Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 318(5): H1296-H1307, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32302494

RESUMEN

Haptoglobin (Hp) is the plasma protein that binds and clears cell-free hemoglobin (Hb), whereas apohemoglobin (apoHb, i.e., Hb devoid of heme) can bind heme. Therefore, the apoHb-Hp protein complex should facilitate holoHb-apoHb αß-dimer exchange and apoHb-heme intercalation. Thus, we hypothesized that apoHb-Hp could facilitate both Hb and heme clearance, which, if not alleviated, could have severe microcirculatory consequences. In this study, we characterized apoHb-Hp and Hb/heme ligand interactions and assessed their in vivo consequences. Hb exchange and heme binding with the apoHb-Hp complex was studied with transfer assays using size-exclusion high-performance liquid chromatography coupled with UV-visible spectrophotometry. Exchange/transfer experiments were conducted in guinea pigs dosed with Hb or heme-albumin followed by a challenge with equimolar amounts of apoHb-Hp. Finally, systemic and microcirculatory parameters were studied in hamsters instrumented with a dorsal window chamber via intravital microscopy. In vitro and in vivo Hb exchange and heme transfer experiments demonstrated proof-of-concept Hb/heme ligand transfer to apoHb-Hp. Dosing with the apoHb-Hp complex reversed Hb- and heme-induced systemic hypertension and microvascular vasoconstriction, reduced microvascular blood flow, and diminished functional capillary density. Therefore, this study highlights the apoHb-Hp complex as a novel therapeutic strategy to attenuate the adverse systemic and microvascular responses to intravascular Hb and heme exposure.NEW & NOTEWORTHY This study highlights the apoHb-Hp complex as a novel therapeutic strategy to attenuate the adverse systemic and microvascular responses to intravascular Hb and heme exposure. In vitro and in vivo Hb exchange and heme transfer experiments demonstrated proof-of-concept Hb/heme ligand transfer to apoHb-Hp. The apoHb-Hp complex reverses Hb- and heme-induced systemic hypertension and microvascular vasoconstriction, preserves microvascular blood flow, and functional capillary density. In summary, the unique properties of the apoHb-Hp complex prevent adverse systemic and microvascular responses to Hb and heme-albumin exposure and introduce a novel therapeutic approach to facilitate simultaneous removal of extracellular Hb and heme.


Asunto(s)
Apoproteínas/metabolismo , Haptoglobinas/metabolismo , Hemo/metabolismo , Hemoglobinas/metabolismo , Hipertensión/sangre , Animales , Apoproteínas/sangre , Transfusión Sanguínea/métodos , Cricetinae , Cobayas , Humanos , Hipertensión/fisiopatología , Hipertensión/terapia , Masculino , Mesocricetus , Microcirculación , Unión Proteica , Vasoconstricción
2.
Haematologica ; 105(8): 2174-2186, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31699790

RESUMEN

Macaques are emerging as a critical animal model in transfusion medicine, because of their evolutionary similarity to humans and perceived utility in discovery and translational science. However, little is known about the metabolism of Rhesus macaque red blood cells (RBC) and how this compares to human RBC metabolism under standard blood banking conditions. Metabolomic and lipidomic analyses, and tracing experiments with [1,2,3-13C3]glucose, were performed using fresh and stored RBC (sampled weekly until storage day 42) obtained from Rhesus macaques (n=20) and healthy human volunteers (n=21). These results were further validated with targeted quantification against stable isotope-labeled internal standards. Metabolomic analyses demonstrated inter-species differences in RBC metabolism independent of refrigerated storage. Although similar trends were observed throughout storage for several metabolic pathways, species- and sex-specific differences were also observed. The most notable differences were in glutathione and sulfur metabolites, purine and lipid oxidation metabolites, acylcarnitines, fatty acyl composition of several classes of lipids (including phosphatidylserines), glyoxylate pathway intermediates, and arginine and carboxylic acid metabolites. Species-specific dietary and environmental compounds were also detected. Overall, the results suggest an increased basal and refrigerator-storage-induced propensity for oxidant stress and lipid remodeling in Rhesus macaque RBC cells, as compared to human red cells. The overlap between Rhesus macaque and human RBC metabolic phenotypes suggests the potential utility of a translational model for simple RBC transfusions, although inter-species storage-dependent differences need to be considered when modeling complex disease states, such as transfusion in trauma/hemorrhagic shock models.


Asunto(s)
Conservación de la Sangre , Eritrocitos , Animales , Bancos de Sangre , Transfusión de Eritrocitos , Femenino , Humanos , Macaca mulatta , Masculino
3.
Transfusion ; 60(3): 513-523, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32064619

RESUMEN

BACKGROUND: Red blood cell (RBC) transfusions result in the sequestration and metabolism of storage-damaged RBCs within the spleen and liver. These events are followed by increased plasma iron concentrations that can contribute to oxidant stress and cellular injury. We hypothesized that administration of a ferroportin inhibitor (FPN-INH) immediately after acute RBC exchange transfusion could attenuate posttransfusion circulatory compartment iron exposure, by retaining iron in spleen and hepatic macrophages. STUDY DESIGN AND METHODS: Donor guinea pig blood was leukoreduced, and RBCs were preserved at 4°C. Recipient guinea pigs (n = 5/group) were exchange transfused with donor RBCs after refrigerator preservation and dosed intravenously with a small-molecule FPN-INH. Groups included transfusion with vehicle (saline), 5 mg/kg or 25 mg/kg FPN-INH. A time course of RBC morphology, plasma non-transferrin-bound iron (NTBI) and plasma hemoglobin (Hb) were evaluated. End-study spleen, liver, and kidney organ iron levels, as well as renal tissue oxidation and injury, were measured acutely (24-hr after transfusion). RESULTS: RBC transfusion increased plasma NTBI, with maximal concentrations occurring 8 hours after transfusion. Posttransfusion iron accumulation resulted in tubule oxidation and acute kidney injury. FPN inhibition increased spleen and liver parenchymal/macrophage iron accumulation, but attenuated plasma NTBI, and subsequent renal tissue oxidation/injury. CONCLUSION: In situations of acute RBC transfusion, minimizing circulatory NTBI exposure by FPN inhibition may attenuate organ-specific adverse consequences of iron exposure.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hierro/sangre , Animales , Conservación de la Sangre , Proteínas de Transporte de Catión/antagonistas & inhibidores , Transfusión de Eritrocitos/métodos , Cobayas , Humanos , Masculino , Estrés Oxidativo/fisiología
4.
Biotechnol Bioeng ; 117(1): 125-145, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31612988

RESUMEN

Apohemoglobin (apoHb) is a dimeric globular protein with two vacant heme-binding pockets that can bind heme or other hydrophobic ligands. Purification of apoHb is based on partial hemoglobin (Hb) unfolding to facilitate heme extraction into an organic solvent. However, current production methods are time consuming, difficult to scale up, and use highly flammable and toxic solvents. In this study, a novel and scalable apoHb production method was developed using an acidified ethanol solution to extract the hydrophobic heme ligand into solution and tangential flow filtration to separate heme from the resultant apoprotein. Total protein and active protein yields were >95% and ~75%, respectively, with <1% residual heme in apoHb preparations and >99% purity from sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis. Virtually no loss of apoHb activity was detected at 4°C, -80°C, and in lyophilized form during long term storage. Structurally, size exclusion chromatography (SEC) and circular dichroism indicated that apoHb was dimeric with a ~25% reduction of helical content compared to Hb. Furthermore, mass spectroscopy and reverse-phase chromatography indicated that the mass of the α and ß subunits were virtually identical to the theoretical mass of these subunits in Hb and had no detectable oxidative modifications upon heme removal from Hb. SEC confirmed that apoHb bound to haptoglobin at a similar ratio to that of native Hb. Finally, reconstituted Hb (rHb) was processed via a hemichrome removal method to isolate functional rHb for biophysical characterization in which the O2 equilibrium curve, O2 dissociation, and CO association kinetics of rHb were virtually identical to native Hb. Overall, this study describes a novel and improved method to produce apoHb, as well as presents a comprehensive biochemical analysis of apoHb and rHb.


Asunto(s)
Apoproteínas , Biotecnología/métodos , Hemoglobinas , Desplegamiento Proteico , Apoproteínas/química , Apoproteínas/aislamiento & purificación , Apoproteínas/metabolismo , Cromatografía Liquida/instrumentación , Cromatografía Liquida/métodos , Eritrocitos/química , Hemo/química , Hemo/aislamiento & purificación , Hemo/metabolismo , Hemoglobinas/química , Hemoglobinas/aislamiento & purificación , Hemoglobinas/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Oxidación-Reducción
5.
Curr Opin Hematol ; 26(6): 453-460, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31483333

RESUMEN

PURPOSE OF REVIEW: The clinical indication for transfusing red blood cells (RBCs) is to restore or maintain adequate oxygenation of respiring tissue. Oxygen (O2) transport, delivery, and utilization following transfusion are impacted by perfusion, hemoglobin (Hb) allosteric saturation/desaturation, and the concentration of tissue O2. Bioavailable O2 maintains tissue utilization and homeostasis; therefore, measuring imbalances in supply and demand could be valuable to assessing blood quality and transfusion effectiveness. O2 homeostasis is critically intertwined with erythropoietic response in blood loss and anemia and the hormones that modulate iron mobilization and RBC production (e.g., erythropoietin, erythroferrone, and hepcidin) are intriguing markers for the monitoring of transfusion effectiveness in acute and chronic settings. The evaluation of RBC donor unit quality and the determination of RBC transfusion needs are emerging areas for biomarker development and minimally invasive O2 measurements. RECENT FINDINGS: Novel methods for assessing circulatory and tissue compartment biomarkers of transfusion effectiveness are suggested. In addition, monitoring of tissue oxygenation by indirect and direct measurements of O2 is available and applied in experimental settings. SUMMARY: Herein, we discuss tissue O2 homeostasis, related aspects of erythropoiesis, molecular markers and measurements of tissue oxygenation, all aimed at optimizing transfusion and assessing blood quality.


Asunto(s)
Biomarcadores , Transfusión de Eritrocitos , Homeostasis , Oxígeno/metabolismo , Anemia/diagnóstico , Anemia/etiología , Anemia/metabolismo , Anemia/terapia , Animales , Diagnóstico por Imagen/métodos , Índices de Eritrocitos , Transfusión de Eritrocitos/efectos adversos , Transfusión de Eritrocitos/métodos , Eritropoyesis , Regulación de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Hierro/metabolismo , Redes y Vías Metabólicas , Oximetría/métodos , Oxígeno/sangre , Resultado del Tratamiento
7.
Blood ; 129(7): 896-905, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-27864296

RESUMEN

Since 2012, a number of case reports have described the occurrence of thrombotic microangiopathy (TMA) following IV abuse of extended-release oxymorphone hydrochloride (Opana ER), an oral opioid for long-term treatment of chronic pain. Here, we present unique clinical features of 3 patients and investigate IV exposure to the tablet's inert ingredients as a possible causal mechanism. Guinea pigs were used as an animal model to understand the hematopathologic and nephrotoxic potential of the inert ingredient mixture (termed here as PEO+) which primarily contains high-molecular-weight polyethylene oxide (HMW PEO). Microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury were found in a group of 3 patients following recent injection of adulterated extended-release oxymorphone tablets. Varying degrees of cardiac involvement and retinal ischemia occurred, with TMA evident on kidney biopsy. A TMA-like state also developed in guinea pigs IV administered PEO+. Acute tubular and glomerular renal injury was accompanied by nonheme iron deposition and hypoxia-inducible factor-1α upregulation in the renal cortex. Similar outcomes were observed following dosing with HMW PEO alone. IV exposure to the inert ingredients in reformulated extended-release oxymorphone can elicit TMA. Although prescription opioid abuse shows geographic variation, all physicians should be highly inquisitive of IV drug abuse when presented with cases of TMA.


Asunto(s)
Analgésicos Opioides/efectos adversos , Oximorfona/efectos adversos , Microangiopatías Trombóticas/inducido químicamente , Microangiopatías Trombóticas/patología , Lesión Renal Aguda/sangre , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/patología , Analgésicos Opioides/administración & dosificación , Animales , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/efectos adversos , Femenino , Cobayas , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Oximorfona/administración & dosificación , Polietilenglicoles/efectos adversos , Microangiopatías Trombóticas/sangre , Microangiopatías Trombóticas/complicaciones
8.
Am J Physiol Lung Cell Mol Physiol ; 315(5): L765-L774, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30047285

RESUMEN

Intravascular sickling and lysis of red blood cells, a hallmark feature of sickle cell disease (SCD), releases hemoglobin (Hb) into the circulation. Increased cell-free Hb has been linked to vasculopathy and in vitro lipid oxidation. Scavenger plasma proteins haptoglobin (Hp) and hemopexin (Hpx) can attenuate cell-free Hb and total plasma heme lipid-oxidative capacity but are depleted in SCD. Here, we isolated lipids from BERK-SS mice, guinea pigs (GP) infused with heme-albumin, and patients with SCD undergoing regular exchange transfusion therapy and evaluated the level of lipid oxidation. Malondialdehyde formation, an end product of lipid peroxidation, was increased in BERK-SS mice, purified lipid fractions of the heme-albumin infused GP, and patients with SCD compared with controls. In humans, the extent of lipid oxidation was associated with the absence of Hp as well as decreased Hpx in plasma samples. Postmortem pulmonary tissue obtained from patients with SCD demonstrated oxidized LDL deposition in the pulmonary artery. The relationship between no Hp and low Hpx levels with greater LDL and HDL oxidation demonstrates the loss of protection against cell-free Hb and total plasma heme-mediated lipid oxidation and tissue injury in SCD. Strategies to protect against plasma lipid oxidation by cell-free Hb and total plasma heme (e.g., therapeutic Hp and Hpx replacement) may diminish the deleterious effects of cell-free Hb and total plasma heme toward the vascular system in SCD.


Asunto(s)
Anemia de Células Falciformes/fisiopatología , Haptoglobinas/metabolismo , Hemoglobinas/deficiencia , Hemopexina/deficiencia , Lípidos/química , Lipoproteínas/química , Adulto , Animales , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Cobayas , Hemo/química , Humanos , Peroxidación de Lípido , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Oxidación-Reducción , Adulto Joven
9.
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
10.
Transfusion ; 58(6): 1474-1485, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29498054

RESUMEN

BACKGROUND: Red blood cell (RBC) oxygen (O2 ) delivery may be impacted at the tissue, cellular, and molecular levels after storage duration, preservation strategies, and pathogen reduction. Collectively, the preclinical measurement of arterial and venous PO2 , systemic blood flow, tissue hypoxia-inducible factors (HIFs), pimonidazole adduction, and erythropoietin (EPO) regulation can serve to elucidate differential RBC quality after storage and processing. STUDY DESIGN AND METHODS: Donor guinea pig blood was collected, leukoreduced, and stored at 4°C in AS-3 for 1 (fresh) or 14 (stored) days. RBC variables-2,3-diphosphoglycerate, adenosine triphosphate, hemoglobin, morphology, deformability, and in vivo recovery at 24 hours-were measured at each storage duration. Recipient guinea pigs were exchange transfused until 80% volume replacement was achieved. Arterial and venous blood gases, systemic blood flow, renal HIF-1α and HIF-2α, renal EPO mRNA, plasma EPO, and renal tissue pimonidazole adduction were measured after transfusion. RESULTS: RBC variables declined significantly with storage; however, hemolysis and in vivo recovery remained within the allowable limits for human blood storage. Posttransfusion arterial and venous PO2 and systemic blood flow decreased, and renal HIFs, EPO mRNA, and pimonidazole adducts increased. Subsequently, EPO accumulated in plasma indicating decreased O2 availability in the kidneys. Conversely, all variables remained at basal levels in the fresh blood group. CONCLUSION: The evaluation of renal O2 homeostasis after transfusion represents an effective approach to defining RBC quality between predicate and novel processing. Methods are adapted from standardized techniques and ideal for preclinical evaluation.


Asunto(s)
Homeostasis , Riñón/metabolismo , Oxígeno/metabolismo , Animales , Conservación de la Sangre , Transfusión de Eritrocitos , Eritrocitos/citología , Eritropoyetina/sangre , Cobayas , Modelos Animales , Oxígeno/fisiología
11.
J Proteome Res ; 14(2): 1089-100, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25566950

RESUMEN

Beta-thalassemia results from mutations of the ß-hemoglobin (Hbb) gene and reduced functional Hbb synthesis. Excess α-Hb causes globin chain aggregation, oxidation, cytoskeletal damage, and increased red blood cell clearance. These events result in anemia, altered iron homeostasis, and expansion of extramedullary erythropoiesis. Serum transferrin (Tf) is suggested to be an important regulator of erythropoiesis in murine models of thalassemia. The present study was conducted to establish a quantitative proteomic and transcriptomic analysis of transferrin-modulated extramedullary erythropoiesis in the spleen of wild type and thalassemic Hbb(th3/+) mice. Our LC-MS/MS protein analysis and mRNA sequencing data provide quantitative expression estimates of 1590 proteins and 24,581 transcripts of the murine spleen and characterize key processes of erythropoiesis and RBC homeostasis such as the whole heme synthesis pathway as well as critical components of the red blood cell antioxidant systems and the proliferative cell cycling pathway. The data confirm that Tf treatment of nontransfused Hbb(th3/+) mice induces a systematic correction of these processes at a molecular level. Tf treatment of Hbb(th3/+) mice for 60 days leads to a complete molecular restoration of the normal murine spleen phenotype. These findings support further investigation of plasma-derived Tf as a treatment for thalassemia.


Asunto(s)
Modelos Animales de Enfermedad , Eritropoyesis , Proteoma , Transcriptoma , Transferrina/uso terapéutico , Talasemia beta/tratamiento farmacológico , Animales , Ratones , Ratones Endogámicos C57BL , Talasemia beta/genética , Talasemia beta/metabolismo
12.
Haematologica ; 100(5): 611-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25616571

RESUMEN

Intermediate beta-thalassemia has a broad spectrum of sequelae and affected subjects may require occasional blood transfusions over their lifetime to correct anemia. Iron overload in intermediate beta-thalassemia results from a paradoxical intestinal absorption, iron release from macrophages and hepatocytes, and sporadic transfusions. Pathological iron accumulation in parenchyma is caused by chronic exposure to non-transferrin bound iron in plasma. The iron scavenger and transport protein transferrin is a potential treatment being studied for correction of anemia. However, transferrin may also function to prevent or reduce iron loading of tissues when exposure to non-transferrin bound iron increases. Here we evaluate the effects of apotransferrin administration on tissue iron loading and early tissue pathology in non-transfused and transfused Hbb(th3/+) mice. Mice with the Hbb(th3/+) phenotype have mild to moderate anemia and consistent tissue iron accumulation in the spleen, liver, kidneys and myocardium. Chronic apotransferrin administration resulted in normalization of the anemia. Furthermore, it normalized tissue iron content in the liver, kidney and heart and attenuated early tissue changes in non-transfused Hbb(th3/+) mice. Apotransferrin treatment was also found to attenuate transfusion-mediated increases in plasma non-transferrin bound iron and associated excess tissue iron loading. These therapeutic effects were associated with normalization of transferrin saturation and suppressed plasma non-transferrin bound iron. Apotransferrin treatment modulated a fundamental iron regulatory pathway, as evidenced by decreased erythroid Fam132b gene (erythroferrone) expression, increased liver hepcidin gene expression and plasma hepcidin-25 levels and consequently reduced intestinal ferroportin-1 in apotransferrin-treated thalassemic mice.


Asunto(s)
Apoproteínas/administración & dosificación , Eliminación de Gen , Hemocromatosis/genética , Hemocromatosis/patología , Transferrina/administración & dosificación , Globinas beta/genética , Animales , Transfusión Sanguínea , Proteínas de Transporte de Catión/sangre , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Índices de Eritrocitos/efectos de los fármacos , Femenino , Expresión Génica , Hemocromatosis/metabolismo , Hemocromatosis/terapia , Hepcidinas/sangre , Hierro/sangre , Hierro/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Miocardio/patología , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/patología , Esplenomegalia/tratamiento farmacológico , Transferrina/metabolismo
13.
Toxicol In Vitro ; 97: 105793, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38401745

RESUMEN

To combat opioid abuse, the U.S. Food and Drug Administration (FDA) released a comprehensive action plan to address opioid addiction, abuse, and overdose that included increasing the prevalence of abuse-deterrent formulations (ADFs) in opioid tablets. Polyethylene oxide (PEO) has been widely used as an excipient to deter abuse via nasal insufflation. However, changes in abuse patterns have led to unexpected shifts in abuse from the nasal route to intravenous injection. Case reports identify adverse effects similar to thrombotic thrombocytopenic purpura (TTP) syndrome following the intravenous (IV) abuse of opioids containing PEO excipient. Increased risk of IV opioid ADF abuse compared to clinical benefit of the drug led to the removal of one opioid product from the market in 2017. Because many generic drugs containing PEO are still in development, there is interest in assessing safety consistent with generic drug regulation and unintended uses. Currently, there are no guidelines or in vitro assessment tools to characterize the safety of PEO excipients taken via intravenous injection. To create a more robust excipient safety evaluation tool and to study the mechanistic basis of HMW PEO-induced TMA, a dynamic in vitro test system involving blood flow through a needle model has been developed.


Asunto(s)
Analgésicos Opioides , Trastornos Relacionados con Opioides , Humanos , Polietilenglicoles/toxicidad , Polímeros , Peso Molecular , Excipientes , Técnicas In Vitro
14.
Blood Transfus ; 22(4): 316-327, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38814883

RESUMEN

BACKGROUND: Quality assessment of modified or processed red blood cell (RBC) components, such as pathogen-reduced RBCs, using only in vitro testing may not always be predictive of in vivo performance. Mouse or rat in vivo models are limited by a lack of applicability to certain aspects of human RBC biology. Here, we used a guinea pig model to study the effects of riboflavin combined with UV light on the integrity of RBCs in vitro and following transfusion in vivo. MATERIALS AND METHODS: Guinea pig RBCs were collected from whole blood (WB) treated with varying UV doses (10, 20, 40 or 80 J/mL) in the presence of riboflavin (UVR-RBCs). In vitro tests for UVR-RBCs included hemolysis, osmotic fragility, and cellular morphology by scanning electron microscopy. Guinea pigs transfused with one-day post-treatment UVR-RBCs were evaluated for plasma hemoglobin (Hb), non-transferrin bound iron (NTBI), total iron and Perls-detectable hemosiderin deposition in the spleen and kidney, and renal uptake of Hb. RESULTS: Acute RBC injury was dose dependently accelerated after treatment with UV light in the presence of riboflavin. Aberrant RBC morphology was evident at 20, 40, and 80 J/mL, and membrane lysis with Hb release was prominent at 80 J/mL. Guinea pigs transfused with 40 and 80 J/mL UVR-RBCs showed increased plasma Hb levels, and plasma NTBI was elevated in all UVR-RBC groups (10-80 J/mL). Total iron levels and Perls-hemosiderin staining in spleen and kidney as well as Hb uptake in renal proximal tubules were increased 8 hours post-transfusion with 40 and 80 J/mL UVR-RBCs. DISCUSSION: UVR-RBCs administered to guinea pigs increased markers of intravascular and extravascular hemolysis in a UV dose-dependent manner. This model may allow for the discrimination of RBC injury during testing of extensively processed RBCs intended for transfusion.


Asunto(s)
Eritrocitos , Hemólisis , Riboflavina , Rayos Ultravioleta , Animales , Riboflavina/farmacología , Cobayas , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Hemólisis/efectos de los fármacos , Transfusión de Eritrocitos , Fragilidad Osmótica/efectos de los fármacos , Humanos , Masculino , Hemoglobinas
15.
Front Med (Lausanne) ; 10: 1158359, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37384048

RESUMEN

Hemoglobin-based oxygen carriers (HBOCs) are being developed as oxygen and volume replacement therapeutics, however, their molecular and cellular effects on the vasculature and different organ systems are not fully defined. Using a guinea pig transfusion model, we examined the renal glomerular and tubular responses to PolyHeme, a highly characterized glutaraldehyde-polymerized human hemoglobin with low tetrameric hemoglobin content. PolyHeme-infused animals showed no major changes in glomerular histology or loss of specific markers of glomerular podocytes (Wilms tumor 1 protein, podocin, and podocalyxin) or endothelial cells (ETS-related gene and claudin-5) after 4, 24, and 72 h. Relative to sham controls, PolyHeme-infused animals also showed similar expression and subcellular distribution of N-cadherin and E-cadherin, two key epithelial junctional proteins of proximal and distal tubules, respectively. In terms of heme catabolism and iron-handling responses, PolyHeme induced a moderate but transient expression of heme oxygenase-1 in proximal tubular epithelium and tubulointerstitial macrophages that was accompanied by increased iron deposition in tubular epithelium. Contrary to previous findings with other modified or acellular hemoglobins, the present data show that PolyHeme does not disrupt the junctional integrity of the renal glomerulus and tubular epithelium, and triggers moderate activation of heme catabolic and iron sequestration systems likely as part of a renal adaptive response.

16.
Int J Pharm ; 632: 122557, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36584863

RESUMEN

Intravenous administration of abuse-deterrent opioid products poses high safety risks, in part due to the presence of high molecular weight polymeric excipients. Previous in vivo studies in animal models have shown that the higher molecular weight (Mw) polymeric excipients like polyethylene oxide (PEO) were directly linked to such adverse responses as intravenous hemolysis and kidney damage. PEO polymers have been widely used in abuse-deterrent formulations (ADF) of opioid products, adding to concerns over the general safety of the opioid category due to the unknown safety risk from abuse via unintended routes. The current study focused on the determination of the critical overlap concentration (c*) at various PEO molecular weights to aid in explaining differences in observed adverse responses from previous animal studies on the intravenous administration of PEO solutions. Adverse in vivo responses may be related to the viscoelastic regime of the polymer solution, which depends not only on Mw but also on concentration. Having a localized polymer concentration in the blood above the c*, i.e., the transition from the dilute to semi-dilute entangled viscoelastic regime, may influence the flow behavior and interactions of cells in the blood. The relationship of c* to this combination of physical, chemical, and rheological effects is a possible driving force behind adverse in vivo responses.


Asunto(s)
Analgésicos Opioides , Trastornos Relacionados con Opioides , Humanos , Excipientes , Polietilenglicoles/efectos adversos , Polietilenglicoles/química , Composición de Medicamentos , Administración Intravenosa , Trastornos Relacionados con Opioides/prevención & control
17.
Biochim Biophys Acta ; 1809(4-6): 262-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21558026

RESUMEN

Hypoxia inducible factor (HIF-1α) is a master regulator of tissue adaptive responses to hypoxia whose stability is controlled by an iron containing prolyl hydroxylase domain (PHD) protein. A catalytic redox cycle in the PHD's iron center that results in the formation of a ferryl (Fe(+4)) intermediate has been reported to be responsible for the hydroxylation and subsequent degradation of HIF-1α under normoxia. We show that induction of HIF-1α in rat kidneys can be achieved by iron reduction by the hydroxypyridin-4 one (CP94), an iron chelator administered intraperitoneally in rats. The extent of HIF protein stabilization as well as the expression of HIF target genes, including erythropoietin (EPO), in kidney tissues was comparable to those induced by known inhibitors of the PHD enzyme, such as desferrioxamine (DFO) and cobalt chloride (CoCl(2)). In human kidney cells and in vitro PHD activity assay, we were able to show that the HIF-1α protein can be stabilized by addition of CP94. This appears to inactivate PHD; and thus prevents the hydroxylation of HIF-1α. In conclusion, we have identified the inhibition of iron-binding pocket of PHD as an underlying mechanism of HIF induction in vivo and in vitro by a bidentate hydroxypyridinone.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quelantes del Hierro/farmacología , Riñón/efectos de los fármacos , Piridonas/farmacología , Animales , Sitios de Unión , Western Blotting , Línea Celular , Cobalto/farmacología , Deferiprona , Deferoxamina/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Hidroxilación/efectos de los fármacos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inyecciones Intraperitoneales , Quelantes del Hierro/administración & dosificación , Riñón/metabolismo , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Procolágeno-Prolina Dioxigenasa/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , Piridonas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sideróforos/farmacología
18.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L312-26, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22728465

RESUMEN

Cell-free hemoglobin (Hb) exposure may be a pathogenic mediator in the development of pulmonary arterial hypertension (PAH), and when combined with chronic hypoxia the potential for exacerbation of PAH and vascular remodeling is likely more pronounced. We hypothesized that Hb may contribute to hypoxia-driven PAH collectively as a prooxidant, inflammatory, and nitric oxide (NO) scavenger. Using programmable micropump technology, we exposed male Sprague-Dawley rats housed under room air or hypoxia to 12 or 30 mg per day Hb for 3, 5, and 7 wk. Blood pressure, cardiac output, right ventricular hypertrophy, and indexes of pulmonary vascular remodeling were evaluated. Additionally, markers of oxidative stress, NO bioavailability and inflammation were determined. Hb increased pulmonary arterial (PA) pressure, pulmonary vessel wall stiffening, and right heart hypertrophy with temporal and dose dependence in both room air and hypoxic cohorts. Hb induced a modest increase in plasma oxidative stress markers (malondialdehyde and 4-hydroxynonenal), no change in NO bioavailability, and increased lung ICAM protein expression. Treatment with the antioxidant Tempol attenuated Hb-induced pulmonary arterial wall thickening, but not PA pressures or ICAM expression. Chronic exposure to low plasma Hb concentrations (range = 3-10 µM) lasting up to 7 wk in rodents induces pulmonary vascular disease via inflammation and to a lesser extent by Hb-mediated oxidation. Tempol demonstrated a modest effect on the attenuation of Hb-induced pulmonary vascular disease. NO bioavailability was found to be of minimal importance in this model.


Asunto(s)
Hemoglobinas/efectos adversos , Inflamación/patología , Enfermedades Pulmonares/inducido químicamente , Enfermedades Vasculares/inducido químicamente , Animales , Presión Sanguínea/efectos de los fármacos , Western Blotting , Gasto Cardíaco/efectos de los fármacos , Óxidos N-Cíclicos/farmacología , Hemodinámica/efectos de los fármacos , Hemoglobinas/administración & dosificación , Hemoglobinas/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Inflamación/complicaciones , Bombas de Infusión , Molécula 1 de Adhesión Intercelular/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Enfermedades Pulmonares/sangre , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/orina , Masculino , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Marcadores de Spin , Enfermedades Vasculares/sangre , Enfermedades Vasculares/patología , Enfermedades Vasculares/orina
19.
Front Physiol ; 13: 845347, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35388289

RESUMEN

Unlike other rodents, guinea pigs (Cavia porcellus) have evolutionarily lost their capacity to synthesize vitamin C (ascorbate) de novo and, like several non-human primates and humans, rely on dietary intake and glutathione-dependent recycling to cope with oxidant stress. This is particularly relevant in red blood cell physiology, and especially when modeling blood storage, which exacerbates erythrocyte oxidant stress. Herein we provide a comprehensive metabolomics analysis of fresh and stored guinea pig red blood cell concentrates (n = 20), with weekly sampling from storage day 0 through 42. Results were compared to previously published ZOOMICS studies on red blood cells from three additional species with genetic loss of L-gulonolactone oxidase function, including humans (n = 21), olive baboons (n = 20), and rhesus macaques (n = 20). While metabolic trends were comparable across all species, guinea pig red blood cells demonstrated accelerated alterations of the metabolic markers of the storage lesion that are consistent with oxidative stress. Compared to the other species, guinea pig red blood cells showed aberrant glycolysis, pentose phosphate pathway end product metabolites, purine breakdown products, methylation, glutaminolysis, and markers of membrane lipid remodeling. Consistently, guinea pig red blood cells demonstrated higher end storage hemolysis, and scanning electron microscopy confirmed a higher degree of morphological alterations of their red blood cells, as compared to the other species. Despite a genetic inability to produce ascorbate that is common to the species evaluated, guinea pig red blood cells demonstrate accelerated oxidant stress under standard storage conditions. These data may offer relevant insights into the basal and cold storage metabolism of red blood cells from species that cannot synthesize endogenous ascorbate.

20.
Biochemistry ; 50(45): 9752-66, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21977904

RESUMEN

We have previously shown that hydrogen peroxide (H(2)O(2)) triggers irreversible oxidation of amino acids exclusive to the ß-chains of purified human hemoglobin (HbAo). However, it is not clear, whether α- or ß-subunit Hb variants exhibit different oxidative resistance to H(2)O(2) when compared to their native HbAo. Hb Providence contains two ß-subunit variants with single amino acid mutations at ßLys82→Asp (ßK82D) and at ßLys82→Asn (ßK82N) positions and binds oxygen at lower affinity than wild type HbA. We have separated Hb Providence into its 3 component fractions, and contrasted oxidative reactions of its ß-mutant fractions with HbAo. Relative to HbAo, both ßK82N and ßK82D fractions showed similar autoxidation kinetics and similar initial oxidation reaction rates with H(2)O(2). However, a more profound pattern of changes was seen in HbAo than in the two Providence fractions. The structural changes in HbAo include a collapse of ß-subunits, and α-α dimer formation in the presence of excess H(2)O(2). Mass spectrometric and amino acid analysis revealed that ßCys93 and ßCys112 were oxidized in the HbAo fraction, consistent with oxidative pathways driven by a ferrylHb and its protein radical. These amino acids were oxidized at a lesser extent in ßK82D fraction. While the 3 isolated components of Hb Providence exhibited similar ligand binding and oxidation reaction kinetics, the variant fractions were more effective in consuming H(2)O(2) and safely internalizing radicals through the ferric/ferryl pseudoperoxidase cycle.


Asunto(s)
Hemoglobina A/química , Hemoglobina A/metabolismo , Hemoglobina J/química , Hemoglobina J/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Óxidos N-Cíclicos , Ácido Cisteico/química , Dimerización , Globinas/química , Hemo/química , Hemoglobina A/genética , Hemoglobina J/genética , Humanos , Técnicas In Vitro , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Estrés Oxidativo , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Subunidades de Proteína , Espectrometría de Masa por Ionización de Electrospray , Marcadores de Spin , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA