Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
BMC Anesthesiol ; 19(1): 13, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30646866

RESUMO

BACKGROUND: Ischemia-reperfusion (I/R) is a critical pathophysiological basis of cognitive dysfunction caused by ischemia stroke. Heme-oxygenase-1 (HO-1) is the rate-limiting enzyme for the elimination of excessive free heme by combining with hemopexin (HPX), a plasma protein that contributes to eliminating excessive free heme during ischemia stroke. This study aimed to elucidate whether HPX could alleviate cognitive dysfunction in rats subjected to cerebral I/R. METHODS: Rats were randomly divided into five groups: sham, MCAO, Vehicle, HPX and HPX + protoporphyrin IX (ZnPPIX). Cerebral I/R was induced by MCAO. Saline, vehicle, HPX and HPX + ZnPPIX were injected intracerebroventricularly at the moment after reperfusion. Morris water maze (MWM) test was used to detect the learning and cognitive function. Western blot was used to detect the expression of HO-1 in ischemic penumbra. CD31/vWF double labeling immunofluorescence was used to detect the neovascularization in the penumbra hippocampus. The structure and function of blood-brain barrier (BBB) was detected by the permeability of Evans Blue (EB), water content of the brain tissue, the Ang1/Ang2 and VE-cadherin expression. RESULTS: Our study verified that HPX improved the learning and memory capacity. Hemopexin up-regulated HO-1 protein expression, the average vessel density in the penumbra hippocampus and the VE- cadherin expression but decreased the permeability of EB, the water content of brain tissue and the ratio of Ang1/Ang2. The effects were reversed by ZnPPIX, an inhibitor of HO-1. CONCLUSION: HPX can maintain the integrity of the blood-brain barrier and alleviate cognitive dysfunction after cerebral I/R through the HO-1 pathway.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle , Hemopexina/administração & dosagem , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Heme Oxigenase-1/metabolismo , Hemopexina/farmacologia , Masculino , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia
2.
BMC Anesthesiol ; 18(1): 2, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29298658

RESUMO

BACKGROUND: Ischemia-reperfusion (I/R) is a critical pathophysiological change of ischemic stroke. Heme-oxygenase-1 (HO-1) is a rate-limiting enzyme of eliminating excessive free heme by combining with hemopexin (HPX), a plasma protein contributing to alleviating infarct size due to ischemia stroke. This study was to investigate whether HPX could improve angiogenesis after cerebral ischemia-reperfusion via up-regulating HO-1. METHODS: Rats were randomly divided into five groups: sham, MCAO, MCAO + Vehicle, MCAO + HPX and MCAO + HPX + protoporphyrin IX (ZnPPIX, an HO-1 inhibitor). Cerebral I/R was induced by MCAO. Saline, vehicle, HPX and HPX + ZnPPIX were respectively given to MCAO group, MCAO + Vehicle group, MCAO + HPX group and MCAO + HPX + ZnPPIX group at the moment after reperfusion by intracerebroventricular injection. Neurological behavioral scores(NBS) was assessed at 24 h and 7d after I/R. Real-time polymerase chain reaction (RT-PCR) was used to analyze the mRNA level of HO-1. Angiogenesis in penumbra area was assessed by immunofluorescence detection at 7d after I/R. Serum endothelial nitric oxide synthase (eNOS) was assessed by enzyme linked immunosorbent assay (ELISA) at 24 h and 7d after I/R. RESULTS: Compared with sham group, the NBS and the mRNA levels of HO-1 at 24 h and 7d after I/R in MCAO group decreased notably (P < 0.05), the new vessel density in ischemia penumbra increased notably at 7d after I/R (P < 0.05), the serum eNOS level increased at 24 h and 7d after I/R (P < 0.05). MCAO group and MCAO + Vehicle group showed no significant differences (P > 0.05). In the MCAO + HPX group, compared with MCAO + Vehicle group, the NBS and the mRNA levels of HO-1 increased drastically at 24 h and 7d after I/R (P < 0.05), the new vessel density in ischemia penumbra increased significantly at 7d after I/R (P < 0.05), the serum eNOS level at 24 h and 7d after I/R ascended notably (P < 0.05). Compared with MCAO + HPX group, the NBS assessment, new vessel density and serum eNOS level decreased at corresponding time points after I/R in MCAO + HPX+ ZnPPIX group (P < 0.05). CONCLUSION: HPX can promote angiogenesis after cerebral ischemia-reperfusion injury in rats via up-regulating HO-1.


Assuntos
Heme Oxigenase-1/metabolismo , Hemopexina/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Indutores da Angiogênese/farmacologia , Animais , Sinergismo Farmacológico , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/biossíntese , Hemopexina/administração & dosagem , Infarto da Artéria Cerebral Média , Infusões Intraventriculares , Masculino , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo III/sangue , Protoporfirinas/administração & dosagem , Protoporfirinas/farmacologia , Ratos , Traumatismo por Reperfusão/sangue , Regulação para Cima/efeitos dos fármacos
3.
Circulation ; 134(13): 945-60, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27515135

RESUMO

BACKGROUND: Extracellular hemoglobin and cell-free heme are toxic breakdown products of hemolyzed erythrocytes. Mammals synthesize the scavenger proteins haptoglobin and hemopexin, which bind extracellular hemoglobin and heme, respectively. Transfusion of packed red blood cells is a lifesaving therapy for patients with hemorrhagic shock. Because erythrocytes undergo progressive deleterious morphological and biochemical changes during storage, transfusion of packed red blood cells that have been stored for prolonged intervals (SRBCs; stored for 35-40 days in humans or 14 days in mice) increases plasma levels of cell-free hemoglobin and heme. Therefore, in patients with hemorrhagic shock, perfusion-sensitive organs such as the kidneys are challenged not only by hypoperfusion but also by the high concentrations of plasma hemoglobin and heme that are associated with the transfusion of SRBCs. METHODS: To test whether treatment with exogenous human haptoglobin or hemopexin can ameliorate adverse effects of resuscitation with SRBCs after 2 hours of hemorrhagic shock, mice that received SRBCs were given a coinfusion of haptoglobin, hemopexin, or albumin. RESULTS: Treatment with haptoglobin or hemopexin but not albumin improved the survival rate and attenuated SRBC-induced inflammation. Treatment with haptoglobin retained free hemoglobin in the plasma and prevented SRBC-induced hemoglobinuria and kidney injury. In mice resuscitated with fresh packed red blood cells, treatment with haptoglobin, hemopexin, or albumin did not cause harmful effects. CONCLUSIONS: In mice, the adverse effects of transfusion with SRBCs after hemorrhagic shock are ameliorated by treatment with either haptoglobin or hemopexin. Haptoglobin infusion prevents kidney injury associated with high plasma hemoglobin concentrations after resuscitation with SRBCs. Treatment with the naturally occurring human plasma proteins haptoglobin or hemopexin may have beneficial effects in conditions of severe hemolysis after prolonged hypotension.


Assuntos
Eritrócitos/efeitos dos fármacos , Haptoglobinas/farmacologia , Hemopexina/farmacologia , Animais , Proteínas Sanguíneas/farmacologia , Eritrócitos/metabolismo , Haptoglobinas/administração & dosagem , Hemopexina/administração & dosagem , Humanos , Inflamação/tratamento farmacológico , Camundongos , Ressuscitação/métodos , Choque Hemorrágico/metabolismo , Reação Transfusional
4.
Am J Physiol Heart Circ Physiol ; 312(6): H1120-H1127, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28314763

RESUMO

Intravascular hemolysis produces injury in a variety of human diseases including hemoglobinopathies, malaria, and sepsis. The adverse effects of increased plasma hemoglobin are partly mediated by depletion of nitric oxide (NO) and result in vasoconstriction. Circulating plasma proteins haptoglobin and hemopexin scavenge extracellular hemoglobin and cell-free heme, respectively. The ability of human haptoglobin or hemopexin to inhibit the adverse effects of NO scavenging by circulating murine hemoglobin was tested in C57Bl/6 mice. In healthy awake mice, the systemic hemodynamic effects of intravenous coinfusion of cell-free hemoglobin and exogenous haptoglobin or of cell-free hemoglobin and hemopexin were compared with the hemodynamic effects of infusion of cell-free hemoglobin or control protein (albumin) alone. We also studied the hemodynamic effects of infusing hemoglobin and haptoglobin as well as injecting either hemoglobin or albumin alone in mice fed a high-fat diet (HFD) and in diabetic (db/db) mice. Coinfusion of a 1:1 weight ratio of haptoglobin but not hemopexin with cell-free hemoglobin prevented hemoglobin-induced systemic hypertension in healthy awake mice. In mice fed a HFD and in diabetic mice, coinfusion of haptoglobin mixed with an equal mass of cell-free hemoglobin did not reverse hemoglobin-induced hypertension. Haptoglobin retained cell-free hemoglobin in plasma, but neither haptoglobin nor hemopexin affected the ability of hemoglobin to scavenge NO ex vivo. In conclusion, in healthy C57Bl/6 mice with normal endothelium, coadministration of haptoglobin but not hemopexin with cell-free hemoglobin prevents acute hemoglobin-induced systemic hypertension by compartmentalizing cell-free hemoglobin in plasma. In murine diseases associated with endothelial dysfunction, haptoglobin therapy appears to be insufficient to prevent hemoglobin-induced vasoconstriction.NEW & NOTEWORTHY Coadministraton of haptoglobin but not hemopexin with cell-free hemoglobin prevents hemoglobin-induced systemic hypertension in mice with a normal endothelium. In contrast, treatment with the same amount of haptoglobin is unable to prevent hemoglobin-induced vasoconstriction in mice with hyperlipidemia or diabetes mellitus, disorders that are associated with endothelial dysfunction.


