Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Exp Biol Med (Maywood) ; 248(10): 897-907, 2023 05.
Article in English | MEDLINE | ID: mdl-36941786

ABSTRACT

Intravascular hemolysis results in the release of cell-free hemoglobin and heme in plasma. In sickle cell disease, the fragility of the sickle red blood cell leads to chronic hemolysis, which can contribute to oxidative damage and activation of inflammatory pathways. The scavenger proteins haptoglobin and hemopexin provide pathways to remove hemoglobin and heme, respectively, from the circulation. Heme also intercalates in membranes of blood cells and endothelial cells in the vasculature and associates with other plasma components such as albumin and lipoproteins. Hemopexin has a much higher affinity and can strip heme from the other pools and detoxify plasma from cell-free circulatory heme. However, due to chronic hemolysis, hemopexin is depleted in individuals with sickle cell disease. Thus, cell-free unbound heme is expected to accumulate in plasma. We developed a methodology for the accurate quantification of the fraction of heme, which is pathologically relevant in sickle cell disease, that does not appear to be sequestered to a plasma compartment. Our data show significant variation in the concentration of unbound heme, and rather unexpectedly, the size of the unbound fraction does not correlate to the degree of hemolysis, as measured by the concentration of bound heme. Very high heme concentrations (>150 µM) were obtained in some plasma with unbound concentrations that were several fold lower than in plasma with much lower hemolysis (<50 µM). These findings underscore the long-term effects of chronic hemolysis on the blood components and of the disruption of the essential equilibrium between release of hemoproteins/heme in the circulation and adaptative response of the scavenging/removal mechanisms. Understanding the clinical implications of this loss of response may provide insights into diagnostic and therapeutic targets in patients with sickle cell disease.


Subject(s)
Anemia, Sickle Cell , Heme , Humans , Hemolysis , Hemopexin/metabolism , Hemopexin/pharmacology , Hemopexin/therapeutic use , Endothelial Cells/metabolism , Anemia, Sickle Cell/drug therapy , Hemoglobins
2.
Cell Cycle ; 22(6): 645-665, 2023.
Article in English | MEDLINE | ID: mdl-36218263

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease, while respiratory infections can elicit exacerbations in COPD patients to mediate increased mortality. Administration of Tanshinones (TS) derivatives has been demonstrated to protect against cigarette smoking (CS) and lipopolysaccharide (LPS)-induced COPD progression. However, the underlying molecular mechanisms and the roles of TS in mitigating the severity of viral-mediated exacerbations of COPD have not been elucidated. Here, we found that TS treatments significantly attenuated lung function decline, inflammatory responses and oxidative stress in CS and LPS-induced COPD mice. Subsequent RNA-seq analysis revealed significantly upregulated Hemopexin expression and enriched interferons (IFNs) signaling pathways in lung tissues of COPD mice upon TS treatments. Moreover, TS administration demonstrated Hemopexin-dependent beneficial roles in BEAS-2B lung cells and RAW264.7 macrophages, which was associated with the suppression of oxidative stress and ERK, NF-κB, and NLRP3 inflammasome signaling pathways-mediated inflammation. Furthermore, TS promoted IFN signaling and rescued impaired antiviral responses in CS and LPS-exposed lung cells that were infected by influenza virus. Notably, hemopexin over-expression in lung cells and macrophages recapitulated the pharmacological activities of TS. Taken together, these results indicate that TS administration is a promising and potential therapeutic strategy for treating COPD and preventing COPD exacerbations.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Mice , Animals , Hemopexin/metabolism , Hemopexin/therapeutic use , Cigarette Smoking/adverse effects , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Lung/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Macrophages/metabolism , Antiviral Agents/therapeutic use
3.
Semin Nephrol ; 40(2): 148-159, 2020 03.
Article in English | MEDLINE | ID: mdl-32303278

ABSTRACT

Sepsis is a heterogeneous clinical syndrome that is complicated commonly by acute kidney injury (sepsis-AKI). Currently, no approved pharmacologic therapies exist to either prevent sepsis-AKI or to treat sepsis-AKI once it occurs. A growing body of evidence supports a connection between red blood cell biology and sepsis-AKI. Increased levels of circulating cell-free hemoglobin (CFH) released from red blood cells during hemolysis are common during sepsis and can contribute to sepsis-AKI through several mechanisms including tubular obstruction, nitric oxide depletion, oxidative injury, and proinflammatory signaling. A number of potential pharmacologic therapies targeting CFH in sepsis have been identified including haptoglobin, hemopexin, and acetaminophen, and early phase clinical trials have suggested that acetaminophen may have beneficial effects on lipid peroxidation and kidney function in patients with sepsis. Bedside measurement of CFH levels may facilitate predictive enrichment for future clinical trials of CFH-targeted therapeutics. However, rapid and reliable bedside tests for plasma CFH will be required for such trials to move forward.


Subject(s)
Acute Kidney Injury/metabolism , Hemoglobins/metabolism , Sepsis/metabolism , Acetaminophen/therapeutic use , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Acute Kidney Injury/prevention & control , Anemia, Sickle Cell/metabolism , Animals , Coronary Artery Bypass , Disseminated Intravascular Coagulation/metabolism , Eryptosis , Erythrocyte Deformability , Haptoglobins/metabolism , Haptoglobins/therapeutic use , Heme/metabolism , Hemoglobins/immunology , Hemolysis , Hemopexin/metabolism , Hemopexin/therapeutic use , Humans , Kidney Tubules , Malaria/metabolism , Nitric Oxide/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Sepsis/complications , Sepsis/immunology , Transfusion Reaction/metabolism
4.
Neurosurg Rev ; 43(5): 1273-1288, 2020 Oct.
Article in English | MEDLINE | ID: mdl-31493061

ABSTRACT

Delayed cerebral ischaemia (DCI) after aneurysmal subarachnoid haemorrhage (aSAH) is a major cause of mortality and morbidity. The pathophysiology of DCI after aSAH is thought to involve toxic mediators released from lysis of red blood cells within the subarachnoid space, including free haemoglobin and haem. Haptoglobin and hemopexin are endogenously produced acute phase proteins that are involved in the clearance of these toxic mediators. The aim of this review is to investigate the pathophysiological mechanisms involved in DCI and the role of both endogenous as well as exogenously administered haptoglobin and hemopexin in the prevention of DCI.


Subject(s)
Brain Ischemia/etiology , Brain Ischemia/prevention & control , Haptoglobins/therapeutic use , Hemopexin/therapeutic use , Subarachnoid Hemorrhage/complications , Humans
5.
PLoS One ; 13(4): e0196455, 2018.
Article in English | MEDLINE | ID: mdl-29694434

ABSTRACT

During hemolysis, hemoglobin and heme released from red blood cells promote oxidative stress, inflammation and thrombosis. Plasma haptoglobin and hemopexin scavenge free hemoglobin and heme, respectively, but can be depleted in hemolytic states. Haptoglobin and hemopexin supplementation protect tissues, including the vasculature, liver and kidneys. It is widely assumed that these protective effects are due primarily to hemoglobin and heme clearance from the vasculature. However, this simple assumption does not account for the consequent cytoprotective adaptation seen in cells and organs. To further address the mechanism, we used a hyperhemolytic murine model (Townes-SS) of sickle cell disease to examine cellular responses to haptoglobin and hemopexin supplementation. A single infusion of haptoglobin or hemopexin (± equimolar hemoglobin) in SS-mice increased heme oxygenase-1 (HO-1) in the liver, kidney and skin several fold within 1 hour and decreased nuclear NF-ĸB phospho-p65, and vaso-occlusion for 48 hours after infusion. Plasma hemoglobin and heme levels were not significantly changed 1 hour after infusion of haptoglobin or hemopexin. Haptoglobin and hemopexin also inhibited hypoxia/reoxygenation and lipopolysaccharide-induced vaso-occlusion in SS-mice. Inhibition of HO-1 activity with tin protoporphyrin blocked the protections afforded by haptoglobin and hemopexin in SS-mice. The HO-1 reaction product carbon monoxide, fully restored the protection, in part by inhibiting Weibel-Palade body mobilization of P-selectin and von Willebrand factor to endothelial cell surfaces. Thus, the mechanism by which haptoglobin and hemopexin supplementation in hyperhemolytic SS-mice induces cytoprotective cellular responses is linked to increased HO-1 activity.


Subject(s)
Anemia, Sickle Cell/prevention & control , Haptoglobins/therapeutic use , Heme Oxygenase-1/metabolism , Hemopexin/therapeutic use , Inflammation/prevention & control , Aldehydes/analysis , Anemia, Sickle Cell/pathology , Animals , Carbon Monoxide/pharmacology , Cytokines/analysis , Disease Models, Animal , Female , Gene Expression/drug effects , Haptoglobins/pharmacology , Hemopexin/pharmacology , Intercellular Adhesion Molecule-1 , Male , Metalloporphyrins/pharmacology , Mice , Microsomes, Liver/metabolism , Protoporphyrins/pharmacology , Skin/metabolism , Skin/pathology , Transcription Factor RelA/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
6.
Nefrología (Madrid) ; 38(1): 13-26, ene.-feb. 2018. ilus, graf, tab
Article in Spanish | IBECS | ID: ibc-170077

ABSTRACT

La hemoglobina y la mioglobina son hemoproteínas que juegan un papel fundamental en el organismo ya que participan en el transporte de oxígeno. Sin embargo, debido a su estructura química, estas moléculas pueden ejercer efectos deletéreos cuando se liberan al torrente sanguíneo de forma masiva, como sucede en determinadas condiciones patológicas asociadas a rabdomiólisis o hemólisis intravascular. Una vez en el plasma, estas hemoproteínas se pueden filtrar y acumular en el riñón, donde resultan citotóxicas, principalmente para el epitelio tubular, e inducen fracaso renal agudo y enfermedad renal crónica. En la presente revisión analizaremos los distintos contextos patológicos que provocan la acumulación renal de estas hemoproteínas, su relación con la pérdida de función renal a corto y largo plazo, los mecanismos fisiopatólogicos responsables de sus efectos adversos y los sistemas de defensa que contrarrestan tales acciones. Por último, describiremos los distintos tratamientos utilizados actualmente y mostraremos nuevas opciones terapéuticas basadas en la identificación de nuevas dianas celulares y moleculares, prestando especial atención a los diversos ensayos clínicos que se encuentran en marcha en la actualidad (AU)


Haemoglobin and myoglobin are haem proteins that play a key role as they help transport oxygen around the body. However, because of their chemical structure, these molecules can exert harmful effects when they are released massively into the bloodstream, as reported in certain pathological conditions associated with rhabdomyolysis or intravascular haemolysis. Once in the plasma, these haem proteins can be filtered and can accumulate in the kidney, where they become cytotoxic, particularly for the tubular epithelium, inducing acute kidney failure and chronic kidney disease. In this review, we will analyse the different pathological contexts that lead to the renal accumulation of these haem proteins, their relation to both acute and chronic loss of renal function, the pathophysiological mechanisms that cause adverse effects and the defence systems that counteract such actions. Finally, we will describe the different treatments currently used and present new therapeutic options based on the identification of new cellular and molecular targets, with particular emphasis on the numerous clinical trials that are currently ongoing (AU)


Subject(s)
Humans , Hemeproteins/adverse effects , Hemeproteins/therapeutic use , Kidney Failure, Chronic/complications , Oxidative Stress , Hemoglobinuria/etiology , Rhabdomyolysis/etiology , Renal Insufficiency, Chronic/physiopathology , Cell Death , Fibrosis/complications , Hemopexin/analysis , Hemopexin/therapeutic use
7.
J Surg Res ; 212: 15-21, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28550901

ABSTRACT

BACKGROUND: Hemopexin (HPX) has been identified as an anti-inflammatory agent, but its role in endotoxemia is unclear. The purpose of this study was to determine whether HPX suppresses systemic and lung inflammation in a mouse model of endotoxemia. MATERIALS AND METHODS: At 30 min of intraperitoneal administration of lipopolysaccharide (LPS; 10 mg/kg), either distilled water (LPS-only treated animals) or HPX (5 mg/kg) was injected into mice via the tail vein, and the survival rates were analyzed after 36 h. Furthermore, the serum levels of tumor necrosis factor-α, interleukin-6 (IL-6), and HPX were determined at 0, 3, and 6 h, and the expression levels and DNA binding activities of phosphorylated cytoplasmic inhibitor κB-α, nuclear factor-κB (NF-κB), and the p65 subunit of NF-κB were evaluated and compared with the rates of histologic lung injury after 6 h. RESULTS: Serum tumor necrosis factor-α and interleukin-6 levels were decreased in HPX-treated animals at 3 and 6 h (P < 0.05). HPX suppressed the NF-κB pathway (P < 0.05) and reduced acute lung injury at 6 h, and 36 h after initial treatment, the survival rate was higher in HPX-treated animals than that in LPS-treated animals (P < 0.05). CONCLUSIONS: HPX downregulated proinflammatory cytokine production and acute lung injury as well as improved survival rates in a mouse model of endotoxemia. These effects were associated with HPX-mediated suppression of the NF-κB pathway.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents/therapeutic use , Endotoxemia/drug therapy , Hemopexin/therapeutic use , Lipopolysaccharides/administration & dosage , Acute Lung Injury/blood , Acute Lung Injury/microbiology , Acute Lung Injury/mortality , Animals , Biomarkers/blood , Blotting, Western , Endotoxemia/blood , Endotoxemia/complications , Endotoxemia/mortality , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred BALB C , Survival Rate , Treatment Outcome
8.
Free Radic Biol Med ; 108: 452-464, 2017 07.
Article in English | MEDLINE | ID: mdl-28400318

ABSTRACT

Heart failure is a leading cause of morbidity and mortality in patients affected by different disorders associated to intravascular hemolysis. The leading factor is the presence of pathologic amount of pro-oxidant free heme in the bloodstream, due to the exhaustion of the natural heme scavenger Hemopexin (Hx). Here, we evaluated whether free heme directly affects cardiac function, and tested the therapeutic potential of replenishing serum Hx for increasing serum heme buffering capacity. The effect of heme on cardiac function was assessed in vitro, on primary cardiomyocytes and H9c2 myoblast cell line, and in vivo, in Hx-/- mice and in genetic and acquired mouse models of intravascular hemolysis. Purified Hx or anti-oxidants N-Acetyl-L-cysteine and α-tocopherol were used to counteract heme cardiotoxicity. In mice, Hx loss/depletion resulted in heme accumulation and enhanced reactive oxygen species (ROS) production in the heart, which ultimately led to severe systolic dysfunction. Similarly, high ROS reduced systolic Ca2+ transient amplitudes and fractional shortening in primary cardiomyocytes exposed to free heme. In keeping with these Ca2+ handling alterations, oxidation and CaMKII-dependent phosphorylation of Ryanodine Receptor 2 were higher in Hx-/- hearts than in controls. Administration of anti-oxidants prevented systolic failure both in vitro and in vivo. Intriguingly, Hx rescued contraction defects of heme-treated cardiomyocytes and preserved cardiac function in hemolytic mice. We show that heme-mediated oxidative stress perturbs cardiac Ca2+ homeostasis and promotes contractile dysfunction. Scavenging heme, Hx counteracts cardiac heme toxicity and preserves left ventricular function. Our data generate the rationale to consider the therapeutic use of Hx to limit the cardiotoxicity of free heme in hemolytic disorders.


Subject(s)
Heart Failure, Systolic/drug therapy , Hemoglobins/metabolism , Hemopexin/therapeutic use , Myocytes, Cardiac/metabolism , beta-Thalassemia/drug therapy , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Heart Failure, Systolic/physiopathology , Hemoglobins/genetics , Hemolysis , Hemopexin/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/pathology , Oxidative Phosphorylation , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , beta-Thalassemia/physiopathology
9.
Antioxid Redox Signal ; 24(2): 99-112, 2016 Jan 10.
Article in English | MEDLINE | ID: mdl-26376667

ABSTRACT

AIMS: Exposure to irritant gases, such as bromine (Br2), poses an environmental and occupational hazard that results in severe lung and systemic injury. However, the mechanism(s) of Br2 toxicity and the therapeutic responses required to mitigate lung damage are not known. Previously, it was demonstrated that Br2 upregulates the heme degrading enzyme, heme oxygenase-1 (HO-1). Since heme is a major inducer of HO-1, we determined whether an increase in heme and heme-dependent oxidative injury underlies the pathogenesis of Br2 toxicity. RESULTS: C57BL/6 mice were exposed to Br2 gas (600 ppm, 30 min) and returned to room air. Thirty minutes postexposure, mice were injected intraperitoneally with a single dose of the heme scavenging protein, hemopexin (Hx) (3 µg/gm body weight), or saline. Twenty-four hours postexposure, saline-treated mice had elevated total heme in bronchoalveolar lavage fluid (BALF) and plasma and acute lung injury (ALI) culminating in 80% mortality after 10 days. Hx treatment significantly lowered heme, decreased evidence of ALI (lower protein and inflammatory cells in BALF, lower lung wet-to-dry weight ratios, and decreased airway hyperreactivity to methacholine), and reduced mortality. In addition, Br2 caused more severe ALI and mortality in mice with HO-1 gene deletion (HO-1-/-) compared to wild-type controls, while transgenic mice overexpressing the human HO-1 gene (hHO-1) showed significant protection. INNOVATION: This is the first study delineating the role of heme in ALI caused by Br2. CONCLUSION: The data suggest that attenuating heme may prove to be a useful adjuvant therapy to treat patients with ALI.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Bromine , Heme/metabolism , Hemopexin , Lung/drug effects , Lung/metabolism , Acute Lung Injury/metabolism , Animals , Female , Hemopexin/pharmacology , Hemopexin/therapeutic use , Male , Mice , Mice, Inbred C57BL
10.
Blood ; 127(4): 473-86, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26675351

ABSTRACT

Hemolytic diseases, such as sickle cell anemia and thalassemia, are characterized by enhanced release of hemoglobin and heme into the circulation, heme-iron loading of reticulo-endothelial system macrophages, and chronic inflammation. Here we show that in addition to activating the vascular endothelium, hemoglobin and heme excess alters the macrophage phenotype in sickle cell disease. We demonstrate that exposure of cultured macrophages to hemolytic aged red blood cells, heme, or iron causes their functional phenotypic change toward a proinflammatory state. In addition, hemolysis and macrophage heme/iron accumulation in a mouse model of sickle disease trigger similar proinflammatory phenotypic alterations in hepatic macrophages. On the mechanistic level, this critically depends on reactive oxygen species production and activation of the Toll-like receptor 4 signaling pathway. We further demonstrate that the heme scavenger hemopexin protects reticulo-endothelial macrophages from heme overload in heme-loaded Hx-null mice and reduces production of cytokines and reactive oxygen species. Importantly, in sickle mice, the administration of human exogenous hemopexin attenuates the inflammatory phenotype of macrophages. Taken together, our data suggest that therapeutic administration of hemopexin is beneficial to counteract heme-driven macrophage-mediated inflammation and its pathophysiologic consequences in sickle cell disease.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/immunology , Anti-Inflammatory Agents/therapeutic use , Heme/immunology , Hemopexin/therapeutic use , Macrophages/immunology , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Animals , Cell Line , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Gene Deletion , Hemopexin/genetics , Humans , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Knockout , Reactive Oxygen Species/immunology , Toll-Like Receptor 4/immunology
11.
J Clin Invest ; 123(11): 4809-20, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24084741

ABSTRACT

The prevention and treatment of acute chest syndrome (ACS) is a major clinical concern in sickle cell disease (SCD). However, the mechanism underlying the pathogenesis of ACS remains elusive. We tested the hypothesis that the hemolysis byproduct hemin elicits events that induce ACS. Infusion of a low dose of hemin caused acute intravascular hemolysis and autoamplification of extracellular hemin in transgenic sickle mice, but not in sickle-trait littermates. The sickle mice developed multiple symptoms typical of ACS and succumbed rapidly. Pharmacologic inhibition of TLR4 and hemopexin replacement therapy prior to hemin infusion protected sickle mice from developing ACS. Replication of the ACS-like phenotype in nonsickle mice revealed that the mechanism of lung injury due to extracellular hemin is independent of SCD. Using genetic and bone marrow chimeric tools, we confirmed that TLR4 expressed in nonhematopoietic vascular tissues mediated this lethal type of acute lung injury. Respiratory failure was averted after the onset of ACS-like symptoms in sickle mice by treating them with recombinant hemopexin. Our results reveal a mechanism that helps to explain the pathogenesis of ACS, and we provide proof of principle for therapeutic strategies to prevent and treat this condition in mice.


Subject(s)
Acute Chest Syndrome/etiology , Anemia, Sickle Cell/complications , Hemin/metabolism , Acute Chest Syndrome/blood , Acute Chest Syndrome/prevention & control , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Animals , Disease Models, Animal , Extracellular Space/metabolism , Hemoglobin, Sickle/genetics , Hemoglobin, Sickle/metabolism , Hemolysis , Hemopexin/therapeutic use , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Recombinant Proteins/therapeutic use , Sickle Cell Trait/blood , Sickle Cell Trait/complications , Sickle Cell Trait/genetics , Sulfonamides/pharmacology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
12.
BMC Neurosci ; 14: 58, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23758755

ABSTRACT

BACKGROUND: The plasma protein hemopexin (HPX) exhibits the highest binding affinity to free heme. In vitro experiments and gene-knock out technique have suggested that HPX may have a neuroprotective effect. However, the expression of HPX in the brain was not well elucidated and its expression after cerebral ischemia-reperfusion injury was also poorly studied. Furthermore, no in vivo data were available on the effect of HPX given centrally on the prognosis of focal cerebral ischemia. RESULTS: In the present study, we systematically investigated expression of HPX in normal rat brain by immunofluorescent staining. The results showed that HPX was mainly expressed in vascular system and neurons, as well as in a small portion of astrocytes adjacent to the vessels in normal rat brain. Further, we determined the role of HPX in the process of focal cerebral ischemic injury and explored the effects of HPX treatment in a rat model of transient focal cerebral ischemia. After 2 h' middle cerebral artery occlusion (MCAO) followed by 24 h' reperfusion, the expression of HPX was increased in the neurons and astrocytes in the penumbra area, as demonstrated by immunohistochemistry and Western blot techniques. Intracerebroventricular injection of HPX at the onset of reperfusion dose-dependently reduced the infarct volumes and improved measurements of neurological function of the rat subjected to transient focal cerebral ischemia. The neuroprotective effects of HPX sustained for up to 7 days after experiments. CONCLUSIONS: Our study provides a new insight into the potential neuroprotective role of HPX as a contributing factor of endogenous protective mechanisms against focal cerebral ischemia injury, and HPX might be developed as a potential agent for treatment of ischemic stroke.


Subject(s)
Hemopexin/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Reperfusion Injury/prevention & control , Animals , Brain/metabolism , Brain/pathology , Brain Edema/etiology , Brain Edema/prevention & control , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Hemopexin/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Injections, Intraventricular , Male , Neuroglia/metabolism , Neurologic Examination , Neurons/metabolism , Neuroprotective Agents/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Time Factors
13.
Circulation ; 127(12): 1317-29, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23446829

ABSTRACT

BACKGROUND: Hemolytic diseases are characterized by enhanced intravascular hemolysis resulting in heme-catalyzed reactive oxygen species generation, which leads to endothelial dysfunction and oxidative damage. Hemopexin (Hx) is a plasma heme scavenger able to prevent endothelial damage and tissue congestion in a model of heme overload. Here, we tested whether Hx could be used as a therapeutic tool to counteract heme toxic effects on the cardiovascular system in hemolytic diseases. METHODS AND RESULTS: By using a model of heme overload in Hx-null mice, we demonstrated that heme excess in plasma, if not bound to Hx, promoted the production of reactive oxygen species and the induction of adhesion molecules and caused the reduction of nitric oxide availability. Then, we used ß-thalassemia and sickle cell disease mice as models of hemolytic diseases to evaluate the efficacy of an Hx-based therapy in the treatment of vascular dysfunction related to heme overload. Our data demonstrated that Hx prevented heme-iron loading in the cardiovascular system, thus limiting the production of reactive oxygen species, the induction of adhesion molecules, and the oxidative inactivation of nitric oxide synthase/nitric oxide, and promoted heme recovery and detoxification by the liver mainly through the induction of heme oxygenase activity. Moreover, we showed that in sickle cell disease mice, endothelial activation and oxidation were associated with increased blood pressure and altered cardiac function, and the administration of exogenous Hx was found to almost completely normalize these parameters. CONCLUSIONS: Hemopexin treatment is a promising novel therapy to protect against heme-induced cardiovascular dysfunction in hemolytic disorders.


