Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(4)2022 Feb 12.
Article in English | MEDLINE | ID: mdl-35216154

ABSTRACT

The state of red blood cells (RBCs) and their functional possibilities depend on the structural organization of the membranes. Cell morphology and membrane nanostructure are compositionally and functionally related to the cytoskeleton network. In this work, the influence of agents (hemin, endogenous oxidation during storage of packed RBCs, ultraviolet (UV) radiation, temperature, and potential of hydrogen (pH) changes) on the relationships between cytoskeleton destruction, membrane nanostructure, and RBC morphology was observed by atomic force microscope. It was shown that the influence of factors of a physical and biochemical nature causes structural rearrangements in RBCs at all levels of organization, forming a unified mechanism of disturbances in relationships "cytoskeleton-membrane nanosurface-cell morphology". Filament ruptures and, consequently, large cytoskeleton pores appeared. The pores caused membrane topological defects in the form of separate grain domains. Increasing loading doses led to an increase in the number of large cytoskeleton pores and defects and their fusion at the membrane nanosurfaces. This caused the changes in RBC morphology. Our results can be used in molecular cell biology, membrane biophysics, and in fundamental and practical medicine.


Subject(s)
Cell Membrane/ultrastructure , Cytoskeleton/ultrastructure , Erythrocytes/pathology , Adult , Cells, Cultured , Erythrocytes/drug effects , Erythrocytes/radiation effects , Female , Hemin/toxicity , Humans , Hydrogen-Ion Concentration , Light/adverse effects , Male , Middle Aged , Oxidants/toxicity
2.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34576050

ABSTRACT

Intracerebral hemorrhage (ICH) occurs when brain blood vessels rupture, causing inflammation and cell death. 2-Fucosyllactose (2FL), a human milk oligosaccharide, has potent antiapoptotic and anti-inflammatory effects. The purpose of this study was to examine the protective effect of 2FL in cellular and rodent models of ICH. Hemin was added to a primary rat cortical neuronal and BV2 microglia coculture to simulate ICH in vitro. IBA1 and MAP2 immunoreactivities were used to determine inflammation and neuronal survival. Hemin significantly increased IBA1, while it reduced MAP2 immunoreactivity. 2FL significantly antagonized both responses. The protective effect of 2FL was next examined in a rat ICH model. Intracerebral administration of type VII collagenase reduced open-field locomotor activity. Early post-treatment with 2FL significantly improved locomotor activity. Brain tissues were collected for immunohistochemistry and qRT-PCR analysis. 2FL reduced IBA1 and CD4 immunoreactivity in the lesioned striatum. 2FL downregulated the expression of ER stress markers (PERK and CHOP), while it upregulated M2 macrophage markers (CD206 and TGFß) in the lesioned brain. Taken together, our data support that 2FL has a neuroprotective effect against ICH through the inhibition of neuroinflammation and ER stress. 2FL may have clinical implications for the treatment of ICH.


Subject(s)
Calcium-Binding Proteins/genetics , Hemorrhagic Stroke/drug therapy , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Trisaccharides/pharmacology , Animals , Cell Line , Coculture Techniques , Collagenases/toxicity , Disease Models, Animal , Gene Expression Regulation , Hemin/toxicity , Hemorrhagic Stroke/chemically induced , Hemorrhagic Stroke/genetics , Hemorrhagic Stroke/pathology , Humans , Locomotion/drug effects , Microglia/drug effects , Microglia/pathology , Milk, Human/chemistry , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Rats , Trisaccharides/chemistry
3.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34080030

ABSTRACT

Spontaneous intracerebral hemorrhage (ICH) is a subtype of stroke with high mortality and morbidity due to the lack of effective therapies. The alpha­amino­3­hydroxy­5­methyl­4­isoxazolepropionic acid receptor antagonist perampanel has been reported to alleviate early brain injury following subarachnoid hemorrhage and traumatic brain injury by reducing reactive oxygen species, apoptosis, autophagy, and necroptosis. Necroptosis is a caspase­independent programmed cell death mechanism that serves a vital role in neuronal cell death following ICH. However, the precise role of necroptosis in perampanel­mediated neuroprotection following ICH has not been confirmed. The present study aimed to investigate the neuroprotective effects and potential molecular mechanisms of perampanel in ICH­induced early brain injury by regulating neural necroptosis in C57BL/6 mice and in a hemin­induced neuron damage cell culture model. Mortality, neurological score, brain water content, and neuronal death were evaluated. The results demonstrated that perampanel treatment increased the survival rate and neurological score, and increased neuron survival. In addition, perampanel treatment downregulated the protein expression levels of receptor interacting serine/threonine kinase (RIP) 1, RIP3, and mixed lineage kinase domain like pseudokinase, and of the cytokines IL­1ß, IL­6, TNF­α, and NF­κB. These results indicated that perampanel­mediated inhibition of necroptosis and neuroinflammation ameliorated neuronal death in vitro and in vivo following ICH. The neuroprotective capacity of perampanel was partly dependent on the PTEN pathway. Taken together, the results of the present study demonstrated that perampanel improved neurological outcomes in mice and reduced neuronal death by protecting against neural necroptosis and neuroinflammation.


Subject(s)
Brain Injuries/drug therapy , Cerebral Hemorrhage/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Inflammation/drug therapy , Necroptosis/drug effects , Neuroprotective Agents/pharmacology , Nitriles/pharmacology , Pyridones/pharmacology , Administration, Oral , Animals , Brain Injuries/etiology , Brain Injuries/metabolism , Cell Death/drug effects , Cell Line , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/metabolism , Cytokines/metabolism , Disease Models, Animal , Hemin/toxicity , Inflammation/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Nitriles/administration & dosage , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pyridones/administration & dosage , Receptors, Glutamate/metabolism , Signal Transduction/drug effects
4.
J Cell Physiol ; 236(10): 6920-6931, 2021 10.
Article in English | MEDLINE | ID: mdl-33792028

ABSTRACT

Subarachnoid hemorrhage (SAH) is a subtype of stroke with high mortality and morbidity due to the lack of effective therapy. Atorvastatin has been reported to alleviate early brain injury (EBI) following subarachnoid hemorrhage (SAH) via reducing reactive oxygen species, antiapoptosis, regulated autophagy, and neuroinflammation. Which was the related to the pyroptosis? Pyroptosis can be defined as a highly specific inflammatory programmed cell death, distinct from classical apoptosis and necrosis. However, the precise role of pyroptosis in atorvastatin-mediated neuroprotection following SAH has not been confirmed. The present study aimed to investigate the neuroprotection and potential molecular mechanisms of atorvastatin in the SAH-induced EBI via regulating neural pyroptosis using the filament perforation model of SAH in male C57BL/6 mice, and the hemin-induced neuron damage model in HT-22. Atorvastatin or vehicle was administrated 2 h after SAH and hemin-induced neuron damage. The mortality, neurological score, brain water content, and neuronal death were evaluated. The results show that the atorvastatin treatment markedly increased survival rate, neurological score, greater survival of neurons, downregulated the protein expression of NLRP1, cleaved caspase-1, interleukin-1ß (IL-1ß), and IL-18, which indicated that atorvastatin-inhibited pyroptosis and neuroinflammation, ameliorated neuron death in vivo/vitro subjected to SAH. Taken together, this study demonstrates that atorvastatin improved the neurological outcome in rats and reduced the neuron death by against neural pyroptosis and neuroinflammation.


Subject(s)
Atorvastatin/pharmacology , Brain Injuries/prevention & control , Brain/drug effects , Encephalitis/prevention & control , Neurons/drug effects , Neuroprotective Agents/pharmacology , Pyroptosis/drug effects , Subarachnoid Hemorrhage/drug therapy , Animals , Brain/metabolism , Brain/pathology , Brain Edema/etiology , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/prevention & control , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Case-Control Studies , Caspase 1/metabolism , Cell Line , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Encephalitis/etiology , Encephalitis/metabolism , Encephalitis/pathology , Hemin/toxicity , Humans , Inflammation Mediators/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurons/metabolism , Neurons/pathology , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology
5.
Int J Nanomedicine ; 16: 775-788, 2021.
Article in English | MEDLINE | ID: mdl-33574665

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) is a common neurological crisis leading to high mortality and morbidity. Oxidative stress-induced secondary injury plays a critical role in neurological deterioration. Previously, we synthesized a porous Se@SiO2 nanocomposite and identified their therapeutic role in osteonecrosis of the femoral head. Whether this nanocomposite is neuroprotective remains to be elucidated. METHODS: A porous Se@SiO2 nanocomposite was synthesized, and its biosafety was determined using a CCK-8 assay. The neuroprotective effect was evaluated by TUNEL staining, and intracellular ROS were detected with a DCFH-DA probe in SH-SY5Y cells exposed to hemin. Furthermore, the effect of the nanocomposite on cell apoptosis, brain edema and blood-brain barrier permeability were evaluated in a collagenase-induced ICH mouse model. The potential mechanism was also explored. RESULTS: The results demonstrated that Se@SiO2 treatment significantly improved neurological function, increased glutathione peroxidase activity and downregulated malonaldehyde levels. The proportion of apoptotic cells, brain edema and blood-brain barrier permeability were reduced significantly in ICH mice treated with Se@SiO2 compared to vehicle-treated mice. In vitro, Se@SiO2 protected SH-SY5Y cells from hemin-induced apoptosis by preventing intracellular reactive oxygen species accumulation. CONCLUSION: These results suggested that the porous Se@SiO2 nanocomposite exerted neuroprotection by suppressing oxidative stress. Se@SiO2 may be a potential candidate for the clinical treatment of ICH and oxidative stress-related brain injuries.


Subject(s)
Brain/pathology , Cerebral Hemorrhage/pathology , Nanocomposites/chemistry , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Selenium/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Biomarkers/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/drug effects , Brain Edema/complications , Brain Edema/drug therapy , Cell Line, Tumor , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/drug therapy , Cytoprotection/drug effects , Disease Models, Animal , Hemin/toxicity , Humans , Male , Malondialdehyde/metabolism , Mice, Inbred C57BL , Nanocomposites/toxicity , Nanocomposites/ultrastructure , Neuroprotection/drug effects , Neuroprotective Agents/therapeutic use , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Selenium/therapeutic use , Silicon Dioxide/pharmacology , Toxicity Tests
6.
Biochem Pharmacol ; 175: 113900, 2020 05.
Article in English | MEDLINE | ID: mdl-32156661

ABSTRACT

During hemolysis, free heme released from damaged RBCs impairs adjacent cells. As a response, heme induces its metabolic degradation via heme oxygenase-1 (HO-1), activated by NF-E2-related factor 2 (NRF2), the master stress response transcription factor. Heme is well considered a signaling molecule, but how heme does activate NRF2 is not well understood. K562, human pro-erythroid cells responding to hemin (ferric chloride heme), were employed to uncover the major role of Kelch-like ECH-associated protein 1 (KEAP1)/NRF2 stress response signaling, embedded in hemin-induced cytotoxicity (HIC), at ≥50 µM. The intracellular pools of hemin were found to determine the progression from the reversible cell growth inhibition to non-apoptotic cell death. Hemin-induced accumulation of both reactive oxygen species (ROS) and ubiquitinated proteins provoked disturbed cellular proteostasis. Immediate accumulation and nuclear translocation of NRF2 were recorded as defensive adaptation. The NRF2-driven genes encoding glutamate-cysteine ligase (GCLC) and cystine/glutamate antiporter (xCT) were substantially activated. Hemin orchestrated a defensive pathway involving the management of cellular non-protein thiols, via an increase in GSH levels and secretion of cysteine. Mechanistically, hemin stabilized NRF2 protein levels selectively by inhibiting the KEAP1-driven ubiquitination of NRF2, while allowing KEAP1 ubiquitination. High-molecular-weight ubiquitinated KEAP1 variants formed in hemin-treated cells degraded in proteasomes, while a portion of them translocated into the nucleus. The KEAP1/NRF2 system can be revealed as a basic homeostatic mechanism, activated in cells encountering free heme, both in healthy and diseased state. Its activation provides a multi-target cytoprotective platform to develop agents preventing heme toxicity.


Subject(s)
Cytotoxins/toxicity , Erythroid Cells/metabolism , Hemin/toxicity , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Dose-Response Relationship, Drug , Erythroid Cells/drug effects , Humans , K562 Cells , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Article in English | MEDLINE | ID: mdl-31585636

ABSTRACT

Red and processed meat consumption has been strongly related to increase the risk of colorectal cancer (CRC), although its impact is largely unknown. Hemin, an iron-containing porphyrin, is acknowledged as a putative factor of red and processed meat pro-carcinogenic effects. The aim of this study was to investigate the effects of high dietary hemin on the promotion/progression stages of 1,2-dimethylhydrazine (1,2-DMH)-induced colon carcinogenesis. Twenty-four Wistar male rats were given four subcutaneous 1,2-DMH injections and received either balanced diet or balanced diet supplemented with hemin 0.5 mmol/kg for 23 weeks. Colon specimens were analyzed for aberrant crypt foci (ACF) and tumor development. Dietary hemin significantly increased ACF number and fecal water cytotoxicity/genotoxicity in Caco-2 cells when compared to 1,2-DMH control group. However, tumor incidence, multiplicity and cell proliferation did not differ between 1,2-DMH + hemin and 1,2-DMH control group. Gene expression analysis of 91 target-genes revealed that only three genes (Figf, Pik3r5 and Tgfbr2) were down-regulated in the tumors from hemin-fed rats compared to those from 1,2-DMH control group. Therefore, the findings of this study show that high hemin intake promotes mainly DNA damage and ACF development and but does not change the number nor incidence of colon tumors induced by 1,2-DMH in male rats.


Subject(s)
Aberrant Crypt Foci/chemically induced , Colonic Neoplasms/chemically induced , DNA Damage , Hemin/toxicity , Precancerous Conditions/chemically induced , 1,2-Dimethylhydrazine , Animal Feed , Animals , Caco-2 Cells , Cocarcinogenesis , Comet Assay , Down-Regulation/drug effects , Feces , Humans , Male , Phosphatidylinositol 3-Kinase/genetics , Rats , Rats, Wistar , Receptor, Transforming Growth Factor-beta Type II/biosynthesis , Receptor, Transforming Growth Factor-beta Type II/genetics , Red Meat , Time Factors , Vascular Endothelial Growth Factor D/biosynthesis , Vascular Endothelial Growth Factor D/genetics
8.
Toxicology ; 426: 152254, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31356851

ABSTRACT

Metamizole is an analgesic and antipyretic with a superior analgesic efficacy than paracetamol. Since metamizole can cause neutropenia and agranulocytosis, it is currently used in only few countries. In a previous study, we have shown that N-methyl-4-aminoantipyrine (MAA), the active metamizole metabolite, reacts with hemin and forms an electrophilic metabolite that is toxic for HL60 cells, but not for mature neutrophil granulocytes. In the current study, we investigated the toxicity of hemin (12.5 µM) and MAA (100 µM) on differentiating HL60 cells. In undifferentiated HL60 cells, hemin decreased the viability and this effect was significantly increased by MAA. Similarly, hemin/MAA was more toxic than hemin alone on human cord blood cells. At 3 days (metamyelocyte stage) and 5 days of differentiation (mature neutrophils), hemin/MAA was not toxic on HL60 cells, whereas hemin alone was still toxic. No toxicity was observed on freshly isolated human neutrophils. The protein expression of enzymes responsible for hemin metabolism increased with HL60 cell differentiation. Inhibition of heme oxygenase-1 or cytochrome P450 reductase increased the toxicity of hemin and hemin/MAA in undifferentiated, but only for hemin in differentiated HL60 cells. Similar to the enzymes involved in hemin metabolism, the protein expression of enzymes involved in antioxidative defense and the cellular glutathione pool increased with HL60 cell differentiation. In conclusion, HL60 cells become resistant to the toxicity of hemin/MAA and partly also of hemin during their differentiation. This resistance is associated with the development of heme metabolism and of the antioxidative defense system including the cellular glutathione pool.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Dipyrone/toxicity , Granulocytes/drug effects , Neutrophils/drug effects , Antioxidants/metabolism , Antipyrine/analogs & derivatives , Antipyrine/toxicity , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Fetal Blood/drug effects , HL-60 Cells , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Hemin/toxicity , Humans , Tumor Stem Cell Assay
9.
J Inorg Biochem ; 193: 166-172, 2019 04.
Article in English | MEDLINE | ID: mdl-30772693

ABSTRACT

Due to the traditional therapies of cancer inducing huge pains to patients, the non-invasive photo-guided therapies are attracting massive attentions of researchers. Herein, the intelligent-designed carbon-dots/hemin nanoplatforms (HCDs NPs) were developed, owning high-authority photo-therapy for cancer. The fluorescence resonance energy transfer (FRET) effect enhanced the photo-thermal ability of HCDs NPs, endowing the synthesized nanoplatforms with photo-dynamic property simultaneously. Therefore, the obtained HCDs NPs could achieve synergetic photo-thermal and photo-dynamic therapies for cancer. Basing on the experimental results, the prepared HCDs NPs could induce the temperature enhancement high to ca 26 °C under laser irradiation, also with the outstanding photo-dynamic efficacy. More than 90% of cancer cells die after 10 min laser treatment. Thus, the dual-modal photo-therapeutic HCDs NPs are promising and excellent nanomaterials for potential application in synergistic cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Hemin/pharmacology , Quantum Dots/chemistry , Antineoplastic Agents/radiation effects , Antineoplastic Agents/toxicity , Carbon/chemistry , Carbon/radiation effects , Carbon/toxicity , Fluorescence Resonance Energy Transfer , Hemin/radiation effects , Hemin/toxicity , Hep G2 Cells , Humans , Hyperthermia, Induced , Light , Particle Size , Photochemotherapy , Quantum Dots/radiation effects , Quantum Dots/toxicity , Reactive Oxygen Species/metabolism , Solubility , Temperature
10.
Ann Neurol ; 84(6): 854-872, 2018 12.
Article in English | MEDLINE | ID: mdl-30294906

ABSTRACT

OBJECTIVES: N-acetylcysteine (NAC) is a clinically approved thiol-containing redox modulatory compound currently in trials for many neurological and psychiatric disorders. Although generically labeled as an "antioxidant," poor understanding of its site(s) of action is a barrier to its use in neurological practice. Here, we examined the efficacy and mechanism of action of NAC in rodent models of hemorrhagic stroke. METHODS: Hemin was used to model ferroptosis and hemorrhagic stroke in cultured neurons. Striatal infusion of collagenase was used to model intracerebral hemorrhage (ICH) in mice and rats. Chemical biology, targeted lipidomics, arachidonate 5-lipoxygenase (ALOX5) knockout mice, and viral-gene transfer were used to gain insight into the pharmacological targets and mechanism of action of NAC. RESULTS: NAC prevented hemin-induced ferroptosis by neutralizing toxic lipids generated by arachidonate-dependent ALOX5 activity. NAC efficacy required increases in glutathione and is correlated with suppression of reactive lipids by glutathione-dependent enzymes such as glutathione S-transferase. Accordingly, its protective effects were mimicked by chemical or molecular lipid peroxidation inhibitors. NAC delivered postinjury reduced neuronal death and improved functional recovery at least 7 days following ICH in mice and can synergize with clinically approved prostaglandin E2 (PGE2 ). INTERPRETATION: NAC is a promising, protective therapy for ICH, which acted to inhibit toxic arachidonic acid products of nuclear ALOX5 that synergized with exogenously delivered protective PGE2 in vitro and in vivo. The findings provide novel insight into a target for NAC, beyond the generic characterization as an antioxidant, resulting in neuroprotection and offer a feasible combinatorial strategy to optimize efficacy and safety in dosing of NAC for treatment of neurological disorders involving ferroptosis such as ICH. Ann Neurol 2018;84:854-872.


Subject(s)
Acetylcysteine/therapeutic use , Arachidonate 5-Lipoxygenase/metabolism , Cation Transport Proteins/metabolism , Dinoprostone/metabolism , Free Radical Scavengers/therapeutic use , Stroke/drug therapy , Acetylcysteine/pharmacology , Animals , Arachidonate 5-Lipoxygenase/genetics , Cation Transport Proteins/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cells, Cultured , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/complications , Collagenases/toxicity , Cytoplasm/metabolism , Disease Models, Animal , Eicosanoids/metabolism , Female , Free Radical Scavengers/pharmacology , Glutathione/metabolism , Hemin/toxicity , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Stroke/etiology , Treatment Outcome
11.
Br J Haematol ; 182(2): 271-275, 2018 07.
Article in English | MEDLINE | ID: mdl-29923176

ABSTRACT

Acute chest syndrome (ACS) mortality in sickle cell disease (SCD) rises sharply in young adult patients and mechanism-based prophylaxis is lacking. In SCD, haem oxygenase-1 (HO-1) declines with age and ACS is associated with low HO-1. To test if enhanced HO-1 can reduce ACS mortality, young SCD mice were treated with D3T (3H-1,2-dithiole-3-thione), an activator of nuclear-factor erythroid 2 like 2, which controls HO-1 expression, for 3 months. Following haem-induced ACS, all vehicle-treated mice succumbed to severe lung injury, while D3T-treated mice had significantly improved survival. Blocking HO-1 activity abrogated the D3T effect. Thus HO-1 may be targeted to reduce ACS severity in adult patients.


Subject(s)
Acute Chest Syndrome/prevention & control , NF-E2-Related Factor 2/physiology , Acute Chest Syndrome/chemically induced , Animals , Hematinics/pharmacology , Heme Oxygenase-1/metabolism , Hemin/toxicity , Mice, Transgenic , Oxygen/blood , Thiones/pharmacology , Thiophenes/pharmacology
12.
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
13.
Antioxid Redox Signal ; 29(2): 111-127, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29065700

ABSTRACT

AIMS: Mesenchymal stromal cells (MSCs) are heterogeneous cells from adult tissues that are able to differentiate in vitro into adipocytes, osteoblasts, or chondrocytes. Such cells are widely studied in regenerative medicine. However, the success of cellular therapy depends on the cell survival. Heme oxygenase-1 (HO-1, encoded by the Hmox1 gene), an enzyme converting heme to biliverdin, carbon monoxide, and Fe2+, is cytoprotective and can affect stem cell performance. Therefore, our study aimed at assessing whether Hmox1 is critical for survival and functions of murine bone marrow MSCs. RESULTS: Both MSC Hmox1+/+ and Hmox1-/- showed similar phenotype, differentiation capacities, and production of cytokines or growth factors. Hmox1+/+ and Hmox1-/- cells showed similar survival in response to 50 µmol/L hemin even in increased glucose concentration, conditions that were unfavorable for Hmox1-/- bone marrow-derived proangiogenic cells (BDMC). Hmox1+/+ MSCs but not fibroblasts retained low ROS levels even after prolonged incubation with 50 µmol/L hemin, although both cell types have a comparable Hmox1 expression and similarly increase its levels in response to hemin. MSCs Hmox1-/- treated with hemin efficiently induced expression of a vast panel of antioxidant genes, especially enzymes of the glutathione pathway. Innovation and Conclusion: Hmox1 overexpression is a popular strategy to enhance viability and performance of MSCs after the transplantation. However, murine MSCs Hmox1-/- do not differ from wild-type MSCs in phenotype and functions. MSC Hmox1-/- show better resistance to hemin than fibroblasts and BDMCs and rapidly react to the stress by upregulation of quintessential genes in antioxidant response. Antioxid. Redox Signal. 00, 000-000.


Subject(s)
Heme Oxygenase-1/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Oxidative Stress , Animals , Antioxidants/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Survival/drug effects , Cytokines/biosynthesis , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Heme Oxygenase (Decyclizing)/metabolism , Hemin/toxicity , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/immunology , Mice , Mice, Knockout , Phenotype
14.
Exp Cell Res ; 361(2): 217-224, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29074371

ABSTRACT

Oxidative stress mediated secondary injury contributes to neurological deterioration after intracerebral hemorrhage (ICH). Astrocytes, the most dominant cells in the central nervous system (CNS), play key roles in maintaining redox homeostasis by providing oxidative stress defense. Hemoglobin (Hb), the primary component released by hemolysis, is an effective activator of astrocytes. Hemin, the product of Hb degradation, is highly toxic due to the induction of reactive oxygen species (ROS). We speculate that Hb-activated astrocytes are resistant to hemin-induced toxicity. To verify our speculation, Hb-pretreated astrocytes were exposed to hemin, intracellular ROS accumulation and cell apoptosis were evaluated. Heme oxygenase 1 (HO-1) and nuclear transcription factor-erythroid 2 related factor (Nrf2) expression were observed to explore the potential mechanism. The results demonstrated that Hb induced upregulation and nuclear translocation of Nrf2 in astrocytes, resulted in HO-1 upregulation, which contributed to reduced ROS accumulation and apoptosis rate. Knocking down Nrf2 expression by siRNA suppressed Hb-induced upregulation of HO-1 expression and increased the susceptibility of Hb-pretreated astrocytes to hemin-induced toxicity. Taken together, Hb-activated astrocytes acquired resistance to hemin-induced toxicity via Nrf2/HO-1 pathway. This phenomenon can be considered as the adaptive self-defense in the pathological process of ICH. Hb pre-warned astrocytes and enhanced their capability of handling the coming hemin "flood". Nrf2/HO-1 may be employed as a target for neuroprotection after ICH.


Subject(s)
Astrocytes/drug effects , Heme Oxygenase (Decyclizing)/genetics , Hemin/toxicity , Hemoglobins/pharmacology , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/pathology , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Gene Expression Regulation , Heme Oxygenase (Decyclizing)/metabolism , Hemin/antagonists & inhibitors , Models, Biological , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
15.
Neuroreport ; 28(10): 551-554, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28489664

ABSTRACT

Hemorrhagic stroke is associated with high morbidity and mortality. Hemin is a decomposition product of hemoglobin that is related to neuronal apoptosis after hemorrhage, although the molecular basis for this association remains unclear. To address this issue, the present study investigated hemin-induced changes in the apoptotic index and mitochondrial ultrastructure in SH-SY5Y cells. Cell viability was evaluated using Cell Counting Kit-8 and by terminal transferase dUTP nick-end labeling, western blotting, and flow cytometry. Changes in mitochondrial ultrastructure were examined by super-resolution three-dimensional structured illumination microscopy. We found that cleaved-caspase-3 expression and the number of apoptotic cells increased in a time-dependent manner upon hemin treatment, which was associated with mitochondrial fragmentation. Our data suggest that hemin induces apoptosis and mitochondrial fission in neuronal cells. Thus, therapeutic strategies that target hemin could mitigate the damage caused by hemorrhagic stroke.


Subject(s)
Apoptosis , Hemin/toxicity , Mitochondria/ultrastructure , Apoptosis/physiology , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , Flow Cytometry , Humans , Imaging, Three-Dimensional , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Neurons/metabolism , Neurons/ultrastructure
16.
Biochem Biophys Res Commun ; 483(3): 930-935, 2017 02 12.
Article in English | MEDLINE | ID: mdl-28082197

ABSTRACT

HBP23, a 23-kDa heme-binding protein identified in rats, is a member of the peroxiredoxin (Prx) family, the primary peroxidases involved in hydrogen peroxide catabolism. Although HBP23 has a characteristic Cys-Pro heme-binding motif, the significance of heme binding to Prx family proteins remains to be elucidated. Here, we examined the effect of heme binding to human peroxiredoxin-1 (PRX1), which has 97% amino acid identity to HBP23. PRX1 was expressed in Escherichia coli and purified to homogeneity. Spectroscopic titration demonstrated that PRX1 binds heme with a 1:1 stoichiometry and a dissociation constant of 0.17 µM. UV-vis spectra of heme-PRX1 suggested that Cys52 is the axial ligand of ferric heme. PRX1 peroxidase activity was lost upon heme binding, reflecting the fact that Cys52 is not only the heme-binding site but also the active center of peroxidase activity. Interestingly, heme binding to PRX1 caused a decrease in the toxicity and degradation of heme, significantly suppressing H2O2-dependent heme peroxidase activity and degradation of PRX1-bound heme compared with that of free hemin. By virtue of its cytosolic abundance (∼20 µM), PRX1 thus functions as a scavenger of cytosolic hemin (<1 µM). Collectively, our results indicate that PRX1 has a dual role; Cys-dependent peroxidase activity and cytosolic heme scavenger.


Subject(s)
Heme/metabolism , Peroxiredoxins/chemistry , Peroxiredoxins/metabolism , Amino Acid Substitution , Animals , Catalytic Domain/genetics , Cysteine/chemistry , Cysteine/genetics , Cytosol/enzymology , Hemin/metabolism , Hemin/toxicity , Humans , Models, Biological , Mutagenesis, Site-Directed , Peroxiredoxins/genetics , Protein Binding , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrophotometry
17.
Sci Transl Med ; 8(328): 328ra29, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26936506

ABSTRACT

Disability or death due to intracerebral hemorrhage (ICH) is attributed to blood lysis, liberation of iron, and consequent oxidative stress. Iron chelators bind to free iron and prevent neuronal death induced by oxidative stress and disability due to ICH, but the mechanisms for this effect remain unclear. We show that the hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) family of iron-dependent, oxygen-sensing enzymes are effectors of iron chelation. Molecular reduction of the three HIF-PHD enzyme isoforms in the mouse striatum improved functional recovery after ICH. A low-molecular-weight hydroxyquinoline inhibitor of the HIF-PHD enzymes, adaptaquin, reduced neuronal death and behavioral deficits after ICH in several rodent models without affecting total iron or zinc distribution in the brain. Unexpectedly, protection from oxidative death in vitro or from ICH in vivo by adaptaquin was associated with suppression of activity of the prodeath factor ATF4 rather than activation of an HIF-dependent prosurvival pathway. Together, these findings demonstrate that brain-specific inactivation of the HIF-PHD metalloenzymes with the blood-brain barrier-permeable inhibitor adaptaquin can improve functional outcomes after ICH in several rodent models.


Subject(s)
Activating Transcription Factor 4/metabolism , Brain/pathology , Intracranial Hemorrhages/pathology , Molecular Targeted Therapy , Neurons/pathology , Oxygen/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Animals , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Genes, Reporter , Hemin/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracranial Hemorrhages/physiopathology , Iron/pharmacology , Iron Chelating Agents/pharmacology , Mice , Neurons/drug effects , Neuroprotective Agents/pharmacology , Procollagen-Proline Dioxygenase/metabolism , Protein Domains , Protein Isoforms/metabolism , Rats , Recovery of Function/drug effects
18.
Neurochem Res ; 41(1-2): 298-306, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26694650

ABSTRACT

Hemin is a breakdown product of the blood protein, hemoglobin and is responsible for much of the secondary damage caused following a hemorrhagic stroke. Hemin is toxic to cultured astrocytes and it is thought that this toxicity is due to iron that is liberated when hemin is degraded. However, free iron applied to astrocytes is not toxic and the reason for this discrepancy is unknown. The present study exposed primary astrocyte cultures from neonatal mice to hemin-iron (25 µM hemin) or non-hemin iron (25 µM ferric ammonium citrate; FAC) for 12 or 24 h. Perls' and Turnbull's staining, as well as measures of cell viability and iron accumulation, were used to assess the valency, solubility and distribution of iron within cells. While cells accumulated similar amounts of iron from both sources, hemin was shown to be highly toxic to astrocytes, whereas FAC was not. Iron released by the degradation of hemin was present in both valencies (Fe(2+) and Fe(3+)), was mostly soluble and did not induce ferritin expression in most cells, whereas non-hemin iron (from FAC) was present in astrocytes almost exclusively as insoluble Fe(3+) and it induced widespread ferritin expression. These results show that the cellular mechanisms for processing hemin-iron and non-hemin iron are very different. The data suggest that hemin-iron has a greater potential to damage astrocytes by participating in unregulated redox reactions.


Subject(s)
Astrocytes/metabolism , Hemin/metabolism , Iron/metabolism , Animals , Cells, Cultured , Hemin/toxicity , Iron/toxicity , Mice , Mice, Inbred C57BL
19.
Nutr Cancer ; 68(1): 105-12, 2016.
Article in English | MEDLINE | ID: mdl-26699517

ABSTRACT

A reported linkage between processed (nitrite-treated) meat products and the incidence of colon cancer could be due to sodium nitrite (NaNO2) itself or to N-nitroso compounds produced from the nitrite. Exposure to nitrite occurs due to residual nitrite in processed meat and to salivary nitrite arising by reduction of nitrate in vegetables and drinking water. Here we tested whether NaNO2 could induce colonic aberrant crypts (ABC) or ABC foci (ACF), which are putative precursors of colon cancer. We fed NaNO2 in drinking water for 20-25 wk to groups of 8-20 adult female mice. After sacrifice, ABC and ACF were counted in 2-cm distal colonic segments. In Experiment 1, no significant differences in ABC/ACF induction were seen between groups of 13-14 A/J mice fed 0, 0.5, or 1.0 g NaNO2/l drinking water. NaNO2 also did not affect fasting blood glucose levels. In Experiment 2, we fed 0, 1.0, 1.25, or 1.5 g NaNO2/l water to groups of 15 CF-1 mice. Five of the mice fed 1.5 g NaNO2/l showed ABC, whereas all other groups showed only 0-2 ABC/group, including 0 ABC for the group fed 1.25 g NaNO2/l. Overall statistical analysis indicated a dose-response p trends of 0.04. Pairwise comparison of ABC between groups fed 1.25 and 1.5 g NaNO2/l showed p 0.02 for both ABC and ACF, but a similar comparison between the untreated and 1.5 g/l groups showed no significant effects. In Experiment 3, hot dogs (18% of diet), which were fed to CF-1 mice previously treated with azoxymethane, inhibited ABC and ACF induction, but this effect was not significant (P = 0.10-0.12). In conclusion, these results support the view that NaNO2 may be a risk factor for colon carcinogenesis.


Subject(s)
Aberrant Crypt Foci/chemically induced , Colorectal Neoplasms/chemically induced , Sodium Nitrite/toxicity , Animals , Azoxymethane/toxicity , Female , Hemin/toxicity , Mice
20.
Mol Med Rep ; 11(6): 4706-12, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25633656

ABSTRACT

Heme oxygenase­1 (HO­1) possesses significant potential as a drug target for hepatitis B, which may be transferable to patient therapy. The aim of the present study was to clarify the dynamic correlation between the hepatitis B virus (HBV) and HO­1. The levels of HBV replication and expression of HO­1 were investigated in HepG2.2.15 hepatoma cells following exposure to 5­50 µM hemin for 1­6 days. The mRNA expression levels of HO­1 were then detected using reverse transcription­quantitative polymerase chain reaction (RT­qPCR). HBV replication levels were determined by enzyme­immunoassay and a PCR­fluorescence quantitation assay. The results of the present study demonstrated that the mRNA expression levels of HO­1 increased in a dose­dependent manner in the HepG2.2.15 cells, following exposure to 5­50 µM hemin. The mRNA expression levels of HO­1 reached a peak following exposure of the cells to hemin for three days, subsequently the expression of HO­1 decreased. Following exposure to hemin at an optimal concentration of 50 µM for 1­6 days, the levels of the hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) in the cells were significantly reduced. This marked reduction in the expression of HBsAg and HBeAg reached its peak on the first day, following which the inhibition weakened as the duration of exposure increased. In addition, the inhibition of HBV DNA replication increased with the a longer duration of exposure. Furthermore, HBV DNA levels were significantly decreased following exposure to hemin for 3­6 days. In conclusion, the present study demonstrated that induced expression of HO­1 interfered with HBV replication in a dose and time­dependent manner, implying that a reduction of the HBV viral load may contribute to upregulation in the expression of HO­1.


Subject(s)
Heme Oxygenase-1/metabolism , Hepatitis B virus/physiology , DNA, Viral/metabolism , Down-Regulation/drug effects , Heme Oxygenase-1/genetics , Hemin/toxicity , Hep G2 Cells , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/metabolism , Humans , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...