Assuntos
Anti-Hipertensivos/farmacologia , Endotélio Vascular/efeitos dos fármacos , Haptoglobinas/farmacologia , Hemoglobinas , Hemopexina/farmacologia , Hipertensão/prevenção & controle , Vasoconstrição/efeitos dos fármacos , Animais , Anti-Hipertensivos/administração & dosagem , Diabetes Mellitus/fisiopatologia , Dieta Hiperlipídica , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Haptoglobinas/administração & dosagem , Hemopexina/administração & dosagem , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Infusões Intravenosas , Rim/metabolismo , Rim/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Fatores de Tempo
5.
Haematologica ; 100(3): 308-14, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25596265

RESUMO

An excess of free heme is present in the blood during many types of hemolytic anemia. This has been linked to organ damage caused by heme-mediated oxidative stress and vascular inflammation. We investigated the mechanism of heme-induced coagulation activation in vivo. Heme caused coagulation activation in wild-type mice that was attenuated by an anti-tissue factor antibody and in mice expressing low levels of tissue factor. In contrast, neither factor XI deletion nor inhibition of factor XIIa-mediated factor XI activation reduced heme-induced coagulation activation, suggesting that the intrinsic coagulation pathway is not involved. We investigated the source of tissue factor in heme-induced coagulation activation. Heme increased the procoagulant activity of mouse macrophages and human PBMCs. Tissue factor-positive staining was observed on leukocytes isolated from the blood of heme-treated mice but not on endothelial cells in the lungs. Furthermore, heme increased vascular permeability in the mouse lungs, kidney and heart. Deletion of tissue factor from either myeloid cells, hematopoietic or endothelial cells, or inhibition of tissue factor expressed by non-hematopoietic cells did not reduce heme-induced coagulation activation. However, heme-induced activation of coagulation was abolished when both non-hematopoietic and hematopoietic cell tissue factor was inhibited. Finally, we demonstrated that coagulation activation was partially attenuated in sickle cell mice treated with recombinant hemopexin to neutralize free heme. Our results indicate that heme promotes tissue factor-dependent coagulation activation and induces tissue factor expression on leukocytes in vivo. We also demonstrated that free heme may contribute to thrombin generation in a mouse model of sickle cell disease.


Assuntos
Anemia Hemolítica/genética , Anemia Falciforme/genética , Coagulação Sanguínea/efeitos dos fármacos , Heme/administração & dosagem , Tromboplastina/genética , Anemia Hemolítica/sangue , Anemia Hemolítica/induzido quimicamente , Anemia Hemolítica/patologia , Anemia Falciforme/sangue , Anemia Falciforme/patologia , Animais , Anticorpos/farmacologia , Coagulação Sanguínea/genética , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Fator XI/genética , Fator XI/metabolismo , Fator XIIa/antagonistas & inibidores , Fator XIIa/genética , Fator XIIa/metabolismo , Feminino , Deleção de Genes , Expressão Gênica , Hemopexina/farmacologia , Humanos , Injeções Intravenosas , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Suínos , Tromboplastina/antagonistas & inibidores , Tromboplastina/metabolismo
6.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(5): 717-21, 743, 2015 Sep.
Artigo em Zh | MEDLINE | ID: mdl-26619542

RESUMO

OBJECTIVE: To explore the effects of the Hemopexin (Hx) on the generation of free radicals and brain edema after intracerebral hemorrhage (ICH). METHODS: One hundred SD rats were randomly and evenly divided into four groups (25 rats in each group) which named Sham group, ICH control group, Hx removal group and Hx intervention group respectively. There were five observation points (1 d, 3 d, 7 d, 14 d, 21 d) and which contain five rats for each. The stereotactic injection technique was used to make the ICH model, which adopted rat autologous Whole blood that was removed or mixed with Hx and then injected to the right caudate nucleus of the brain. Sham group were only injected with 50 µL saline to the right caudate nucleus and ICH control group were injected with 50 pL autologous whole blood; Hx removal group were injected 50 µL autologous whole blood of removal Hx and Hx intervention group were injected 50 µL autologous whole blood which contain 0. 25 mg (5 µg/ µL) Hx. Bederson's method was applied to evaluate whether the model was established successfully or not. Garcia' s method was used to estimate the neurological dysfunction scores by. Water contents of brain tissue around the hematoma was detected by dry-wet weigh method . The superoxide dismutase (SOD) activity were measured with the xanthine oxidase method. The content of the malonyldialdehyde (MDA) was measured by the thiobarbituric acid method. Pathological changes of brain tissue around the hematoma were detected by immunohistochemical method at each observation time points; and the immunohistochemical scores result was judged by the double semiquantitative evaluation method. RESULTS: Compared with Sham group, at 3-21 d, there were statistically significant differences (P<0. 05) in the neurological disorders and water content of the brain tissue and immunohistochemistry scores within ICH control group, Hx intervention group and Hx removal group. Compared with Sham group, at 1-21 d, there were statistically significant differences (P<0. 05) in SOD activity and the content of the MDA within ICH control group, Hx intervention group and Hx removal group. All the indexes above were superior in Hx intervention group to ICH control group (P<0. 05), and inferior in Hx removal group to ICH control group (P< 0. 05). CONCLUSION: The Hemopexin may attenuate the generation of the free radicals and encephalaedema in the brain tissue around the hematoma after intracerebral hemorrhage.


Assuntos
Edema Encefálico/terapia , Encéfalo/patologia , Hemorragia Cerebral/patologia , Radicais Livres/metabolismo , Hemopexina/farmacologia , Animais , Hematoma/patologia , Imuno-Histoquímica , Malondialdeído/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Circulation ; 127(12): 1317-29, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23446829

RESUMO

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.


Assuntos
Anemia Falciforme/tratamento farmacológico , Sistema Cardiovascular/fisiopatologia , Endotélio Vascular/fisiopatologia , Heme/efeitos adversos , Hemopexina/uso terapêutico , Talassemia beta/tratamento farmacológico , Anemia Falciforme/metabolismo , Anemia Falciforme/fisiopatologia , Animais , Sistema Cardiovascular/efeitos dos fármacos , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Heme/metabolismo , Hemopexina/genética , Hemopexina/farmacologia , Camundongos , Camundongos Knockout , Camundongos SCID , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Resultado do Tratamento , Talassemia beta/metabolismo , Talassemia beta/fisiopatologia
8.
Exp Biol Med (Maywood) ; 248(13): 1103-1111, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37452705

RESUMO

Heme is a fundamental molecule for several biological processes, but when released in the extracellular space such as in hemolytic diseases, it can be toxic to cells and tissues. Hemopexin (HPX) is a circulating protein responsible for removing free heme from the circulation, whose levels can be severely depleted in conditions such as sickle cell diseases. Accordingly, increasing HPX levels represents an attractive strategy to mitigate the deleterious effects of heme in these conditions. Gene transfer of liver-produced proteins with adeno-associated virus (AAV) has been shown to be an effective and safety strategy in animal and human studies mainly in hemophilia. Here, we report the feasibility of increasing HPX levels using an AAV8 vector expressing human HPX (hHPX). C57Bl mice were injected with escalating doses of our vector, and expression was assessed by enzyme immunoassay (ELISA), Western blot, and quantitative polymerase chain reaction (qPCR). In addition, the biological activity of transgenic hHPX was confirmed using two different models of heme challenge consisting of serial heme injections or phenylhydrazine-induced hemolysis. Sustained expression of hHPX was confirmed for up to 26 weeks in plasma. Expression was dose-dependent and not associated with clinical signs of toxicity. hHPX levels were significantly reduced by heme infusions and phenylhydrazine-induced hemolysis. No clinical toxicity or laboratory signs of liver damage were observed in preliminary short-term heme challenge studies. Our results confirm that long-term expression of hHPX is feasible and safe in mice, even in the presence of heme overload. Additional studies are needed to explore the effect of transgenic HPX protein in animal models of chronic hemolysis.


Assuntos
Heme , Hemopexina , Camundongos , Humanos , Animais , Hemopexina/genética , Hemopexina/metabolismo , Hemopexina/farmacologia , Hemólise , Estudos de Viabilidade , Fatores de Transcrição , Fenil-Hidrazinas
9.
Exp Biol Med (Maywood) ; 248(10): 897-907, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36941786

RESUMO

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.


Assuntos
Anemia Falciforme , Heme , Humanos , Hemólise , Hemopexina/metabolismo , Hemopexina/farmacologia , Hemopexina/uso terapêutico , Células Endoteliais/metabolismo , Anemia Falciforme/tratamento farmacológico , Hemoglobinas
10.
Diabetes ; 72(12): 1841-1852, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37722135

RESUMO

Hemopexin (HPX) is overexpressed in the retina of patients with diabetes and induces the breakdown of the blood-retinal barrier in vitro. The aim of this study was to evaluate whether HPX blockade by specific antibodies (aHPX) could avoid vascular leakage in vivo and microvascular angiogenesis in vitro and ex vivo. For this purpose, the effect of intravitreal (IVT) injections of aHPX on vascular leakage was evaluated in db/db mice and rats with streptozotocin-induced diabetes using the Evans Blue method. Retinal neurodegeneration and inflammation were also evaluated. The antiangiogenic effect of aHPX on human retinal endothelial cells (HRECs) was tested by scratch wound healing and tube formation using standardized methods, as well as by choroidal sprouting assays from retinal explants obtained in rats. We found that IVT injection of aHPX significantly reduced vascular leakage, retinal neurodegeneration, and inflammation. In addition, treatment with aHPX significantly reduced HREC migration and tube formation induced by high glucose concentration and suppressed choroidal sprouting even after vascular endothelial growth factor stimulation, with this effect being higher than obtained with bevacizumab. The antipermeability and antiangiogenic effects of IVT injection of aHPX suggest the blockade or inhibition of HPX as a new strategy for the treatment of advanced stages of diabetic retinopathy. ARTICLE HIGHLIGHTS: Hemopexin (HPX) is the best-characterized permeability factor in steroid-sensitive nephrotic syndrome. We have previously reported that HPX is overexpressed in the retina of patients with diabetes and induces the breakdown of the blood-retinal barrier in vitro. Here, we report that intravitreal injection of anti-HPX antibodies significantly reduces vascular leakage, retinal neurodegeneration, and inflammation in diabetic murine models and that the immunoneutralization of HPX exerts a significant antiangiogenic effect in vitro and in retinal explants. The blockade of HPX can be considered as a new therapy for advanced stages of diabetic retinopathy.


Assuntos
Diabetes Mellitus Experimental , Retinopatia Diabética , Ratos , Humanos , Camundongos , Animais , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/metabolismo , Hemopexina/metabolismo , Hemopexina/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Células Endoteliais/metabolismo , Retina/metabolismo , Barreira Hematorretiniana/metabolismo , Anticorpos/farmacologia , Diabetes Mellitus Experimental/metabolismo , Inflamação/metabolismo
11.
Sci Adv ; 8(51): eadc9245, 2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36563141

RESUMO

Anthracyclines such as doxorubicin (Dox) are effective chemotherapies, but their use is limited by cardiac toxicity. We hypothesized that plasma proteomics in women with breast cancer could identify new mechanisms of anthracycline cardiac toxicity. We measured changes in 1317 proteins in anthracycline-treated patients (n = 30) and replicated key findings in a second cohort (n = 31). An increase in the heme-binding protein hemopexin (Hpx) 3 months after anthracycline initiation was associated with cardiac toxicity by echocardiography. To assess the functional role of Hpx, we administered Hpx to wild-type (WT) mice treated with Dox and observed improved cardiac function. Conversely, Hpx-/- mice demonstrated increased Dox cardiac toxicity compared to WT mice. Initial mechanistic studies indicate that Hpx is likely transported to the heart by circulating monocytes/macrophages and that Hpx may mitigate Dox-induced ferroptosis to confer cardioprotection. Together, these observations suggest that Hpx induction represents a compensatory response during Dox treatment.


Assuntos
Antraciclinas , Cardiotoxicidade , Animais , Feminino , Camundongos , Antraciclinas/toxicidade , Antibióticos Antineoplásicos , Cardiotoxicidade/etiologia , Doxorrubicina , Hemopexina/metabolismo , Hemopexina/farmacologia
12.
Front Immunol ; 13: 901876, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35935964

RESUMO

Hemolysis, as a result of disease or exposure to biomaterials, is characterized by excess amounts of cell-free heme intravascularly and consumption of the protective heme-scavenger proteins in plasma. The liberation of heme has been linked to the activation of inflammatory systems, including the complement system, through alternative pathway activation. Here, we investigated the impact of heme on the regulatory function of the complement system. Heme dose-dependently inhibited factor I-mediated degradation of soluble and surface-bound C3b, when incubated in plasma or buffer with complement regulatory proteins. Inhibition occurred with factor H and soluble complement receptor 1 as co-factors, and the mechanism was linked to the direct heme-interaction with factor I. The heme-scavenger protein hemopexin was the main contaminant in purified factor I preparations. This led us to identify that hemopexin formed a complex with factor I in normal human plasma. These complexes were significantly reduced during acute vasoocclusive pain crisis in patients with sickle cell disease, but the complexes were normalized at their baseline outpatient clinic visit. Hemopexin exposed a protective function of factor I activity in vitro, but only when it was present before the addition of heme. In conclusion, we present a mechanistic explanation of how heme promotes uncontrolled complement alternative pathway amplification by interfering with the regulatory capacity of factor I. Reduced levels of hemopexin and hemopexin-factor I complexes during an acute hemolytic crisis is a risk factor for heme-mediated factor I inhibition.


Assuntos
Anemia Falciforme , Hemopexina , Anemia Falciforme/metabolismo , Fator I do Complemento , Fibrinogênio , Heme/metabolismo , Hemopexina/farmacologia , Humanos
13.
Biosensors (Basel) ; 12(10)2022 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-36291022

RESUMO

Matrix metalloproteinases (MMPs) are essential proteins acting directly in the breakdown of the extra cellular matrix and so in cancer invasion and metastasis. Given its impact on tumor angiogenesis, monitoring MMP-14 provides strategic insights on cancer severity and treatment. In this work, we report a new approach to improve the electrochemical interaction of the MMP-14 with the electrode surface while preserving high specificity. This is based on the detection of the hemopexin (PEX) domain of MMP-14, which has a greater availability with a stable and low-cost commercial molecule, as a recognition element. This molecule, called NSC-405020, is specific of the PEX domain of MMP-14 within the binding pocket. Through the covalent grafting of the NSC-405020 molecule on carbon nanotubes (CNTs), we were able to detect and quantify MMP-14 using electrochemical impedance spectroscopy with a linear range of detection of 10 ng⋅mL-1 to 100 ng⋅mL-1, and LOD of 7.5 ng⋅mL-1. The specificity of the inhibitory small molecule was validated against the PEX domain of MMP-1. The inhibitor loaded CNTs system showed as a desirable candidate to become an alternative to the conventional recognition bioelements for the detection of MMP-14.


Assuntos
Metaloproteinase 14 da Matriz , Nanotubos de Carbono , Metaloproteinase 14 da Matriz/química , Metaloproteinase 14 da Matriz/metabolismo , Hemopexina/química , Hemopexina/metabolismo , Hemopexina/farmacologia , Metaloproteinase 1 da Matriz/metabolismo , Estrutura Terciária de Proteína
14.
Brain Res ; 1765: 147507, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-33930375

RESUMO

Heme release from hemoglobin may contribute to secondary injury after intracerebral hemorrhage (ICH). The primary endogenous defense against heme toxicity is hemopexin, a 57 kDa glycoprotein that is depleted in the CNS after hemorrhagic stroke. We hypothesized that systemic administration of exogenous hemopexin would reduce perihematomal injury and improve outcome after experimental ICH. Intraperitoneal treatment with purified human plasma hemopexin beginning 2 h after striatal ICH induction and repeated daily for the following two days reduced blood-brain barrier disruption and cell death at 3 days. However, it had no effect on neurological deficits at 4 or 7 days or striatal cell viability at 8 days. Continuous daily hemopexin administration had no effect on striatal heme content at 3 or 7 days, and did not attenuate neurological deficits, inflammatory cell infiltration, or perihematomal cell viability at 8 days. These results suggest that systemic hemopexin treatment reduces early injury after ICH, but this effect is not sustained, perhaps due to an imbalance between striatal tissue heme and hemopexin content at later time points. Future studies should investigate its effect when administered by methods that more efficiently target CNS delivery.


Assuntos
Hemorragia Cerebral/tratamento farmacológico , Hemopexina/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/metabolismo , Morte Celular/efeitos dos fármacos , Hemorragia Cerebral/metabolismo , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Feminino , Heme/metabolismo , Heme Oxigenase-1/metabolismo , Hemoglobinas/metabolismo , Hemopexina/metabolismo , Injeções Intraperitoneais , Masculino , Camundongos , Resultado do Tratamento
15.
Front Immunol ; 12: 627944, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33763072

RESUMO

Sickle cell disease (SCD) is an inherited hemolytic disorder, defined by a point mutation in the ß-globin gene. Stress conditions such as infection, inflammation, dehydration, and hypoxia trigger erythrocyte sickling. Sickled red blood cells (RBCs) hemolyze more rapidly, show impaired deformability, and increased adhesive properties to the endothelium. In a proinflammatory, pro-coagulative environment with preexisting endothelial dysfunction, sickled RBCs promote vascular occlusion. Hepatobiliary involvement related to the sickling process, such as an acute sickle hepatic crisis, is observed in about 10% of acute sickle cell crisis incidents. In mice, ligation of CD40 with an agonistic antibody leads to a macrophage activation in the liver, triggering a sequence of systemic inflammation, endothelial cell activation, thrombosis, and focal ischemia. We found that anti-CD40 antibody injection in sickle cell mice induces a systemic inflammatory and hemodynamic response with accelerated hemolysis, extensive vaso-occlusion, and large ischemic infarctions in the liver mimicking an acute hepatic crisis. Administration of the tumor necrosis factor-α (TNF-α) blocker, etanercept, and the heme scavenger protein, hemopexin attenuated end-organ damage. These data collectively suggest that anti-CD40 administration offers a novel acute liver crisis model in humanized sickle mice, allowing for evaluation of therapeutic proof-of-concept.


Assuntos
Anemia Falciforme/complicações , Anticorpos/toxicidade , Antígenos CD40/agonistas , Inflamação/etiologia , Hepatopatias/etiologia , Anemia Falciforme/sangue , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/imunologia , Animais , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Citocinas/sangue , Modelos Animais de Doenças , Etanercepte/farmacologia , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/imunologia , Hemólise , Hemopexina/farmacologia , Humanos , Inflamação/sangue , Inflamação/imunologia , Inflamação/prevenção & controle , Mediadores da Inflamação/sangue , Hepatopatias/sangue , Hepatopatias/imunologia , Hepatopatias/prevenção & controle , Camundongos Transgênicos , Inibidores do Fator de Necrose Tumoral/farmacologia , Disfunção Ventricular Direita/sangue , Disfunção Ventricular Direita/etiologia , Disfunção Ventricular Direita/imunologia
16.
Front Immunol ; 11: 1684, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849588

