Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 167(1): 90-103, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37702203

RESUMEN

During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub-lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub-lethal Hb concentrations, we find that synaptic AMPAR-driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co-application can prevent these deficits by scavenging free Hb to reduce it to sub-threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage.


Asunto(s)
Lesiones Encefálicas , Hemorragia Subaracnoidea , Humanos , Haptoglobinas/farmacología , Haptoglobinas/uso terapéutico , Hemorragia Subaracnoidea/metabolismo , Hemoglobinas/farmacología , Hemoglobinas/uso terapéutico , Neuronas/metabolismo , Lesiones Encefálicas/metabolismo
2.
Molecules ; 27(18)2022 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-36144832

RESUMEN

(1) Background: This study evaluated the effects of a plant bioactive (Phyto Ax'Cell, Phytosynthese, Mozac, France) on the inflammatory status and health of dairy cows during calving. (2) Methods: 46 Holstein crossbred cows were randomized into a control group (CON, n = 23) and the Phyto Ax'Cell group (PAC, n = 23). PAC received Phyto Ax'Cell at 25 g/cow/day, from 15 days prepartum to 7 days postpartum. Blood analyses were performed weekly from D-7 to D14 to evaluate the energy metabolism and inflammatory status; rectal temperature was measured daily within 14 days from calving day (D0). (3) Results: PAC showed lower serum haptoglobin at D7 (0.55 vs. 0.79 mg/mL; p < 0.05) and D14 (0.44 vs. 0.66 mg/mL; p < 0.05). CON had a higher number of circulating white blood cells and granulocytes on D7 (p < 0.05). Fewer cows from PAC showed hyperthermia (≥39 °C) during the first 2 weeks postpartum (−7%, p < 0.05). Energy metabolism, which was represented by the NEFA/cholesterol ratio, improved (0.21 vs. 0.36 at D0, p < 0.1; 0.19 and 0.15 vs. 0.36 and 0.32, respectively, at D+7 and D+14, p < 0.05) under the plant bioactive supplementation. (4) Conclusions: The results suggest that the anti-inflammatory plant bioactive compound with Brazilian green propolis administered during calving had a beneficial effect on the energy and inflammatory status of dairy cows.


Asunto(s)
Leche , Própolis , Animales , Bovinos , Dieta/veterinaria , Suplementos Dietéticos , Ácidos Grasos no Esterificados/metabolismo , Femenino , Haptoglobinas/metabolismo , Haptoglobinas/farmacología , Lactancia , Leche/metabolismo , Fitoquímicos/farmacología , Própolis/farmacología
3.
Folia Med Cracov ; 62(3): 5-18, 2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-36309828

RESUMEN

Platelet aggregation contributes to the pathogenesis of cardiovascular diseases. After activation it leads to dense granule secretion and 5-HT release. The question arises; how platelet aggregation is endogenously controlled during blood circulation. In preliminary studies, we observed that human platelets aggregate more rapidly when suspended in buffer as compared to those suspended in plasma (PRP). These observations point to the presence of an endogenous substance that may inhibit arachidonic acid- induced platelet aggregation. An analysis of plasma Cohn fractions demonstrated that most of the plasma inhibitory activity was associated with albumin-rich and α-globulin rich protein fractions. The identity of plasma endogenous inhibitors of platelet aggregation (EIPA) was established by affinity chromatography on Cibacron Blue F3G-A for specific removal of albumin. The association of α-globulins to EIPA activity was recognized as due to haptoglobin by affinity chromatography on a column of hemoglobin-sepharose. In addition, we also found that the distribution of EIPA activity varies according to sex and physiological state. These findings reveal that EIPA may act by modulation of arachidonic acid metabolism or sequestering the fatty acid substrate.


Asunto(s)
Agregación Plaquetaria , Serotonina , Humanos , Serotonina/metabolismo , Serotonina/farmacología , Haptoglobinas/metabolismo , Haptoglobinas/farmacología , Albúmina Sérica , Ácido Araquidónico/farmacología , Ácido Araquidónico/metabolismo , Albúmina Sérica Humana/metabolismo , Albúmina Sérica Humana/farmacología , Plaquetas/metabolismo
4.
Wound Repair Regen ; 28(4): 493-505, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32428978

RESUMEN

Nonhealing wounds possess elevated numbers of pro-inflammatory M1 macrophages, which fail to transition to anti-inflammatory M2 phenotypes that promote healing. Hemoglobin (Hb) and haptoglobin (Hp) proteins, when complexed (Hb-Hp), can elicit M2-like macrophages through the heme oxygenase-1 (HO-1) pathway. Despite the fact that nonhealing wounds are chronically inflamed, previous studies have focused on non-inflammatory systems, and do not thoroughly compare the effects of complexed vs individual proteins. We aimed to investigate the effect of Hb/Hp treatments on macrophage phenotype in an inflammatory, lipopolysaccharide (LPS)-stimulated environment, similar to chronic wounds. Human M1 macrophages were cultured in vitro and stimulated with LPS. Concurrently, Hp, Hb, or Hb-Hp complexes were delivered. The next day, 27 proteins related to inflammation were measured in the supernatants. Hp treatment decreased a majority of inflammatory factors, Hb increased many, and Hb-Hp had intermediate trends, indicating that Hp attenuated overall inflammation to the greatest extent. From this data, Ingenuity Pathway Analysis software identified high motility group box 1 (HMGB1) as a key canonical pathway-strongly down-regulated from Hp, strongly up-regulated from Hb, and slightly activated from Hb-Hp. HMGB1 measurements in macrophage supernatants confirmed this trend. In vivo results in diabetic mice with biopsy punch wounds demonstrated accelerated wound closure with Hp treatment, and delayed wound closure with Hb treatment. This work specifically studied Hb/Hp effects on macrophages in a highly inflammatory environment relevant to chronic wound healing. Results show that Hp-and not Hb-Hp, which is known to be superior in noninflammatory conditions-reduces inflammation in LPS-stimulated macrophages, and HMGB1 signaling is also implicated. Overall, Hp treatment on M1 macrophages in vitro reduced the inflammatory secretion profile, and also exhibited benefits in in silico and in vivo wound-healing models.


Asunto(s)
Proteína HMGB1/efectos de los fármacos , Haptoglobinas/farmacología , Hemoglobinas/farmacología , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Diabetes Mellitus , Proteína HMGB1/metabolismo , Hemo-Oxigenasa 1 , Humanos , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Ratones Obesos , Receptores de Superficie Celular/metabolismo , Transducción de Señal
5.
BMC Biotechnol ; 18(1): 15, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29544494

RESUMEN

BACKGROUND: Preclinical studies have evaluated haptoglobin (Hp) polymers from pooled human plasma as a therapeutic protein to attenuate toxic effects of cell-free hemoglobin (Hb). Proof of concept studies have demonstrated efficacy of Hp in hemolysis associated with transfusion and sickle cell anemia. However, phenotype-specific Hp products might be desirable to exploit phenotype specific activities of Hp 1-1 versus Hp 2-2, offering opportunities for recombinant therapeutics. Prohaptoglobin (proHp) is the primary translation product of the Hp mRNA. ProHp is proteolytically cleaved by complement C1r subcomponent-like protein (C1r-LP) in the endoplasmic reticulum. Two main allelic Hp variants, HP1 and HP2 exist. The larger HP2 is considered to be the ancestor variant of all human Hp alleles and is characterized by an α2-chain, which contains an extra cysteine residue that pairs with additional α-chains generating multimers with molecular weights of 200-900 kDa. The two human HP1 alleles (HP1F and HP1S) differ by a two-amino-acid substitution polymorphism within the α-chain and are derived from HP2 by recurring exon deletions. RESULTS: In the present study, we describe a process for the production of recombinant phenotype specific Hp polymers in mammalian FS293F cells. This approach demonstrates that efficient expression of mature and fully functional protein products requires co-expression of active C1r-LP. The functional characterization of our proteins, which included monomer/polymer distribution, binding affinities as well as NO-sparing and antioxidant functions, demonstrated that C1r-LP-processed recombinant Hp demonstrates equal protective functions as plasma derived Hp in vitro as well as in animal studies. CONCLUSIONS: We present a recombinant production process for fully functional phenotype-specific Hp therapeutics. The proposed process could accelerate the development of Hb scavengers to treat patients with cell-free Hb associated disease states, such as sickle cell disease and other hemolytic conditions.


Asunto(s)
Haptoglobinas/genética , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Ingeniería de Proteínas/métodos , Serina Endopeptidasas/genética , Animales , Vasos Coronarios/efectos de los fármacos , Cobayas , Haptoglobinas/farmacología , Hemo/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Óxido Nítrico/metabolismo , Fenotipo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Serina Endopeptidasas/metabolismo , Porcinos
6.
Circulation ; 134(13): 945-60, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27515135

RESUMEN

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.


Asunto(s)
Eritrocitos/efectos de los fármacos , Haptoglobinas/farmacología , Hemopexina/farmacología , Animales , Proteínas Sanguíneas/farmacología , Eritrocitos/metabolismo , Haptoglobinas/administración & dosificación , Hemopexina/administración & dosificación , Humanos , Inflamación/tratamiento farmacológico , Ratones , Resucitación/métodos , Choque Hemorrágico/metabolismo , Reacción a la Transfusión
7.
Am J Physiol Heart Circ Physiol ; 312(6): H1120-H1127, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28314763

RESUMEN

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.


Asunto(s)
Antihipertensivos/farmacología , Endotelio Vascular/efectos de los fármacos , Haptoglobinas/farmacología , Hemoglobinas , Hemopexina/farmacología , Hipertensión/prevención & control , Vasoconstricción/efectos de los fármacos , Animales , Antihipertensivos/administración & dosificación , Diabetes Mellitus/fisiopatología , Dieta Alta en Grasa , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Haptoglobinas/administración & dosificación , Hemopexina/administración & dosificación , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Hipertensión/fisiopatología , Infusiones Intravenosas , Riñón/metabolismo , Riñón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Factores de Tiempo
8.
Crit Care ; 21(1): 245, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-28946897

RESUMEN

BACKGROUND: Intravascular haemolysis has been associated with acute kidney injury (AKI) in different clinical settings (cardiac surgery, sickle cell disease). Haemolysis occurs frequently in critically ill burn patients. The aim of this study was to assess the predictive value of haptoglobin at admission to predict major adverse kidney events (MAKE) and AKI in critically ill burn patients. METHODS: We conducted a retrospective, single-centre cohort study in a burn critical care unit in a tertiary centre, including all consecutive severely burned patients (total burned body surface > 20% and/or shock and/or mechanical ventilation at admission) from January 2012 to April 2017 with a plasmatic haptoglobin dosage at admission. RESULTS: A total of 130 patients were included in the analysis. Their mean age was 49 (34-62) years, their median total body surface area burned was 29% (15-51%) and the intensive care unit (ICU) mortality was 25%. Early haemolysis was defined as an undetectable plasmatic haptoglobin at admission. We used logistic regression to identify MAKE and AKI risk factors. In multivariate analysis, undetectable haptoglobin was associated with MAKE and AKI (respectively, OR 6.33, 95% CI 2.34-16.45, p < 0.001; OR 8.32, 95% CI 2.86-26.40, p < 0.001). CONCLUSIONS: Undetectable plasmatic haptoglobin at ICU admission is an independent risk factor for MAKE and AKI in critically ill burn patients. This study provides a rationale for biomarker-guided therapy using haptoglobin in critically ill burn patients.


Asunto(s)
Lesión Renal Aguda/etiología , Quemaduras/complicaciones , Haptoglobinas/análisis , Haptoglobinas/farmacología , Lesión Renal Aguda/mortalidad , Adulto , Quemaduras/metabolismo , Quemaduras/mortalidad , Estudios de Cohortes , Creatinina/análisis , Creatinina/sangre , Femenino , Haptoglobinas/uso terapéutico , Humanos , Unidades de Cuidados Intensivos/organización & administración , Masculino , Persona de Mediana Edad , Paris , Respiración Artificial/efectos adversos , Estudios Retrospectivos , Factores de Riesgo , Estadísticas no Paramétricas
9.
Am J Respir Crit Care Med ; 193(10): 1111-22, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26694989

RESUMEN

RATIONALE: Hemolysis occurs not only in conditions such as sickle cell disease and malaria but also during transfusion of stored blood, extracorporeal circulation, and sepsis. Cell-free Hb depletes nitric oxide (NO) in the vasculature, causing vasoconstriction and eventually cardiovascular complications. We hypothesize that Hb-binding proteins may preserve vascular NO signaling during hemolysis. OBJECTIVES: Characterization of an archetypical function by which Hb scavenger proteins could preserve NO signaling during hemolysis. METHODS: We investigated NO reaction kinetics, effects on arterial NO signaling, and tissue distribution of cell-free Hb and its scavenger protein complexes. MEASUREMENTS AND MAIN RESULTS: Extravascular translocation of cell-free Hb into interstitial spaces, including the vascular smooth muscle cell layer of rat and pig coronary arteries, promotes vascular NO resistance. This critical disease process is blocked by haptoglobin. Haptoglobin does not change NO dioxygenation rates of Hb; rather, the large size of the Hb:haptoglobin complex prevents Hb extravasation, which uncouples NO/Hb interaction and vasoconstriction. Size-selective compartmentalization of Hb functions as a substitute for red blood cells after hemolysis and preserves NO signaling in the vasculature. We found that evolutionarily and structurally unrelated Hb-binding proteins, such as PIT54 found in avian species, functionally converged with haptoglobin to protect NO signaling by sequestering cell-free Hb in large protein complexes. CONCLUSIONS: Sequential compartmentalization of Hb by erythrocytes and scavenger protein complexes is an archetypical mechanism, which may have supported coevolution of hemolysis and normal vascular function. Therapeutic supplementation of Hb scavengers may restore vascular NO signaling and attenuate disease complications in patients with hemolysis.


Asunto(s)
Haptoglobinas/farmacología , Hemólisis/efectos de los fármacos , Óxido Nítrico/metabolismo , Resistencia Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Animales , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiología , Modelos Animales de Enfermedad , Humanos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Ratas , Porcinos , Resistencia Vascular/fisiología
10.
J Neurochem ; 139(4): 586-595, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27364920

RESUMEN

Haptoglobin (Hp) binds hemoglobin (Hb) with high affinity and provides the primary defense against its toxicity after intravascular hemolysis. Neurons are exposed to extracellular Hb after CNS hemorrhage, and a therapeutic effect of Hp via Hb sequestration has been hypothesized. In this study, we tested the hypothesis that Hp protects neurons from Hb in primary mixed cortical cell cultures. Treatment with low micromolar concentrations of human Hb for 24 h resulted in loss of 10-20% of neurons without injuring glia. Concomitant treatment with Hp surprisingly increased neuronal loss five-sevenfold, with similar results produced by Hp 1-1 and 2-2 phenotypes. Consistent with a recent in vivo observation, neurons expressed the CD163 receptor for Hb and the Hb-Hp complex in these cultures. Hp reduced overall Hb uptake, directed it away from the astrocyte-rich CD163-negative glial monolayer, and decreased induction of the iron-binding protein ferritin. Hb-Hp complex neuronal toxicity, like that of Hb per se, was iron-dependent and reduced by deferoxamine and 2,2' bipyridyl. These results suggest that Hp increases the vulnerability of CD163+ neurons to Hb by permitting Hb uptake while attenuating the protective response of ferritin induction by glial cells. Cover Image for this issue: doi: 10.1111/jnc.13342.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Haptoglobinas/farmacología , Hemoglobinas/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de Superficie Celular/biosíntesis , Animales , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Expresión Génica , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Receptores de Superficie Celular/genética
11.
Br J Haematol ; 175(4): 714-723, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27507623

RESUMEN

There is growing evidence that extracellular haemoglobin and haem mediate inflammatory and oxidative damage in sickle cell disease. Haptoglobin (Hp), the scavenger for free haemoglobin, is depleted in most patients with sickle cell disease due to chronic haemolysis. Although single infusions of Hp can ameliorate vaso-occlusion in mouse models of sickle cell disease, prior studies have not examined the therapeutic benefits of more chronic Hp dosing on sickle cell disease manifestations. In the present study, we explored the effect of Hp treatment over a 3-month period in sickle mice at two dosing regimens: the first at a moderate dose of 200 mg/kg thrice weekly and the second at a higher dose of 400 mg/kg thrice weekly. We found that only the higher dosing regimen resulted in increased haem-oxygenase-1 and heavy chain ferritin (H-ferritin) expression and decreased iron deposition in the kidney. Despite the decreased kidney iron deposition following Hp treatment, there was no significant improvement in kidney function. However, there was a nearly significant trend towards decreased liver infarction.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Apoferritinas/metabolismo , Haptoglobinas/farmacología , Hierro/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Animales , Apoferritinas/genética , Recuento de Células Sanguíneas , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Haptoglobinas/administración & dosificación , Haptoglobinas/efectos adversos , Haptoglobinas/farmacocinética , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Enfermedades Renales/etiología , Enfermedades Renales/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Resultado del Tratamiento
12.
Biol Chem ; 392(4): 371-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21294680

RESUMEN

Haptoglobin (Hpt) is known to capture circulating free hemoglobin (Hb) and bind apolipoprotein (Apo) A-I or E. Here, we report that Hb can be tightly bound by most of Hpt molecules (TB-Hpt, 80%), whereas loosely bound by a minor part of them (LB-Hpt, 20%). LB-Hpt amount was significantly increased (over 60%) in patients with acute coronary syndrome. LB-Hpt bound ApoA-I and ApoE less efficiently than TB-Hpt (8- and 4-fold less, respectively) and did not affect their activity of stimulating the enzyme lecithin-cholesterol acyltransferase. LB-Hpt and TB-Hpt displayed comparable levels of nitrotyrosine residues, but differences in glycan chains. Changes in LB-Hpt level might be associated with changes in Hpt functions.


Asunto(s)
Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/enzimología , Apolipoproteína A-I/metabolismo , Apolipoproteínas E/metabolismo , Estudios de Casos y Controles , Haptoglobinas/farmacología , Humanos , Lectinas/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Unión Proteica
13.
Blood ; 113(11): 2578-86, 2009 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-19131549

RESUMEN

Detoxification and clearance of extracellular hemoglobin (Hb) have been attributed to its removal by the CD163 scavenger receptor pathway. However, even low-level hydrogen peroxide (H(2)O(2)) exposure irreversibly modifies Hb and severely impairs Hb endocytosis by CD163. We show here that when Hb is bound to the high-affinity Hb scavenger protein haptoglobin (Hp), the complex protects Hb from structural modification by preventing alpha-globin cross-links and oxidations of amino acids in critical regions of the beta-globin chain (eg, Trp15, Cys93, and Cys112). As a result of this structural stabilization, H(2)O(2)-exposed Hb-Hp binds to CD163 with the same affinity as nonoxidized complex. Endocytosis and lysosomal translocation of oxidized Hb-Hp by CD163-expressing cells were found to be as efficient as with nonoxidized complex. Hp complex formation did not alter Hb's ability to consume added H(2)O(2) by redox cycling, suggesting that within the complex the oxidative radical burden is shifted to Hp. We provide structural and functional evidence that Hp protects Hb when oxidatively challenged with H(2)O(2) preserving CD163-mediated Hb clearance under oxidative stress conditions. In addition, our data provide in vivo evidence that unbound Hb is oxidatively modified within extravascular compartments consistent with our in vitro findings.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Haptoglobinas/fisiología , Hemoglobinas/efectos de los fármacos , Hemoglobinas/metabolismo , Peróxido de Hidrógeno/farmacología , Receptores de Superficie Celular/metabolismo , Aminoácidos/metabolismo , Animales , Células Cultivadas , Perros , Haptoglobinas/química , Haptoglobinas/metabolismo , Haptoglobinas/farmacología , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemoglobinas/química , Humanos , Oxidación-Reducción , Unión Proteica/fisiología , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Receptores Depuradores/fisiología , Transducción de Señal/fisiología
14.
J Exp Med ; 183(3): 1023-9, 1996 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-8642243

RESUMEN

Natural immunity of humans to the cattle pathogen Trypanosoma brucei brucei has been attributed to the presence in normal human serum (NHS) of lytic factors for the parasites. We and others have shown that NHS contains two trypanolytic factors (herein termed TLF1 and TLF2) that can be separated by gel filtration. TLF1 copurifies with a subclass of high density lipoprotein (HDL), whereas TLF2 has a much higher molecular weight and does not appear to be a lipoprotein. We find that the trypanolytic activity of purified TLF1 is totally inhibited by exogenous haptoglobin (Hp) at concentrations (0.1 mg/ml) lower than those present in NHS (0.2-2 mg/ml). In contrast, exogenous Hp (up to 2.5 mg/ml) has no effect on the lytic activity of either NHS or isolated TLF2. Hp-depleted sera from patients with intravascular hemolysis is severalfold more trypanolytic than NHS. These sera contain only TLF1, and their lytic activity is totally abolished upon the addition of Hp (0.1 mg/ml). When NHS containing different Hp allotypes is fractionated by gel filtration, TLF1 activity is either revealed or remains masked, depending on whether it coelutes with Hp. Masked TLF1 activity in the column fractions is revealed if Hp is removed by density gradient ultracentrifugation. We conclude that endogenous Hp inhibits TLF1 activity, and that TLF2 is the main trypanolytic factor in NHS.


Asunto(s)
Factores Biológicos/sangre , Lipoproteínas/sangre , Trypanosoma brucei brucei/inmunología , Tripanosomiasis Africana/inmunología , Cloruro de Amonio/farmacología , Animales , Factores Biológicos/aislamiento & purificación , Bovinos , Cromatografía en Gel , Ensayo de Inmunoadsorción Enzimática , Haptoglobinas/farmacología , Haptoglobinas/fisiología , Humanos , Inmunidad Innata , Lipoproteínas/aislamiento & purificación , Lipoproteínas HDL/sangre , Tripanosomiasis Africana/sangre
15.
Trends Mol Med ; 26(7): 683-697, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32589936

RESUMEN

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


Asunto(s)
Haptoglobinas/farmacología , Haptoglobinas/uso terapéutico , Hemoglobinas/toxicidad , Anemia de Células Falciformes/tratamiento farmacológico , Animales , Eritrocitos/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Sepsis/tratamiento farmacológico , Reacción a la Transfusión/tratamiento farmacológico
16.
Science ; 215(4533): 691-3, 1982 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-7036344

RESUMEN

The combination of bacteria and blood in a wound can have lethal consequences, probably because hemoglobin iron supports prolific bacterial growth. Rats inoculated intraperitoneally with pathogenic Escherichia coli and small amounts of hemoglobin die. Simultaneous administration of haptoglobin, a naturally occurring hemoglobin-binding protein, fully protects against lethality. Therefore, haptoglobin may not only accelerate the clearance of free hemoglobin, but also limit its utilization by adventitious bacteria. Haptoglobin may have therapeutic potential in the treatment of life-threatening, hemoglobin-driven bacterial infections.


Asunto(s)
Escherichia coli/crecimiento & desarrollo , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Haptoglobinas/farmacología , Hierro/metabolismo
17.
J Clin Invest ; 129(12): 5219-5235, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31454333

RESUMEN

Delayed ischemic neurological deficit (DIND) is a major driver of adverse outcomes in patients with aneurysmal subarachnoid hemorrhage (aSAH), defining an unmet need for therapeutic development. Cell-free hemoglobin that is released from erythrocytes into the cerebrospinal fluid (CSF) is suggested to cause vasoconstriction and neuronal toxicity, and correlates with the occurrence of DIND. Cell-free hemoglobin in the CSF of patients with aSAH disrupted dilatory NO signaling ex vivo in cerebral arteries, which shifted vascular tone balance from dilation to constriction. We found that selective removal of hemoglobin from patient CSF with a haptoglobin-affinity column or its sequestration in a soluble hemoglobin-haptoglobin complex was sufficient to restore physiological vascular responses. In a sheep model, administration of haptoglobin into the CSF inhibited hemoglobin-induced cerebral vasospasm and preserved vascular NO signaling. We identified 2 pathways of hemoglobin delocalization from CSF into the brain parenchyma and into the NO-sensitive compartment of small cerebral arteries. Both pathways were critical for hemoglobin toxicity and were interrupted by the large hemoglobin-haptoglobin complex that inhibited spatial requirements for hemoglobin reactions with NO in tissues. Collectively, our data show that compartmentalization of hemoglobin by haptoglobin provides a novel framework for innovation aimed at reducing hemoglobin-driven neurological damage after subarachnoid bleeding.


Asunto(s)
Haptoglobinas/administración & dosificación , Hemoglobinas/administración & dosificación , Hemorragia Subaracnoidea/metabolismo , Espacio Subaracnoideo/metabolismo , Vasoespasmo Intracraneal/metabolismo , Animales , Arteria Basilar/metabolismo , Encéfalo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Modelos Animales de Enfermedad , Femenino , Haptoglobinas/química , Haptoglobinas/farmacología , Hemoglobinas/química , Hemoglobinas/farmacología , Humanos , Aneurisma Intracraneal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteómica , Ovinos , Transducción de Señal , Porcinos
18.
JCI Insight ; 3(18)2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30232287

RESUMEN

During the last half-century, numerous antiinflammatory agents were tested in dozens of clinical trials and have proven ineffective for treating septic shock. The observation in multiple studies that cell-free hemoglobin (CFH) levels are elevated during clinical sepsis and that the degree of increase correlates with higher mortality suggests an alternative approach. Human haptoglobin binds CFH with high affinity and, therefore, can potentially reduce iron availability and oxidative activity. CFH levels are elevated over approximately 24-48 hours in our antibiotic-treated canine model of S. aureus pneumonia that simulates the cardiovascular abnormalities of human septic shock. In this 96-hour model, resuscitative treatments, mechanical ventilation, sedation, and continuous care are translatable to management in human intensive care units. We found, in this S. aureus pneumonia model inducing septic shock, that commercial human haptoglobin concentrate infusions over 48-hours bind canine CFH, increase CFH clearance, and lower circulating iron. Over the 96-hour study, this treatment was associated with an improved metabolic profile (pH, lactate), less lung injury, reversal of shock, and increased survival. Haptoglobin binding compartmentalized CFH to the intravascular space. This observation, in combination with increasing CFHs clearance, reduced available iron as a potential source of bacterial nutrition while decreasing the ability for CFH and iron to cause extravascular oxidative tissue injury. In contrast, haptoglobin therapy had no measurable antiinflammatory effect on elevations in proinflammatory C-reactive protein and cytokine levels. Haptoglobin therapy enhances normal host defense mechanisms in contrast to previously studied antiinflammatory sepsis therapies, making it a biologically plausible novel approach to treat septic shock.


Asunto(s)
Haptoglobinas/farmacología , Lesión Pulmonar/tratamiento farmacológico , Neumonía/tratamiento farmacológico , Choque Séptico/tratamiento farmacológico , Animales , Antibacterianos , Antiinflamatorios/farmacología , Análisis de los Gases de la Sangre , Anomalías Cardiovasculares , Citocinas , Modelos Animales de Enfermedad , Perros , Haptoglobinas/uso terapéutico , Hematócrito , Humanos , Inmunidad Innata , Hierro , Estimación de Kaplan-Meier , Neumonía/microbiología , Neumonía/mortalidad , Arteria Pulmonar , Staphylococcus aureus
19.
PLoS One ; 13(4): e0196455, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29694434

RESUMEN

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.


Asunto(s)
Anemia de Células Falciformes/prevención & control , Haptoglobinas/uso terapéutico , Hemo-Oxigenasa 1/metabolismo , Hemopexina/uso terapéutico , Inflamación/prevención & control , Aldehídos/análisis , Anemia de Células Falciformes/patología , Animales , Monóxido de Carbono/farmacología , Citocinas/análisis , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Haptoglobinas/farmacología , Hemopexina/farmacología , Molécula 1 de Adhesión Intercelular , Masculino , Metaloporfirinas/farmacología , Ratones , Microsomas Hepáticos/metabolismo , Protoporfirinas/farmacología , Piel/metabolismo , Piel/patología , Factor de Transcripción ReIA/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
20.
J Clin Invest ; 91(3): 977-85, 1993 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7680672

RESUMEN

Angiogenesis is an important process in chronic inflammatory diseases. We observed that sera from patients with systemic vasculitis stimulated angiogenesis in an in vitro model using human umbilical vein endothelial cells cultured on a basement membrane (Matrigel) substrate. After 40% ammonium sulfate precipitation, angiogenic activity remained in the low molecular weight fraction and could be inactivated by heat. SDS-page of serum FPLC fractions exhibiting maximal angiogenic activity demonstrated two prominent species of 45 and 16-20 kD in patients' sera. These bands were much less apparent in sera obtained from control subjects. Amino-terminal sequencing of the 45-kD protein demonstrated that it was haptoglobin. Purified haptoglobin stimulated angiogenesis in a dose-dependent manner. The angiogenic activity of vasculitis patients' sera was partially inhibited by an antihaptoglobin antibody. Furthermore, serum haptoglobin levels in vasculitis patients correlated both with disease and angiogenic activity. Haptoglobin angiogenic activity was confirmed in two in vivo models using an implanted disc and a subcutaneous injection of basement membrane. Stimulation of angiogenesis is a newly recognized biological function of haptoglobin. The increased levels of haptoglobin found in chronic inflammatory conditions may play an important role in tissue repair. In systemic vasculitis, haptoglobin might also compensate for ischemia by promoting development of collateral vessels.


Asunto(s)
Inductores de la Angiogénesis/sangre , Haptoglobinas/análisis , Neovascularización Patológica/patología , Vasculitis/sangre , Secuencia de Aminoácidos , Inductores de la Angiogénesis/análisis , Inductores de la Angiogénesis/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Haptoglobinas/farmacología , Humanos , Ratones , Ratones Desnudos , Valores de Referencia , Venas Umbilicales , Vasculitis/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA