Dawley (SD) rats were randomly divided into sham group and SAH groups at 2 h, 12 h and on day 1, day 2. SAH groups suffered experimental subarachnoid hemorrhage by injection of 0.3 ml autoblood into the pre-chiasmatic cistern. Rats injected by recombinant HMGB1(rHMGB1) solution were divided into four groups according to different time points. Cultured neurons were assigned into control group and four hemoglobin (Hb) incubated groups. Mixed glial cells were cultured and stimulated in medium from neurons incubated by Hb. HMGB1 expression is measured by western blot analysis, real-time polymerase chain reaction (PCR), immunohistochemistry and immunofluorescence. Downstream nuclear factor kappa B (NF-κB) subunit P65 and inflammatory factor Interleukin 1ß (IL-1ß) were measured by western blot and real-time PCR, respectively. Brain injury was evaluated by cleaved caspase-3 staining. RESULTS: Our results demonstrated HMGB1 translocation occurred as early as 2 h after experimental SAH with mRNA and protein level increased. Immunohistochemistry and immunofluorescence results indicated cytosolic HMGB1 was mainly located in neurons while translocated HMGB1 could also be found in some microglia. After subarachnoid injection of rHMGB1, NF-κB, downstream inflammatory response and cleaved caspase-3 were up-regulated in the cortex compared to the saline control group. In-vitro, after Hb incubation, HMGB1 was also rapidly released from neurons to medium. Incubation with medium from neurons up-regulated IL-1ß in mixed glial cells. This effect could be inhibited by HMGB1 specific inhibitor glycyrrhizic acid (GA) treatment. CONCLUSION: HMGB1 was released from neurons early after SAH onset and might trigger inflammation as an upstream inflammatory mediator. Extracellular HMGB1 contributed to the brain injury after SAH. These results might have important implications during the administration of specific HMGB1 antagonists early in order to prevent or reduce inflammatory response following SAH.


Subject(s)
Gene Expression Regulation/physiology , HMGB1 Protein/metabolism , Neurons/metabolism , Subarachnoid Hemorrhage/pathology , Animals , Brain Injuries/chemically induced , Brain Injuries/pathology , Cells, Cultured , Cerebral Cortex/pathology , Culture Media, Conditioned/chemistry , Disease Models, Animal , Embryo, Mammalian , Female , Gene Expression Regulation/drug effects , HMGB1 Protein/administration & dosage , HMGB1 Protein/genetics , Hemoglobins/toxicity , Male , Neuroglia/drug effects , Neurons/drug effects , Phosphopyruvate Hydratase/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/etiology , Time Factors
8.
J Neuroinflammation ; 10: 100, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23915174

ABSTRACT

BACKGROUND: Cerebral intraventricular hemorrhage (IVH) is a major cause of severe neurodevelopmental impairment in preterm infants. To date, no therapy is available that prevents infants from developing serious neurological disability following IVH. Thus, to develop treatment strategies for IVH, it is essential to characterize the initial sequence of molecular events that leads to brain damage. In this study, we investigated extracellular hemoglobin (Hb) as a causal initiator of inflammation in preterm IVH. METHODS: Using a preterm rabbit pup model, we investigated the molecular mechanisms and events following IVH. We also characterized the concentrations of cell-free Hb metabolites and pro-inflammatory mediators in the cerebrospinal fluid (CSF) of preterm human infants and rabbit pups. Finally, Hb metabolites were evaluated as causal initiators of inflammation in primary rabbit astrocyte cell cultures. RESULTS: Following IVH in preterm rabbit pups, the intraventricular CSF concentration of cell-free methemoglobin (metHb) increased from 24 to 72 hours and was strongly correlated with the concentration of TNFα at 72 hours (r2 = 0.896, P <0.001). Also, the mRNA expression of TNFα, IL-1ß, and Toll-like receptor-4 and TNFα protein levels were significantly increased in periventricular tissue at 72 hours, which was accompanied by extensive astrocyte activation (that is, glial fibrillary acidic protein (GFAP)staining). Furthermore, exposure of primary rabbit astrocyte cell cultures to metHb caused a dose-dependent increase in TNFα mRNA and protein levels, which was not observed following exposure to oxyhemoglobin (oxyHb) or hemin. Finally, a positive correlation (r2 = 0.237, P <0.03) between metHb and TNFα concentrations was observed in the CSF of preterm human infants following IVH. CONCLUSIONS: Following preterm IVH, increased metHb formation in the intraventricular space induces expression of pro-inflammatory cytokines. Thus, the formation of metHb might be a crucial initial event in the development of brain damage following preterm IVH. Accordingly, removal, scavenging, or neutralization of Hb could present a therapeutic opportunity and plausible approach to decreasing the damage in the immature brain following preterm IVH.


Subject(s)
Hemoglobins/toxicity , Inflammation/chemically induced , Intracranial Hemorrhages/metabolism , Methemoglobin/metabolism , Animals , Astrocytes/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Ventricles/pathology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Hemin/cerebrospinal fluid , Hemin/metabolism , Humans , Infant, Newborn , Infant, Premature , Inflammation/pathology , Intracranial Hemorrhages/pathology , Methemoglobin/cerebrospinal fluid , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Oxyhemoglobins/cerebrospinal fluid , Oxyhemoglobins/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/cerebrospinal fluid , Tumor Necrosis Factor-alpha/metabolism
9.
Parasite Immunol ; 33(4): 250-4, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21204852

ABSTRACT

To determine whether purified Ascaris suum haemoglobin (AsHb) is a suitable vaccine candidate for the control of Ascaris infections, pigs were vaccinated with AsHb in combination with QuilA adjuvant and challenged with A. suum eggs. The number of liver lesions and worms in the intestine was assessed on day 14, 28 and 56 post-infection (p.i.). No significant differences were found in the number of worms recovered between vaccinated and control pigs on any of these days. However, significantly more white spots were counted on the livers of vaccinated pigs on day 14 (+86%) and day 28 (+118%) p.i. compared with nonvaccinated controls. To investigate whether the increased immunoreactivity against the liver stage L3s in vaccinated pigs was triggered by and directed against AsHb, the transcription and expression of AsHb in this larval life stage was analysed by RT-PCR and immunoblotting. The results showed that neither the AsHb transcript nor protein was detectable in freshly hatched L3. However, the immunoblot analysis showed that vaccination with AsHb resulted in the production of antibodies binding to several other antigens of the L3, suggesting that these might be involved in the increased white spot development.


Subject(s)
Ascaris suum/immunology , Chemical and Drug Induced Liver Injury/pathology , Hemoglobins/immunology , Liver/pathology , Vaccines/adverse effects , Adjuvants, Immunologic/administration & dosage , Animals , Ascaris suum/pathogenicity , Hemoglobins/toxicity , Quillaja Saponins , Saponins/administration & dosage , Swine , Vaccines/immunology
10.
Mutat Res ; 718(1-2): 56-61, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-20974287

ABSTRACT

Iron is an important element that modulates the production of reactive oxygen species, which are thought to play a causative role in biological processes such as mutagenesis and carcinogenesis. The potential genotoxicity of dietary iron has been seldom studied in human leukocyte and only few reports have investigated in human colon tumor cells. Therefore, DNA damage and repair capacity of human leukocytes were examined using comet assay for screening the potential toxicity of various iron-overloads such as ferric-nitrilotriacetate (Fe-NTA), FeSO(4), hemoglobin and myoglobin, and compared with 200µM of H(2)O(2) and HNE. The iron-overloads tested were not cytotoxic in the range of 10-1000 microM by trypan blue exclusion assay. The exposure of leukocytes to Fe-NTA (500 and 1000 microM), FeSO(4) (250-1000 microM), hemoglobin (10 microM) and myoglobin (250 microM) for 30 min induced significantly higher DNA damage than NC. Treatment with 500 and 1000 microM of Fe-NTA showed a similar genotoxic effect to H(2)O(2), and a significant higher genotoxic effect than HNE. The genotoxicity of FeSO(4) (250-1000 microM), hemoglobin (10 microM) and myoglobin (250 microM) was not significantly different from that of H(2)O(2) and HNE. Iron-overloads generated DNA strand break were rejoined from the first 1h. Their genotoxic effect was not observed at 24h. These data from this study provide additional information on the genotoxicity of iron-overloads and self-repair capacity in human leukocytes.


Subject(s)
DNA Damage , DNA Repair , Iron Overload/genetics , Iron Overload/metabolism , Leukocytes/metabolism , Cell Survival/drug effects , Comet Assay , Ferric Compounds/toxicity , Hemoglobins/toxicity , Humans , In Vitro Techniques , Leukocytes/cytology , Leukocytes/drug effects , Mutagens/toxicity , Myoglobin/toxicity , Nitrilotriacetic Acid/analogs & derivatives , Nitrilotriacetic Acid/toxicity , Oxidative Stress/drug effects
11.
Int J Biol Macromol ; 187: 409-421, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34271050

ABSTRACT

The presence of excess glucose promotes hemoglobin glycation via the biochemical modification of hemoglobin by dicarbonyl products. However, the precise effects of Hb-AGEs in human umbilical vein endothelial cells (HUVECs) are not known to date. Therefore, we investigated the tentative effects of Hb-AGEs in HUVECs. Initially, we used the AGE formation assay to examine the selectivity of MGO toward various proteins. Among all proteins, MGO-Hb-AGEs formation was higher compared to the formation of other dicarbonyl-mediated AGEs. Our next data demonstrated that treatment with 0.5 mg/mL of Hb-AGEs-4w significantly reduced cell viability in HUVECs. Further, we evaluated the role of MGO in conformational and structural changes in Hb. The results showed that Hb demonstrated a highly altered conformation upon incubation with MGO. Moreover, Hb-AGEs-4w treatment strongly increased ROS production, and decreased mitochondrial membrane potential in HUVECs, and moderately reduced the expression of phosphorylated forms of p-38 and JNK. We observed that Hb-AGEs-4w treatment increased the number of apoptotic cells and the Bax/Bcl-2 ratio and cleaved the nuclear enzyme PARP in HUVECs. Finally, Hb-AGEs also inhibited migration and proliferation of HUVECs, thus be physiologically significant in endothelial dysfunction. Taken together, our data suggest that Hb-AGEs may play a critical role in inducing vascular endothelial cell damage. Therefore, this study may provide a plausible explanation for the potential Hb-AGEs in human endothelial cell dysfunction of diabetic patients.


Subject(s)
Apoptosis/drug effects , Glycation End Products, Advanced/toxicity , Hemoglobins/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Oxidative Stress/drug effects , Pyruvaldehyde/toxicity , Reactive Oxygen Species/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Phosphorylation , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Neurochem ; 114(4): 1063-73, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20497302

ABSTRACT

A growing body of experimental evidence suggests that an intracerebral hematoma is toxic to neighboring cells. However, injury mechanisms remain largely undefined, due in part to conflicting results from in vivo studies. In order to investigate blood toxicity in a more controlled environment, murine clots were co-cultured on porous membrane inserts with primary neurons and glia. Erythrocyte lysis was apparent within 48 h, but was reduced by almost 80% in cultures lacking neurons, and by over 90% in the absence of both neurons and glial cells. By 72 h, most released hemoglobin had oxidized to methemoglobin or its hemichrome degradation products. At this time point, approximately 50% of neurons were non-viable, as detected by propidium iodide staining; glia were not injured. Deferoxamine, Trolox and the NMDA receptor antagonist MK-801 prevented most neuronal death, but had no effect on hemolysis at neuroprotective concentrations. The 27-fold increase in culture malondialdehyde and 5.8-fold increase in heme oxygenase-1 expression were also attenuated by deferoxamine and Trolox, but not by MK-801. These results suggest that hemoglobin release from clotted blood is accelerated by adjacent neurons and glia. Subsequent neurotoxicity is mediated by both iron-dependent and excitotoxic injury pathways.


Subject(s)
Hematoma, Subdural, Chronic/pathology , Hemolysis/physiology , Neuroglia/pathology , Neurons/pathology , Neurotoxins/toxicity , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Coculture Techniques , Dizocilpine Maleate/administration & dosage , Hematoma, Subdural, Chronic/chemically induced , Hematoma, Subdural, Chronic/physiopathology , Heme Oxygenase-1/biosynthesis , Hemoglobins/toxicity , Hemolysis/drug effects , Iron/metabolism , Iron/toxicity , Malondialdehyde/metabolism , Mice , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Time Factors
13.
Trends Mol Med ; 26(7): 683-697, 2020 07.
Article in English | MEDLINE | ID: mdl-32589936

ABSTRACT

Hemolysis and accumulation of cell-free hemoglobin (Hb) in the circulation or in confined tissue compartments such as the subarachnoid space is an important driver of disease. Haptoglobin is the Hb binding and clearance protein in human plasma and an efficient antagonist of Hb toxicity resulting from physiological red blood cell turnover. However, endogenous concentrations of haptoglobin are insufficient to provide protection against Hb-driven disease processes in conditions such as sickle cell anemia, sepsis, transfusion reactions, medical-device associated hemolysis, or after a subarachnoid hemorrhage. As a result, there is increasing interest in developing haptoglobin therapeutics to target 'toxic' cell-free Hb exposures. Here, we discuss key concepts of Hb toxicity and provide a perspective on the use of haptoglobin as a therapeutic protein.


Subject(s)
Haptoglobins/pharmacology , Haptoglobins/therapeutic use , Hemoglobins/toxicity , Anemia, Sickle Cell/drug therapy , Animals , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , Sepsis/drug therapy , Transfusion Reaction/drug therapy
14.
Biochim Biophys Acta ; 1784(10): 1415-20, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18457681

ABSTRACT

Acellular hemoglobins developed as oxygen bridging agents with volume expanding properties ("blood substitutes") are prone to autoxidation and oxidant-mediated structural changes in circulation. In the presence of hydrogen peroxide and either ascorbate or urate we show that ferric hemoglobin functions as a true enzymatic peroxidase. The activity saturates with both substrates and is linearly dependent on protein concentration. The activity is enhanced at low pH with a pKa of 4.7, consistent with protonation of the ferryl species (Fe(IV)-OH) as the active intermediate. To test whether these redox reactions define its behaviour in vivo we exchanged transfused guinea pigs with 50% polymerized bovine Hb (PolyHbBv) and monitored plasma levels of endogenous ascorbate and urate. Immediately after transfusion, met PolyHbBv levels increased up to 30% of total Hb and remained at this level during the first 24 h post transfusion. Plasma ascorbate decreased by 50% whereas urate levels remained unchanged after transfusion. A simple kinetic model, assuming that ascorbate was a more active ferric heme reductase and peroxidase substrate than urate, was consistent with the in vivo data. The present finding confirms the primary and secondary roles of ascorbate and urate respectively in maintaining the oxidative stability of infused Hb.


Subject(s)
Ascorbic Acid/blood , Blood Substitutes/toxicity , Drug Carriers/toxicity , Hemoglobins/metabolism , Oxyhemoglobins/therapeutic use , Peroxidases/blood , Uric Acid/blood , Animals , Ascorbic Acid/pharmacology , Blood Substitutes/therapeutic use , Buffers , Ferric Compounds , Guinea Pigs , Hemoglobins/toxicity , Humans , Kinetics , Oxygen/blood , Oxygen/metabolism
15.
Circulation ; 117(15): 1982-90, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18391111

ABSTRACT

BACKGROUND: One of the major obstacles hindering the clinical development of a cell-free, hemoglobin-based oxygen carrier (HBOC) is systemic vasoconstriction. METHODS AND RESULTS: Experiments were performed in healthy mice and lambs by infusion of either murine tetrameric hemoglobin (0.48 g/kg) or glutaraldehyde-polymerized bovine hemoglobin (HBOC-201, 1.44 g/kg). We observed that intravenous infusion of either murine tetrameric hemoglobin or HBOC-201 induced prolonged systemic vasoconstriction in wild-type mice but not in mice congenitally deficient in endothelial nitric oxide (NO) synthase (NOS3). Treatment of wild-type mice by breathing NO at 80 ppm in air for 15 or 60 minutes or with 200 ppm NO for 7 minutes prevented the systemic hypertension induced by subsequent intravenous administration of murine tetrameric hemoglobin or HBOC-201 and did not result in conversion of plasma hemoglobin to methemoglobin. Intravenous administration of sodium nitrite (48 nmol) 5 minutes before infusion of murine tetrameric hemoglobin also prevented the development of systemic hypertension. In awake lambs, breathing NO at 80 ppm for 1 hour prevented the systemic hypertension caused by subsequent infusion of HBOC-201. CONCLUSIONS: These findings demonstrate that HBOC can cause systemic vasoconstriction by scavenging NO produced by NOS3. Moreover, in 2 species, inhaled NO administered before the intravenous infusion of HBOC can prevent systemic vasoconstriction without causing methemoglobinemia.


Subject(s)
Blood Substitutes/therapeutic use , Hemoglobins/therapeutic use , Hypertension/prevention & control , Nitric Oxide/therapeutic use , Vasoconstriction/drug effects , Vasodilator Agents/therapeutic use , Administration, Inhalation , Animals , Blood Substitutes/administration & dosage , Blood Substitutes/toxicity , Blood Transfusion , Drug Evaluation, Preclinical , Hemodynamics/drug effects , Hemoglobins/administration & dosage , Hemoglobins/toxicity , Hypertension/chemically induced , Infusions, Intravenous , Methemoglobinemia/prevention & control , Mice , Mice, Knockout , Nitric Oxide/administration & dosage , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III , Premedication , Sheep , Sodium Nitrite/administration & dosage , Sodium Nitrite/therapeutic use , Vasodilator Agents/administration & dosage , Wakefulness
16.
Toxicol In Vitro ; 23(3): 400-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19444923

ABSTRACT

Increased risk for development of colon cancer is associated with red meat intake and iron toxicity is discussed for one underlying mechanism. Anyhow, for iron itself only limited evidence is found. In this study, effects of different iron compounds on proliferation of HT29 carcinoma and LT97 adenoma human colon cells were investigated. After treatment of cells with inorganic (ferrous sulfate: FeSO4 and ferric nitrilotriacetate: FeNTA) and organic (hemoglobin and hemin) iron sources (24-72 h), number of cells and metabolic activity were measured. Under normal cell culture conditions, neither iron compound elevated cell growth in either cell line with the exception of FeNTA which induced LT97 cell growth significantly. Distinct inhibition of cell proliferation was measured for organic iron. Serum-free incubation of HT29 cells revealed growth promoting properties of iron under deficiency. Even though organic iron, especially hemin, was a potent growth factor, both substances showed also dose-dependent cytotoxic effects. In conclusion, these data emphasize that not iron itself, but merely organic iron may promote carcinogenic events. Since promotion of proliferation was only detectable under deficiency, cytotoxic properties of organic iron may be of more importance in colon carcinogenesis.


Subject(s)
Colon/drug effects , Hemin/toxicity , Hemoglobins/toxicity , Iron Compounds/toxicity , Adult , Cell Proliferation/drug effects , Cell Survival/drug effects , Colon/metabolism , Colon/pathology , Female , Ferric Compounds/toxicity , Ferrous Compounds/toxicity , HT29 Cells , Humans , Nitrilotriacetic Acid/analogs & derivatives , Nitrilotriacetic Acid/toxicity
17.
J Mater Chem B ; 7(31): 4821-4832, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31389959

ABSTRACT

Preparation of stable and effective artificial oxygen carriers (AOCs) is a promising strategy to temporarily replace transfused blood and solve tissue hypoxia. Developing hemoglobin (Hb) loaded particles is one of the main ways to prepare suitable AOCs. Particles with a hierarchical micro/nanostructure can be loaded with plenty of proteins and have attracted great attention. Therefore, multiwall carbon nanotubes (MWCNTs) were chosen to fabricate AOCs. To improve the Hb-loading capacity of MWCNTs, functionalized MWCNTs, including carboxyl-functionalized MWCNTs (MWCNT-COOH), amino-functionalized MWCNTs (MWCNT-NH2), and heparin-conjugated MWCNTs (MWCNT-Hep), were prepared. Then, in this study, Hb was coupled to the functionalized MWCNTs to fabricate the AOCs. The functionalized MWCNTs and the AOCs were characterized by FTIR, SEM, TEM, and zeta potential analysis. The oxygen/Hb-loading capacity of the AOCs was also measured. The adverse effects of the AOCs on human umbilical vein endothelial cells (HUVECs) and human red blood cells (RBCs) were evaluated. The results showed that (1) the functional groups were grafted on the surface of the MWCNTs, and Hb was bound to the functionalized MWCNTs, thus the AOCs were successfully prepared; (2) MWCNT-Hep-Hb had the most stable dispersibility (i.e., the most negative zeta potential) in 0.9 wt% NaCl solution (MWCNT-Hep-Hb < MWCNT-COOH-Hb < MWCNT-Hb < MWCNT-NH2-Hb < 0); (3) MWCNT-Hep had the best Hb-loading capability, which was three times that of purified MWCNTs; (4) with concentrations increased up to 400 µg mL-1, MWCNT-Hep-Hb still had the highest cell viability (97.63% > 80%, ISO 10993-5:2009) and excellent blood biocompatibility. Therefore, MWCNT-Hep-Hb might be a satisfactory candidate as a blood substitute.


Subject(s)
Blood Substitutes/pharmacology , Hemoglobins/pharmacology , Nanotubes, Carbon/chemistry , Oxygen/pharmacology , Blood Substitutes/chemistry , Blood Substitutes/toxicity , Cell Survival/drug effects , Erythrocytes/drug effects , Hemoglobins/chemistry , Hemoglobins/toxicity , Hemolysis/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Nanotubes, Carbon/toxicity , Oxygen/chemistry
18.
Neurobiol Dis ; 31(2): 242-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18571425

ABSTRACT

The effect of iron regulatory protein-2 (IRP2) on ferritin expression and neuronal vulnerability to hemoglobin was assessed in primary cortical cell cultures prepared from wild-type and IRP2 knockout mice. Baseline levels of H and L-ferritin subunits were significantly increased in IRP2 knockout neurons and astrocytes. Hemoglobin was toxic to wild-type neurons in mixed neuron-astrocyte cultures, with an LC(50) near 3 microM for a 24 h exposure. Neuronal death was reduced by 85-95% in knockout cultures, and also in cultures containing knockout neurons plated on wild-type astrocytes. Protein carbonylation, reactive oxygen species formation, and heme oxygenase-1 expression after hemoglobin treatment were also attenuated by IRP2 gene deletion. These results suggest that IRP2 binding activity increases the vulnerability of neurons to hemoglobin, possibly by reducing ferritin expression. Therapeutic strategies that target this regulatory mechanism may be beneficial after hemorrhagic CNS injuries.


Subject(s)
Cerebral Cortex/metabolism , Drug Resistance/genetics , Hemoglobins/toxicity , Iron Metabolism Disorders/metabolism , Iron Regulatory Protein 2/metabolism , Neurons/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Cerebral Cortex/physiopathology , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/physiopathology , Female , Ferritins/metabolism , Genetic Predisposition to Disease/genetics , Heme Oxygenase-1/metabolism , Hemoglobins/metabolism , Iron Metabolism Disorders/genetics , Iron Regulatory Protein 2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neurons/drug effects , Oxidative Stress/genetics , Protein Binding/genetics
19.
Toxicology ; 402-403: 37-49, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29689364

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.


Subject(s)
Glutaral/toxicity , Hemoglobins/toxicity , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Polymerization , Polymers/toxicity , Animals , Guinea Pigs , Kidney Glomerulus/physiopathology , Male , Proteinuria/chemically induced , Proteinuria/pathology , Proteinuria/physiopathology
20.
Toxicol Sci ; 166(1): 180-191, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30085279

ABSTRACT

Plasma hemoglobin (Hb) is elevated in some hematologic disease states, during exposures to certain toxicants, and with the use of some medical devices. Exposure to free Hb can precipitate oxidative reactions within tissues and alter the normal physiological function of critical organ systems. As kidney structures can be highly sensitive to Hb exposures, we evaluated the acute dose dependent renal toxicologic response to purified Hb isolated from RBCs. Male Hartley guinea pigs (n = 5 per group) were dosed with 0.9% saline (2 ml), 15, 75, 150, or 300 mg of purified Hb, infused over a 2-h period. The primary endpoints of this study were to define toxicokinetic parameters after increasing doses of purified Hb, identify clinically recognized and experimental markers of acute kidney injury (AKI), and determine relevant toxicological parameters and potential causes of renal toxicity in this model. Experimental findings demonstrated a dose dependent increase in Cmax after a 2-h infusion, which correlated with an elevation in serum creatinine, renal Kim-1 mRNA expression and increased urinary Kim-1. Renal NGAL mRNA expression and urinary NGAL excretion were also increased in several groups, but these parameters did not correlate with exposure. Iron increased in the renal cortex as Hb exposure increased and its deposition colocalized with 4-hydroxy-nonenal and 8-Oxo-2-deoxyguanosine immune reactivity, suggesting oxidative stressors may contribute to AKI in animals exposed to Hb. The results presented here suggest that Cmax may effectively predict the risk of AKI in normal healthy animals exposed to Hb.


Subject(s)
Acute Kidney Injury/chemically induced , Hemoglobins/pharmacokinetics , Hemoglobins/toxicity , Kidney/drug effects , Acute Kidney Injury/blood , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Biomarkers/metabolism , Dose-Response Relationship, Drug , Guinea Pigs , Hemoglobins/administration & dosage , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Iron/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Function Tests , Lipocalin-2/metabolism , Male , Models, Biological , Oxidative Stress/drug effects , Toxicokinetics
SELECTION OF CITATIONS
SEARCH DETAIL