Subject(s)
Anemia, Sickle Cell/drug therapy , Cardiovascular System/physiopathology , Endothelium, Vascular/physiopathology , Heme/adverse effects , Hemopexin/therapeutic use , beta-Thalassemia/drug therapy , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/physiopathology , Animals , Cardiovascular System/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Heme/metabolism , Hemopexin/genetics , Hemopexin/pharmacology , Mice , Mice, Knockout , Mice, SCID , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Treatment Outcome , beta-Thalassemia/metabolism , beta-Thalassemia/physiopathology
14.
Blood ; 121(8): 1276-84, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23264591

ABSTRACT

Hemolysis occurs in many hematologic and nonhematologic diseases. Extracellular hemoglobin (Hb) has been found to trigger specific pathophysiologies that are associated with adverse clinical outcomes in patients with hemolysis, such as acute and chronic vascular disease, inflammation, thrombosis, and renal impairment. Among the molecular characteristics of extracellular Hb, translocation of the molecule into the extravascular space, oxidative and nitric oxide reactions, hemin release, and molecular signaling effects of hemin appear to be the most critical. Limited clinical experience with a plasma-derived haptoglobin (Hp) product in Japan and more recent preclinical animal studies suggest that the natural Hb and the hemin-scavenger proteins Hp and hemopexin have a strong potential to neutralize the adverse physiologic effects of Hb and hemin. This includes conditions that are as diverse as RBC transfusion, sickle cell disease, sepsis, and extracorporeal circulation. This perspective reviews the principal mechanisms of Hb and hemin toxicity in different disease states, updates how the natural scavengers efficiently control these toxic moieties, and explores critical issues in the development of human plasma-derived Hp and hemopexin as therapeutics for patients with excessive intravascular hemolysis.


Subject(s)
Haptoglobins/therapeutic use , Hematologic Diseases/blood , Hematologic Diseases/drug therapy , Hemolysis/drug effects , Hemolysis/physiology , Hemopexin/therapeutic use , Animals , Haptoglobins/metabolism , Hemin/metabolism , Hemoglobins/metabolism , Hemopexin/metabolism , Humans
15.
Clin Cancer Res ; 11(2 Pt 1): 768-76, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15701867

ABSTRACT

PURPOSE: There is an urgent need for modalities that can localize and prolong the administration of the antitumor agents, particularly antiangiogenic, to achieve long-term tumor inhibition. However, one of the major obstacles is designing a device in which the biological activity of sensitive endogenous inhibitors is retained. We have designed a biodegradable polymeric device, which provides a unique and practical means of localizing and continuously delivering hemopexin (PEX) or platelet factor 4 fragment (PF-4/CTF) at the tumor site while maintaining their biological activity. The potential and efficacy of this system is shown in vitro and in vivo in a human glioma mouse model. EXPERIMENTAL DESIGN: Polymeric microspheres made of poly(lactic-co-glycolic acid) (PLGA) were loaded with very low amounts of PEX and PF-4/CTF. The release profiles of these factors from PLGA and their biological activity were confirmed in vitro using proliferation assays done on endothelial and tumor cells. Tumor inhibition using this system was studied in nude mice bearing a human s.c. glioma. RESULTS: PEX and PF-4/CTF released in vitro from PLGA microspheres were biologically active and significantly inhibited the proliferation of human umbilical vein endothelial cells, bovine capillary endothelial cells, and U87-MG cells. A single local s.c. injection of PLGA microspheres loaded with low amounts of PEX or PF-4/CTF resulted in an 88% and 95% reduction in glioma tumor volume 30 days post-treatment. Immunohistochemical analysis of the treated tumors showed a marked decrease in tumor vessel density compared with untreated tumors. CONCLUSION: Our findings show that polymeric microspheres are a very promising approach to locally and efficiently deliver endogenous inhibitors to the tumor site leading to a significant inhibition of the tumor.


Subject(s)
Biocompatible Materials , Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioblastoma/drug therapy , Hemopexin/therapeutic use , Lactic Acid , Platelet Factor 4/therapeutic use , Polyglycolic Acid , Polymers , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cattle , Coagulants/therapeutic use , Endothelial Cells/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microspheres , Peptide Fragments/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...