RESUMO

Hemopexin is the main plasmatic scavenger of cell-free heme, released in the context of intravascular hemolysis or major cell injury. Heme is indispensable for the oxygen transport by hemoglobin but when released outside of the erythrocytes it becomes a danger-associated molecular pattern, contributing to tissue injury. One of the mechanisms of pro-inflammatory action of heme is to activate the innate immune complement cascade. Therefore, we hypothesized that injection of hemopexin will prevent hemolysis-induced complement activation. Human plasma-derived hemopexin is compatible with the heme clearance machinery of the mice. 100 or 500 mg/kg of hemopexin was injected in C57Bl/6 mice before treatment with phenylhydrazine (inducer of erythrocytes lysis) or with PBS as a control. Blood was taken at different timepoints to determine the pharmacokinetic of injected hemopexin in presence and absence of hemolysis. Complement activation was determined in plasma, by the C3 cleavage (western blot) and in the kidneys (immunofluorescence). Kidney injury was evaluated by urea and creatinine in plasma and renal NGAL and HO-1 gene expression were measured. The pharmacokinetic properties of hemopexin (mass spectrometry) in the hemolytic mice were affected by the target-mediated drug disposition phenomenon due to the high affinity of binding of hemopexin to heme. Hemolysis induced complement overactivation and signs of mild renal dysfunction at 6 h, which were prevented by hemopexin, except for the NGAL upregulation. The heme-degrading capacity of the kidney, measured by the HO-1 expression, was not affected by the treatment. These results encourage further studies of hemopexin as a therapeutic agent in models of diseases with heme overload.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Hemopexina/farmacologia , Hemopexina/farmacocinética , Animais , Humanos , Rim/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
17.
Biometals ; 22(3): 421-37, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19039664

RESUMO

Hemopexin (HPX) binds heme tightly, thus protecting cells from heme toxicity during hemolysis, trauma and ischemia-reperfusion injury. Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Normal iron homeostasis requires copper-replete cells. When heme-HPX induces HMOX1, the copper-storing metallothioneins (MTs) are also induced whereas the copper-responsive copper chaperone that delivers copper to Cu, Zn superoxide dismutase, CCS1, is decreased; both are known responses when cellular copper levels rise. Endocytosis of heme-HPX is needed to regulate CCS1 since the signaling ligand cobalt-protoporphyrin (CoPP)-HPX, which does not induce HMOX1 but does co-localize with heme-HPX in endosomes, also decreased CCS1. These observations support that heme-HPX mobilizes copper in cells. The regulation of both hmox1 and mt1 is prevented by the copper-chelator, bathocuproinedisulfonate (BCDS), but not uptake of heme-AlexaFluor-labeled HPX into endosomes. Supporting a role for copper in HMOX1 regulation by heme-HPX, nutritional copper deficiency generated by tetraethylene pentamine or 232 tetraamine prevented HMOX1 induction. Using conditions that mimic maturing endosomes, we found that copper prevents rebinding of heme to apo-HPX. A model is presented in which copper endocytosis together with that of heme-HPX provides a means to facilitate heme export from HPX in the maturing endosomes: heme is needed for hmox1 transcription, while cytosolic copper and CCS1 provide a link for the known simultaneous regulation of hmox1 and mt1 by heme-HPX.


Assuntos
Cobre/fisiologia , Heme Oxigenase-1/metabolismo , Heme/farmacologia , Hemopexina/farmacologia , Proteínas de Membrana/metabolismo , Animais , Linhagem Celular Tumoral , Cobre/química , Cobre/deficiência , Endocitose/efeitos dos fármacos , Endossomos/metabolismo , Ativação Enzimática/efeitos dos fármacos , Etilenodiaminas/química , Heme/química , Hemopexina/química , Concentração de Íons de Hidrogênio , Metalotioneína/metabolismo , Camundongos , Chaperonas Moleculares/metabolismo
18.
J Am Soc Nephrol ; 19(11): 2140-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18753258

RESUMO

Hemopexin is an abundant plasma protein that effectively scavenges heme. When infused into rats, hemopexin induces reversible proteinuria, and activated hemopexin is increased in children with minimal change nephrotic syndrome. These observations suggest a role for hemopexin in glomerular disease; in this study, the effects of active hemopexin on human podocytes and glomerular endothelial cells, the two cell types that compose the glomerular filtration barrier, were investigated. Within 30 min of treatment with hemopexin, actin reorganized from stress fibers to cytoplasmic aggregates and membrane ruffles in wild-type podocytes. This did not occur in nephrin-deficient podocytes unless they were transfected with nephrin-expressing plasmids. Furthermore, hemopexin did not affect actin organization in cells that do not express nephrin, specifically human glomerular endothelial cells, fibroblasts, and HEK293 cells. The effects of hemopexin on wild-type podocytes reversed within 4 h and were inhibited by preincubation with human plasma. Treatment with hemopexin activated protein kinase B in both wild-type and nephrin-deficient podocytes but activated RhoA only in wild-type cells. In addition, hemopexin led to a selective increase in the passage of albumin across monolayers of glomerular endothelial cells and to a reduction in glycocalyx. In summary, active hemopexin causes nephrin-dependent remodeling of podocytes and affects permeability of the glomerular filtration barrier by degrading the glycocalyx.


Assuntos
Actinas/metabolismo , Hemopexina/farmacologia , Proteínas de Membrana/metabolismo , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Sequência de Bases , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Glicocálix/efeitos dos fármacos , Glicocálix/metabolismo , Proteínas de Fluorescência Verde/genética , Hemopexina/genética , Hemopexina/metabolismo , Humanos , Glomérulos Renais/citologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Podócitos/ultraestrutura , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transfecção , Proteína rhoA de Ligação ao GTP/metabolismo
19.
JCI Insight ; 3(12)2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29925688

RESUMO

In hemolytic diseases, such as sickle cell disease (SCD), intravascular hemolysis results in the release of hemoglobin, heme, and heme-loaded membrane microvesicles in the bloodstream. Intravascular hemolysis is thus associated with inflammation and organ injury. Complement system can be activated by heme in vitro. We investigated the mechanisms by which hemolysis and red blood cell (RBC) degradation products trigger complement activation in vivo. In kidney biopsies of SCD nephropathy patients and a mouse model with SCD, we detected tissue deposits of complement C3 and C5b-9. Moreover, drug-induced intravascular hemolysis or injection of heme or hemoglobin in mice triggered C3 deposition, primarily in kidneys. Renal injury markers (Kim-1, NGAL) were attenuated in C3-/- hemolytic mice. RBC degradation products, such as heme-loaded microvesicles and heme, induced alternative and terminal complement pathway activation in sera and on endothelial surfaces, in contrast to hemoglobin. Heme triggered rapid P selectin, C3aR, and C5aR expression and downregulated CD46 on endothelial cells. Importantly, complement deposition was attenuated in vivo and in vitro by heme scavenger hemopexin. In conclusion, we demonstrate that intravascular hemolysis triggers complement activation in vivo, encouraging further studies on its role in SCD nephropathy. Conversely, heme inhibition using hemopexin may provide a novel therapeutic opportunity to limit complement activation in hemolytic diseases.


Assuntos
Sistema Livre de Células , Heme/metabolismo , Hemólise/fisiologia , Injúria Renal Aguda , Anemia Falciforme , Animais , Complemento C3/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Células Endoteliais , Eritrócitos , Feminino , Hemopexina/farmacologia , Receptor Celular 1 do Vírus da Hepatite A , Rim , Camundongos , Camundongos Endogâmicos C57BL , Selectina-P , Receptor da Anafilatoxina C5a/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
20.
Cell Death Dis ; 9(2): 181, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29415995

RESUMO

Spinal cord injury (SCI) is a devastating type of central nervous system (CNS) trauma with limited therapeutic treatments. The polarization of microglia into the M1 or M2 state has been documented to play important roles in the pathogenesis of SCI, although the complete repertoire of underlying factors has not been identified. Interestingly, the time point at which hematomyelia (intramedullary spinal cord hemorrhage) is alleviated coincides with a decrease in the number of M2 microglia. Here the function of Hemopexin (Hpx), a hematogenous glycoprotein, was examined in the crush model of SCI. Hpx levels were elevated at the lesion site during hematomyelia and were synchronously correlated with the level of the M2 marker Arginase-1 (Arg-1). Ablation of Hpx in vivo affected the polarization state of lipopolysaccharide (LPS)-stimulated microglia, as mirrored by a lower percentage of M2 microglia and a higher percentage of M1 microglia in the lesion site, which delayed the recovery and exacerbated the behavioral dysfunction after SCI. However, Hpx induced a rapid switch from the M1 to M2 phenotype in LPS-stimulated primary cultured microglia in a heme scavenging-independent manner. The supernant of Hpx-treated microglia ameliorated neuronal degeneration, alleviated demyelination, and promoted oligodendrocyte precursor cell (OPC) maturation. This modulatory effect of Hpx on microglia polarization was at least partially mediated by the LRP-1 receptor. Based on these results, Hpx is considered a novel modulator of the polarization of microglia during the pathogenesis of SCI and may play a crucial role in the recovery from SCI.


Assuntos
Arginase/metabolismo , Hemopexina/metabolismo , Microglia/metabolismo , Traumatismos da Medula Espinal/sangue , Animais , Polaridade Celular/efeitos dos fármacos , Polaridade Celular/fisiologia , Células Cultivadas , Hemopexina/farmacologia , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/patologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA