Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Biomacromolecules ; 18(5): 1532-1543, 2017 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-28350442

RESUMEN

Polymer-protein conjugates can be engineered to self-assemble into discrete and well-defined drug delivery systems, which combine the advantages of receptor targeting and controlled drug release. We designed specific conjugates of the iron-binding and transport protein, transferrin (Tf), to combine the advantages of this serum-stable protein as a targeting agent for cancer cells with self-assembling polymers to act as carriers of cytotoxic drugs. Tf variants were expressed with cysteine residues at sites spanning different regions of the protein surface, and the polymer conjugates grown from these variants were compared with polymer conjugates grown from nonselectively derivatized sites on native Tf. The resulting synthetic biopolymer hybrids were evaluated for self-assembly properties, size and topology, ability to carry an anticancer drug (paclitaxel), and cytotoxicity with and without a drug payload in a representative human colon cancer cell line. The results demonstrated that the engineered Tf variant polymer conjugates formed better-defined self-assembled nanoparticles than the nonselectively derivatized conjugates and showed greater efficacy in paclitaxel delivery. A polymer conjugate grown from a specific Tf variant, S415C was found to be taken up rapidly into cancer cells expressing the Tf-receptor, and, while tolerated well by cells in the absence of drugs, was as cytotoxic as free paclitaxel, when loaded with the drug. Importantly, the S415C conjugate polymer was not the most active variant in Tf-receptor binding, suggesting that the nanoscale self-assembly of the polymer-protein hybrid is also a key factor in delivery efficacy. The data overall suggest new design rules for polymer-biopolymer hybrids and therapeutic delivery systems, which include engineering specific residues for conjugation that mediate nanoscale assembly as well as control of ligand-receptor interactions to target specific cell types.


Asunto(s)
Nanoconjugados/química , Transferrina/química , Antineoplásicos/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Células HCT116 , Humanos , Células MCF-7 , Nanoconjugados/efectos adversos , Paclitaxel/administración & dosificación
2.
J Biol Chem ; 289(19): 13492-502, 2014 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-24652290

RESUMEN

A major challenge for the therapeutic use of many peptides and proteins is their short circulatory half-life. Albumin has an extended serum half-life of 3 weeks because of its size and FcRn-mediated recycling that prevents intracellular degradation, properties shared with IgG antibodies. Engineering the strictly pH-dependent IgG-FcRn interaction is known to extend IgG half-life. However, this principle has not been extensively explored for albumin. We have engineered human albumin by introducing single point mutations in the C-terminal end that generated a panel of variants with greatly improved affinities for FcRn. One variant (K573P) with 12-fold improved affinity showed extended serum half-life in normal mice, mice transgenic for human FcRn, and cynomolgus monkeys. Importantly, favorable binding to FcRn was maintained when a single-chain fragment variable antibody was genetically fused to either the N- or the C-terminal end. The engineered albumin variants may be attractive for improving the serum half-life of biopharmaceuticals.


Asunto(s)
Albúminas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Albúminas/genética , Albúminas/farmacología , Sustitución de Aminoácidos , Animales , Femenino , Semivida , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/farmacología , Humanos , Macaca fascicularis , Ratones , Mutación Missense , Receptores Fc/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
3.
J Biol Chem ; 289(24): 17228-39, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24764301

RESUMEN

Albumin is the most abundant protein in blood and plays a pivotal role as a multitransporter of a wide range of molecules such as fatty acids, metabolites, hormones, and toxins. In addition, it binds a variety of drugs. Its role as distributor is supported by its extraordinary serum half-life of 3 weeks. This is related to its size and binding to the cellular receptor FcRn, which rescues albumin from intracellular degradation. Furthermore, the long half-life has fostered a great and increasing interest in utilization of albumin as a carrier of protein therapeutics and chemical drugs. However, to fully understand how FcRn acts as a regulator of albumin homeostasis and to take advantage of the FcRn-albumin interaction in drug design, the interaction interface needs to be dissected. Here, we used a panel of monoclonal antibodies directed towards human FcRn in combination with site-directed mutagenesis and structural modeling to unmask the binding sites for albumin blocking antibodies and albumin on the receptor, which revealed that the interaction is not only strictly pH-dependent, but predominantly hydrophobic in nature. Specifically, we provide mechanistic evidence for a crucial role of a cluster of conserved tryptophan residues that expose a pH-sensitive loop of FcRn, and identify structural differences in proximity to these hot spot residues that explain divergent cross-species binding properties of FcRn. Our findings expand our knowledge of how FcRn is controlling albumin homeostasis at a molecular level, which will guide design and engineering of novel albumin variants with altered transport properties.


Asunto(s)
Albúminas/metabolismo , Antígenos de Histocompatibilidad Clase I/química , Receptores Fc/química , Secuencia de Aminoácidos , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Receptores Fc/genética , Receptores Fc/inmunología , Receptores Fc/metabolismo
4.
J Biol Chem ; 288(33): 24277-85, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818524

RESUMEN

Albumin has a serum half-life of 3 weeks in humans. This has been utilized to extend the serum persistence of biopharmaceuticals that are fused to albumin. In light of the fact that the neonatal Fc receptor (FcRn) is a key regulator of albumin homeostasis, it is crucial to address how fusion of therapeutics to albumin impacts binding to FcRn. Here, we report on a detailed molecular investigation on how genetic fusion of a short peptide or an single-chain variable fragment (scFv) fragment to human serum albumin (HSA) influences pH-dependent binding to FcRn from mouse, rat, monkey, and human. We have found that fusion to the N- or C-terminal end of HSA only slightly reduces receptor binding, where the most noticeable effect is seen after fusion to the C-terminal end. Furthermore, in contrast to the observed strong binding to human and monkey FcRn, HSA and all HSA fusions bound very poorly to mouse and rat versions of the receptor. Thus, we demonstrate that conventional rodents are limited as preclinical models for analysis of serum half-life of HSA-based biopharmaceuticals. This finding is explained by cross-species differences mainly found within domain III (DIII) of albumin. Our data demonstrate that although fusion, particularly to the C-terminal end, may slightly reduce the affinity for FcRn, HSA is versatile as a carrier of biopharmaceuticals.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Albúmina Sérica/metabolismo , Albúmina Sérica/uso terapéutico , Anticuerpos de Cadena Única/metabolismo , Animales , Unión Competitiva , Semivida , Humanos , Cinética , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Especificidad de la Especie
5.
Biochim Biophys Acta ; 1830(12): 5526-34, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23639804

RESUMEN

BACKGROUND: Albumin is the most abundant plasma protein, is highly soluble, very stable and has an extraordinarily long circulatory half-life as a direct result of its size and interaction with the FcRn mediated recycling pathway. In contrast, many therapeutic molecules are smaller than the renal filtration threshold and are rapidly lost from the circulation thereby limiting their therapeutic potential. Albumin can be used in a variety of ways to increase the circulatory half-life of such molecules. SCOPE OF REVIEW: This article will review the mechanisms which underpin albumin's extraordinarily long circulatory half-life and how the understanding of these processes are currently being employed to extend the circulatory half-life of drugs which can be engineered to bind to albumin, or are conjugated to, or genetically fused to, albumin. MAJOR CONCLUSIONS: The recent and growing understanding of the pivotal role of FcRn in maintaining the extended circulatory half-life of albumin will necessitate a greater and more thorough investigation of suitable pre-clinical model systems for assessing the pharmacokinetic profiles of drugs associated, conjugated or fused to albumin. GENERAL SIGNIFICANCE: Association, conjugation or fusion of therapeutic drugs to albumin is a well-accepted and established half-life extension technology. The manipulation of the albumin-FcRn interaction will facilitate the modulation of the circulatory half-life of albumin-enabled drugs, leading to superior pharmacokinetics tailored to the disease state and increased patient compliance. This article is part of a Special Issue entitled Serum Albumin.


Asunto(s)
Farmacocinética , Albúmina Sérica/metabolismo , Animales , Semivida , Humanos , Inmunoglobulina G/metabolismo , Receptores Fc/metabolismo
6.
J Am Chem Soc ; 134(3): 1454-7, 2012 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-22239162

RESUMEN

Albumin transports both fatty acids and zinc in plasma. Competitive binding studied by isothermal titration calorimetry revealed that physiologically relevant levels of fatty acids modulate the Zn-binding capacity of albumin, with far-reaching implications for biological zinc speciation. The molecular mechanism for this effect is likely due to a large conformational change elicited by fatty acid binding to a high-affinity interdomain site that disrupts at least one Zn site. Albumin may be a molecular device to "translate" certain aspects of the organismal energy state into global zinc signals.


Asunto(s)
Ácidos Grasos/sangre , Ácidos Grasos/metabolismo , Albúmina Sérica/metabolismo , Zinc/sangre , Zinc/metabolismo , Transporte Biológico , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Albúmina Sérica/química
7.
Front Immunol ; 13: 892534, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35757719

RESUMEN

Serum albumin (SA), the most abundant soluble protein in the body, maintains plasma oncotic pressure and regulates the distribution of vascular fluid and has a range of other important functions. The goals of this review are to expand clinical knowledge regarding the functions of SA, elucidate effects of dysregulated SA concentration, and discuss the clinical relevance of hypoalbuminemia resulting from various diseases. We discuss potential repercussions of SA dysregulation on cholesterol levels, liver function, and other processes that rely on its homeostasis, as decreased SA concentration has been shown to be associated with increased risk for cardiovascular disease, hyperlipidemia, and mortality. We describe the anti-inflammatory and antioxidant properties of SA, as well as its ability to bind and transport a plethora of endogenous and exogenous molecules. SA is the primary serum protein involved in binding and transport of drugs and as such has the potential to affect, or be affected by, certain medications. Of current relevance are antibody-based inhibitors of the neonatal Fc receptor (FcRn), several of which are under clinical development to treat immunoglobulin G (IgG)-mediated autoimmune disorders; some have been shown to decrease SA concentration. FcRn acts as a homeostatic regulator of SA by rescuing it, as well as IgG, from intracellular degradation via a common cellular recycling mechanism. Greater clinical understanding of the multifunctional nature of SA and the potential clinical impact of decreased SA are needed; in particular, the potential for certain treatments to reduce SA concentration, which may affect efficacy and toxicity of medications and disease progression.


Asunto(s)
Enfermedades Autoinmunes , Inmunoglobulina G , Enfermedades Autoinmunes/tratamiento farmacológico , Homeostasis , Humanos , Inmunoglobulina G/metabolismo , Recién Nacido , Receptores Fc , Albúmina Sérica/metabolismo
8.
J Biol Chem ; 284(34): 23116-24, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19520864

RESUMEN

Most blood plasma zinc is bound to albumin, but the structure of the binding site has not been determined. Zn K-edge extended x-ray absorption fine structure spectroscopy and modeling studies show that the major Zn(2+) site on albumin is a 5-coordinate site with average Zn-O/N distances of 1.98 A and a weak sixth O/N bond of 2.48 A, consistent with coordination to His(67) and Asn(99) from domain I, His(247) and Asp(249) from domain II (residues conserved in all sequenced mammalian albumins), plus a water ligand. The dynamics of the domain I/II interface, thought to be important to biological function, are affected by Zn(2+) binding, which induces cooperative allosteric effects related to those of the pH-dependent neutral-to-base transition. N99D and N99H mutations enhance Zn(2+) binding but alter protein stability, whereas mutation of His(67) to alanine removes an interdomain H-bond and weakens Zn(2+) binding. Both wild-type and mutant albumins promote the safe management of high micromolar zinc concentrations for cells in cultures.


Asunto(s)
Modelos Moleculares , Albúmina Sérica/química , Albúmina Sérica/metabolismo , Zinc/química , Zinc/metabolismo , Sitios de Unión , Línea Celular , Humanos , Espectroscopía de Resonancia Magnética , Mutación , Unión Proteica , Ingeniería de Proteínas , Estructura Secundaria de Proteína , Albúmina Sérica/genética , Relación Estructura-Actividad
9.
Protein Expr Purif ; 73(2): 113-24, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20546898

RESUMEN

An expression system is described for the production of monomeric scFvs and scFv antibody fragments genetically fused to human albumin (at either the N- or C-terminus or both). Based upon strains of Saccharomyces cerevisiae originally developed for the production of a recombinant human albumin (Recombumin) this system has delivered high levels of secreted product into the supernatant of shake flask and high cell density fed-batch fermentations. Specific binding to the corresponding ligand was demonstrated for each of the scFvs and scFv-albumin fusions and pharmacokinetic studies showed that the fusion products had greatly extended circulatory half-lives. The system described provides an attractive alternative to other microbial systems for the manufacture of this type of product.


Asunto(s)
Albúminas/metabolismo , Fusión Artificial Génica , Saccharomyces cerevisiae/metabolismo , Albúmina Sérica/genética , Anticuerpos de Cadena Única/inmunología , Albúminas/genética , Animales , Área Bajo la Curva , Reactores Biológicos , Fermentación/genética , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/metabolismo , Semivida , Humanos , Tasa de Depuración Metabólica , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/farmacocinética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/inmunología , Albúmina Sérica/metabolismo , Anticuerpos de Cadena Única/genética
10.
Microb Cell Fact ; 9: 87, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21083917

RESUMEN

BACKGROUND: Animal-free recombinant proteins provide a safe and effective alternative to tissue or serum-derived products for both therapeutic and biomanufacturing applications. While recombinant insulin and albumin already exist to replace their human counterparts in cell culture media, until recently there has been no equivalent for serum transferrin. RESULTS: The first microbial system for the high-level secretion of a recombinant transferrin (rTf) has been developed from Saccharomyces cerevisiae strains originally engineered for the commercial production of recombinant human albumin (Novozymes' Recombumin® USP-NF) and albumin fusion proteins (Novozymes' albufuse®). A full-length non-N-linked glycosylated rTf was secreted at levels around ten-fold higher than from commonly used laboratory strains. Modification of the yeast 2 µm-based expression vector to allow overexpression of the ER chaperone, protein disulphide isomerase, further increased the secretion of rTf approximately twelve-fold in high cell density fermentation. The rTf produced was functionally equivalent to plasma-derived transferrin. CONCLUSIONS: A Saccharomyces cerevisiae expression system has enabled the cGMP manufacture of an animal-free rTf for industrial cell culture application without the risk of prion and viral contamination, and provides a high-quality platform for the development of transferrin-based therapeutics.


Asunto(s)
Saccharomyces cerevisiae/metabolismo , Transferrina/biosíntesis , Recuento de Células , Fermentación , Glicosilación , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Transferrina/química , Transferrina/genética
11.
Biochem Biophys Res Commun ; 386(4): 666-70, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19555658

RESUMEN

Thymidine phosphorylase (TP) first identified as platelet derived endothelial cell growth factor (PD-ECGF) plays a key role in nucleoside metabolism. Human TP (hTP) is implicated in angiogenesis and is overexpressed in several solid tumors. Here, we report the crystal structures of recombinant hTP and its complex with a substrate 5-iodouracil (5IUR) at 3.0 and 2.5A, respectively. In addition, we provide information on the role of specific residues in the enzymatic activity of hTP through mutagenesis and kinetic studies.


Asunto(s)
Timidina Fosforilasa/química , Uracilo/análogos & derivados , Cristalografía por Rayos X , Humanos , Mutación , Fosfatos/química , Conformación Proteica , Timidina Fosforilasa/genética , Uracilo/química
12.
FEBS J ; 272(2): 353-62, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15654874

RESUMEN

Cys34 in domain I of the three-domain serum protein albumin is the binding site for a wide variety of biologically and clinically important small molecules, provides antioxidant activity, and constitutes the largest portion of free thiol in blood. Analysis of X-ray structures of albumin reveals that the loop containing Tyr84 occurs in multiple conformations. In structures where the loop is well defined, there appears to be an H-bond between the OH of Tyr84 and the sulfur of Cys34. We show that the reaction of 5,5'-dithiobis(2-nitrobenzoic acid) (DTNB) with Tyr84Phe mutant albumin is approximately four times faster than with the wild-type protein between pH 6 and pH 8. In contrast, the His39Leu mutant reacts with DTNB more slowly than the wild-type protein at pH < 8, but at a similar rate at pH 8. Above pH 8 there is a dramatic increase in reactivity for the Tyr84Phe mutant. We also report (1)H NMR studies of disulfide interchange reactions with cysteine. The tethering of the two loops containing Tyr84 and Cys34 not only appears to control the redox potential and accessibility of Cys34, but also triggers the transmission of information about the state of Cys34 throughout domain I, and to the domainI/II interface.


Asunto(s)
Albúmina Sérica/química , Sitios de Unión , Disulfuros/química , Humanos , Concentración de Iones de Hidrógeno , Oxidación-Reducción , Proteínas Recombinantes/química
13.
Expert Opin Drug Deliv ; 12(5): 793-812, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25518870

RESUMEN

INTRODUCTION: Rapid clearance of drugs from the body results in short therapeutic half-life and is an integral property of many protein and peptide-based drugs. To maintain the desired therapeutic effect patients are required to administer higher doses more frequently, which is inconvenient and risks undesirable side effects. Drug delivery technologies aim to minimise the number of administrations and dose-related toxicity while maximising therapeutic efficacy. AREAS COVERED: This review describes albumin's inherent biochemical and biophysical properties, which make it an attractive drug delivery platform and the developmental status of drugs that are associated, conjugated or genetically fused with albumin. Albumin interacts with a number of cell surface receptors including gp18, gp30, gp60, FcRn, cubilin and megalin. The importance of albumin's interaction with the FcRn receptor, the basis for albumin's long circulatory half-life, is described, as are engineered albumins with improved pharmacokinetics. Albumin naturally accumulates at tumours and sites of inflammation, a characteristic which can be augmented by the addition of targeting ligands. The development of albumin drug conjugates which reply upon this property is described. EXPERT OPINION: Albumin's inherent biochemical and biophysical properties make it an ideal drug delivery platform. Recent advances in our understanding of albumin physiology and the improvement in albumin-based therapies strongly suggest that albumin-based therapies have a significant advantage over alternative technologies in terms of half-life, stability, versatility, safety and ease of manufacture. Given the importance of the albumin:FcRn interaction, the interpretation of the pharmacokinetic and pharmacodynamic profiles of albumin-based therapeutics with disturbed albumin:FcRn interaction may have to be reassessed. The FcRn receptor has additional functionality, especially in relation to immunology, antigen presentation and delivery of proteins across mucosal membranes, consequently albumin-based fusions and conjugates may have a future role in oral and pulmonary-based vaccines and drug delivery.


Asunto(s)
Albúminas/química , Sistemas de Liberación de Medicamentos , Receptores de Superficie Celular/metabolismo , Animales , Semivida , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Farmacocinética , Receptores Fc/metabolismo
14.
Nat Commun ; 3: 610, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22215085

RESUMEN

Albumin is the most abundant protein in blood where it has a pivotal role as a transporter of fatty acids and drugs. Like IgG, albumin has long serum half-life, protected from degradation by pH-dependent recycling mediated by interaction with the neonatal Fc receptor, FcRn. Although the FcRn interaction with IgG is well characterized at the atomic level, its interaction with albumin is not. Here we present structure-based modelling of the FcRn-albumin complex, supported by binding analysis of site-specific mutants, providing mechanistic evidence for the presence of pH-sensitive ionic networks at the interaction interface. These networks involve conserved histidines in both FcRn and albumin domain III. Histidines also contribute to intramolecular interactions that stabilize the otherwise flexible loops at both the interacting surfaces. Molecular details of the FcRn-albumin complex may guide the development of novel albumin variants with altered serum half-life as carriers of drugs.


Asunto(s)
Albúminas/química , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/genética , Mutagénesis , Receptores Fc/química , Receptores Fc/genética , Sitios de Unión , Escherichia coli/metabolismo , Histidina/química , Humanos , Concentración de Iones de Hidrógeno , Inmunoglobulina G/química , Iones , Modelos Moleculares , Conformación Molecular , Mutagénesis Sitio-Dirigida , Mutación , Estructura Terciaria de Proteína , Albúmina Sérica/química , Factores de Tiempo
15.
Proc Natl Acad Sci U S A ; 100(7): 3701-6, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12598656

RESUMEN

Albumin is the major transport protein in blood for Zn(2+), a metal ion required for physiological processes and recruited by various drugs and toxins. However, the Zn(2+)-binding site(s) on albumin is ill-defined. We have analyzed the 18 x-ray crystal structures of human albumin in the PDB and identified a potential five-coordinate Zn site at the interface of domains I and II consisting of N ligands from His-67 and His-247 and O ligands from Asn-99, Asp-249, and H(2)O, which are the same amino acid ligands as those in the zinc enzymes calcineurin, endonucleotidase, and purple acid phosphatase. The site is preformed in unliganded apo-albumin and highly conserved in mammalian albumins. We have used (111)Cd NMR as a probe for Zn(2+) binding to recombinant human albumin. We show that His-67 --> Ala (His67Ala) mutation strongly perturbs Cd(2+) binding, whereas the mutations Cys34Ala, or His39Leu and Tyr84Phe (residues which may H-bond to Cys-34) have no effect. Weak Cl(-) binding to the fifth coordination site of Cd(2+) was demonstrated. Cd(2+) binding was dramatically affected by high fatty acid loading of albumin. Analysis of the x-ray structures suggests that fatty acid binding to site 2 triggers a spring-lock mechanism, which disengages the upper (His-67Asn-99) and lower (His-247Asp-249) halves of the metal site. These findings provide a possible mechanism whereby fatty acids (and perhaps other small molecules) could influence the transport and delivery of zinc in blood.


Asunto(s)
Albúmina Sérica/química , Zinc/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Calcio/metabolismo , Cristalografía por Rayos X/métodos , Histidina , Humanos , Cinética , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Albúmina Sérica/metabolismo , Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA