Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Nat Chem Biol ; 16(4): 379-382, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32198493

RESUMEN

Tyrosine sulfation is an important post-translational modification found in higher eukaryotes. Here we report an engineered tyrosyl-tRNA synthetase/tRNA pair that co-translationally incorporates O-sulfotyrosine in response to UAG codons in Escherichia coli and mammalian cells. This platform enables recombinant expression of eukaryotic proteins homogeneously sulfated at chosen sites, which was demonstrated by expressing human heparin cofactor II in mammalian cells in different states of sulfation.


Asunto(s)
Ingeniería de Proteínas/métodos , Somatomedinas/química , Tirosina/análogos & derivados , Animales , Codón de Terminación/metabolismo , Escherichia coli/metabolismo , Código Genético , Cofactor II de Heparina/metabolismo , Humanos , Mamíferos , Procesamiento Proteico-Postraduccional , Proteínas/química , Tirosina/química , Tirosina-ARNt Ligasa/metabolismo
2.
IUBMB Life ; 72(10): 2180-2193, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32827448

RESUMEN

Heparin cofactor II (HCII) is predominantly expressed in the liver and inhibits thrombin in blood plasma to influence the blood coagulation cascade. Its deficiency is associated with arterial thrombosis. Its cleavage by neutrophil elastase produces fragment that helps in neutrophil chemotaxis in the acute inflammatory response in human. In the present study, we have identified a novel alternatively spliced transcript of the HCII gene in human liver. This novel transcript includes an additional novel region in continuation with exon 3 called exon 3b. Exon 3b acts like an alternate last exon, and hence its inclusion in the transcript due to alternative splicing removes exon 4 and encodes for a different C-terminal region to give a novel protein, HCII-N. MD simulations of HCII-N and three-dimensional structure showed a unique 51 amino acid sequence at the C-terminal having unique RCL-like structure. The HCII-N protein purified from bacterial culture showed a protein migrating at lower molecular weight (MW 55 kDa) as compared to native HCII (MW 66 kDa). A fluorescence-based analysis revealed a more compact structure of HCII-N that was in a more hydrophilic environment. The HCII-N protein, however, showed no inhibitory activity against thrombin. Due to large conformational variation observed in comparison with native HCII, HCII-N may have alternate protease specificity or a non-inhibitory role. Western blot of HCII purified from large plasma volume showed the presence of a low MW 59 kDa band with no thrombin activity. This study provides the first evidence of alternatively spliced novel isoform of the HCII gene.


Asunto(s)
Cofactor II de Heparina/química , Cofactor II de Heparina/genética , Cofactor II de Heparina/metabolismo , Hígado/metabolismo , Empalme Alternativo , Factor Xa/metabolismo , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Isoformas de Proteínas , Espectrometría de Fluorescencia , Trombina/metabolismo , Activador de Tejido Plasminógeno/antagonistas & inhibidores , Activador de Tejido Plasminógeno/metabolismo
3.
Cytokine ; 126: 154876, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31629109

RESUMEN

Premature ovarian insufficiency (POI) is a primary ovarian defect characterized by premature depletion of ovarian follicles before 40 years of age. The disorder has been attributed to various causes, but the study of altered proteins in serum levels as the cause is rare. Additionally, identifying novel biomarkers can contribute to more accurate diagnosis or prognosis of POI. In the present study, a solid-phase antibody array simultaneously detecting multiple proteins was used to analyze POI serum with menopausal and healthy fertile subjects as control groups. As a result, compared to the menopause and healthy fertile groups, eleven proteins, including Neurturin, Frizzled-5, Serpin D1, MMP-7, ICAM-3, IL-17F, IFN-gamma R1, IL-29, IL-17R, IL-17C and Soggy-1, were uniquely down-regulated, and Afamin was particularly up-regulated in POI serum. More importantly, all of these factors were firstly found to be associated with POI in this study, suggesting that these proteins may participate in the pathogenesis of POI and may be novel serum biomarkers for POI.


Asunto(s)
Biomarcadores/sangre , Menopausia Prematura/sangre , Insuficiencia Ovárica Primaria/sangre , Adulto , Anticuerpos , Proteínas Portadoras/sangre , Regulación hacia Abajo , Estradiol/sangre , Femenino , Receptores Frizzled/sangre , Glicoproteínas/sangre , Cofactor II de Heparina/metabolismo , Humanos , Molécula 3 de Adhesión Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/sangre , Interferón gamma/sangre , Interferones/sangre , Interleucina-17/sangre , Interleucinas/sangre , Metaloproteinasa 7 de la Matriz/sangre , Persona de Mediana Edad , Neurturina/sangre , Insuficiencia Ovárica Primaria/inmunología , Insuficiencia Ovárica Primaria/patología , Análisis por Matrices de Proteínas , Receptores de Interleucina-17/sangre , Albúmina Sérica Humana , Regulación hacia Arriba
4.
Glycobiology ; 29(6): 446-451, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30869126

RESUMEN

The glycosaminoglycan dermatan sulfate (DS) is a well-known activator of heparin cofactor II-dependent inactivation of thrombin. In contrast to heparin, dermatan sulfate has never been prepared recombinantly from material of non-animal origin. Here we report on the enzymatic synthesis of structurally well-defined DS with high anticoagulant activity. Using a microbial K4 polysaccharide and the recombinant enzymes DS-epimerase 1, dermatan 4-O-sulfotransferase 1, uronyl 2-O-sulfotransferase and N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase, several new glycostructures have been prepared, such as a homogenously sulfated IdoA-GalNAc-4S polymer and its 2-O-, 6-O- and 2,6-O-sulfated derivatives. Importantly, the recombinant highly 2,4-O-sulfated DS inhibits thrombin via heparin cofactor II, approximately 20 times better than heparin, enabling manipulation of vascular and extravascular coagulation. The potential of this method can be extended to preparation of specific structures that are of importance for binding and activation of cytokines, and control of inflammation and metastasis, involving extravasation and migration.


Asunto(s)
Dermatán Sulfato/farmacología , Cofactor II de Heparina/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Trombina/antagonistas & inhibidores , Conformación de Carbohidratos , Dermatán Sulfato/síntesis química , Dermatán Sulfato/química , Humanos , Modelos Moleculares , Inhibidores de Serina Proteinasa/síntesis química , Inhibidores de Serina Proteinasa/química , Trombina/metabolismo
5.
Biochem Biophys Res Commun ; 483(3): 964-971, 2017 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-28087279

RESUMEN

Rupture of carotid atherosclerotic plaque may cause stroke, while few biomarker in clinic can evaluate carotid plaque vulnerability. In this study, we divided the recruited participants into no plaque, stable plaque, and vulnerable plaque group according to carotid ultrasonography, and screened the differentially expressed proteins in plasma of these participants using isobaric tags for relative and absolute quantitation labeling coupled with liquid chromatography-tandem mass spectrometry. 28 proteins were identified differentially expressed, among which alpha-2-macroglobulin (α2M) and heparin cofactor II (HCII) were found to be at hub position in the interactions of these proteins by STRING analysis and were selected for enzyme-linked immunosorbent assay measurement to assess their relevance with carotid plaques vulnerability and diagnostic efficiency. The plasma level of α2M was found positively correlated, while HCII level was negatively correlated with higher vulnerability of carotid plaques. Both proteins were efficient in differentiating stable and vulnerable carotid plaques. These findings provide potential new targets for the research of carotid plaque vulnerability. Plasma α2M and HCII may be potential biomarkers for evaluation of the vulnerability of carotid plaques if further studied.


Asunto(s)
Estenosis Carotídea/sangre , Cofactor II de Heparina/metabolismo , Placa Aterosclerótica/sangre , alfa-Macroglobulinas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Análisis Químico de la Sangre/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad
6.
Arterioscler Thromb Vasc Biol ; 36(5): e41-50, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27034473

RESUMEN

OBJECTIVE: Thrombin signaling promotes atherosclerosis by initiating inflammatory events indirectly through platelet activation and directly via protease-activated receptors. Therefore, endogenous thrombin inhibitors may be relevant modulators of atheroprogression and cardiovascular risk. In addition, endogenous thrombin inhibitors may affect the response to non-vitamin K-dependent oral anticoagulants. Here, the question was addressed whether the small leucine-rich proteoglycan biglycan acts as an endogenous thrombin inhibitor in atherosclerosis through activation of heparin cofactor II. APPROACH AND RESULTS: Biglycan concentrations were elevated in the plasma of patients with acute coronary syndrome and in male Apolipoprotein E-deficient (ApoE(-/-)) mice. Biglycan was detected in the glycocalyx of capillaries and the subendothelial matrix of arterioles of ApoE(-/-) mice and in atherosclerotic plaques. Thereby a vascular compartment is provided that may mediate the endothelial and subendothelial activation of heparin cofactor II through biglycan. ApoE and Bgn double-deficient (ApoE(-/-)/Bgn(-/0)) mice showed higher activity of circulating thrombin, increased platelet activation and platelet adhesion in vivo, supporting a role of biglycan in balancing thrombin activity. Furthermore, concentrations of circulating cytokines and aortic macrophage content were elevated in ApoE(-/-)/Bgn(-/0) mice, suggesting a proinflammatory phenotype. Elevated platelet activation and macrophage accumulation were reversed by treating ApoE(-/-)/Bgn(-/0) mice with the thrombin inhibitor argatroban. Ultimately, ApoE(-/-)/Bgn(-/0) mice developed aggravated atherosclerosis. CONCLUSIONS: The present results indicate that biglycan plays a previously unappreciated protective role during the progression of atherosclerosis by inhibiting thrombin activity, platelet activation, and finally macrophage-mediated plaque inflammation.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Biglicano/deficiencia , Inflamación/metabolismo , Trombina/metabolismo , Síndrome Coronario Agudo/sangre , Animales , Antitrombinas/farmacología , Aorta/efectos de los fármacos , Aorta/patología , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/prevención & control , Biglicano/sangre , Biglicano/genética , Citocinas/sangre , Modelos Animales de Enfermedad , Genotipo , Cofactor II de Heparina/metabolismo , Humanos , Inflamación/sangre , Inflamación/genética , Inflamación/prevención & control , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Placa Aterosclerótica , Activación Plaquetaria , Factores de Tiempo
7.
Proc Natl Acad Sci U S A ; 111(41): 14894-9, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25267637

RESUMEN

Patients with mucopolysaccharidosis type I (MPS I), a genetic deficiency of the lysosomal enzyme α-l-iduronidase (IDUA), exhibit accumulation of glycosaminoglycans in tissues, with resulting diverse clinical manifestations including neurological, ocular, skeletal, and cardiac disease. MPS I is currently treated with hematopoietic stem cell transplantation or weekly enzyme infusions, but these therapies have significant drawbacks for patient safety and quality of life and do not effectively address some of the most critical clinical sequelae, such as life-threatening cardiac valve involvement. Using the naturally occurring feline model of MPS I, we tested liver-directed gene therapy as a means of achieving long-term systemic IDUA reconstitution. We treated four MPS I cats at 3-5 mo of age with an adeno-associated virus serotype 8 vector expressing feline IDUA from a liver-specific promoter. We observed sustained serum enzyme activity for 6 mo at ∼ 30% of normal levels in one animal, and in excess of normal levels in three animals. Remarkably, treated animals not only demonstrated reductions in glycosaminoglycan storage in most tissues, but most also exhibited complete resolution of aortic valve lesions, an effect that has not been previously observed in this animal model or in MPS I patients treated with current therapies. These data point to clinically meaningful benefits of the robust enzyme expression achieved with hepatic gene transfer that extend beyond the economic and quality of life advantages over lifelong enzyme infusions.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Terapia Genética , Hígado/metabolismo , Mucopolisacaridosis I/terapia , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Enfermedades Cardiovasculares/patología , Gatos , Dependovirus/genética , Femenino , Vectores Genéticos/metabolismo , Glicosaminoglicanos/metabolismo , Cofactor II de Heparina/metabolismo , Iduronidasa/sangre , Iduronidasa/genética , Iduronidasa/uso terapéutico , Hígado/patología , Masculino , Datos de Secuencia Molecular , Mucopolisacaridosis I/sangre , Mucopolisacaridosis I/patología , Miocardio/metabolismo , Miocardio/patología , Trombina/metabolismo , Distribución Tisular , Transducción Genética
8.
Angew Chem Int Ed Engl ; 56(9): 2312-2317, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28124818

RESUMEN

Glycosaminoglycan (GAG) sequences that selectively target heparin cofactor II (HCII), a key serpin present in human plasma, remain unknown. Using a computational strategy on a library of 46 656 heparan sulfate hexasaccharides we identified a rare sequence consisting of consecutive glucuronic acid 2-O-sulfate residues as selectively targeting HCII. This and four other unique hexasaccharides were chemically synthesized. The designed sequence was found to activate HCII ca. 250-fold, while leaving aside antithrombin, a closely related serpin, essentially unactivated. This group of rare designed hexasaccharides will help understand HCII function. More importantly, our results show for the first time that rigorous use of computational techniques can lead to discovery of unique GAG sequences that can selectively target GAG-binding protein(s), which may lead to chemical biology or drug discovery tools.


Asunto(s)
Glucuronatos/farmacología , Cofactor II de Heparina/agonistas , Heparitina Sulfato/farmacología , Descubrimiento de Drogas , Glucuronatos/química , Cofactor II de Heparina/metabolismo , Heparitina Sulfato/química , Humanos , Unión Proteica , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
9.
Int J Mol Sci ; 17(5)2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27136540

RESUMEN

Follicular fluid (FF) accumulates in the antrum of the ovarian follicle and provides the microenvironment for oocyte development. FF plays an important role in follicle growth and oocyte maturation. The FF provides a unique window to investigate the processes occurring during buffalo follicular development. The observed low quality of buffalo oocytes may arise from the poor follicular microenvironment. Investigating proteins found in buffalo FF (BFF) should provide insight into follicular development processes and provide further understanding of intra-follicular maturation and oocytes quality. Here, a proteomic-based approach was used to analyze the proteome of BFF. SDS-PAGE separation combined with mass spectrometry was used to generate the proteomic dataset. In total, 363 proteins were identified and classified by Gene Ontology terms. The proteins were assigned to 153 pathways, including signaling pathways. To evaluate difference in proteins expressed between BFF with different follicle size (small, <4 mm; and large, >8 mm), a quantitative proteomic analysis based on multi-dimensional liquid chromatography pre-fractionation tandem Orbitrap mass spectrometry identification was performed. Eleven differentially expressed proteins (six downregulated and five upregulated in large BFF) were identified and assigned to a variety of functional processes, including serine protease inhibition, oxidation protection and the complement cascade system. Three differentially expressed proteins, Vimentin, Peroxiredoxin-1 and SERPIND1, were verified by Western blotting, consistent with the quantitative proteomics results. Our datasets offers new information about proteins present in BFF and should facilitate the development of new biomarkers. These differentially expressed proteins illuminate the size-dependent protein changes in follicle microenvironment.


Asunto(s)
Búfalos/metabolismo , Folículo Ovárico/metabolismo , Proteoma/análisis , Proteómica , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Estradiol/análisis , Femenino , Líquido Folicular/metabolismo , Cofactor II de Heparina/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Péptidos/análisis , Peroxirredoxinas/metabolismo , Progesterona/análisis , Espectrometría de Masas en Tándem , Vimentina/metabolismo
10.
Biochem Biophys Res Commun ; 457(4): 585-8, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25600805

RESUMEN

Heparan sulfate normally binds to heparin cofactor II and modulates the coagulation pathway by inhibiting thrombin. However, when human heparin cofactor II was incubated with heparan sulfate, heparin cofactor II became degraded. Other glycosaminoglycans were tested, including hyaluronic acid, chondroitin sulfates, dermatan sulfate, and heparin, but only dextran sulfate also degraded heparin cofactor II. Pretreatment of heparan sulfate with heparinase reduced its heparin cofactor II-degrading activity. Heparan sulfate and dextran sulfate diminished the thrombin inhibitory activity of heparin cofactor II. Other serpins, including antithrombin III and pigment epithelium-derived factor, were also degraded by heparan sulfate. This is the first evidence of acidic polysaccharides exhibiting protein-degrading activity without the aid of other proteins.


Asunto(s)
Antitrombinas/metabolismo , Sulfato de Dextran/metabolismo , Cofactor II de Heparina/metabolismo , Heparitina Sulfato/metabolismo , Proteolisis , Animales , Antitrombina III/metabolismo , Antitrombinas/farmacología , Bovinos , Flavobacterium/enzimología , Cofactor II de Heparina/farmacología , Liasa de Heparina/metabolismo , Humanos , Indicadores y Reactivos/metabolismo
11.
J Immunol ; 190(12): 6303-10, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23656734

RESUMEN

The abundant serine proteinase inhibitor heparin cofactor II (HCII) has been proposed to inhibit extravascular thrombin. However, the exact physiological role of this plasma protein remains enigmatic. In this study, we demonstrate a previously unknown role for HCII in host defense. Proteolytic cleavage of the molecule induced a conformational change, thereby inducing endotoxin-binding and antimicrobial properties. Analyses employing representative peptide epitopes mapped these effects to helices A and D. Mice deficient in HCII showed increased susceptibility to invasive infection by Pseudomonas aeruginosa, along with a significantly increased cytokine response. Correspondingly, decreased levels of HCII were observed in wild-type animals challenged with bacteria or endotoxin. In humans, proteolytically cleaved HCII forms were detected during wounding and in association with bacteria. Thus, the protease-induced uncovering of cryptic epitopes in HCII, which transforms the molecule into a host defense factor, represents a previously unknown regulatory mechanism in HCII biology and innate immunity.


Asunto(s)
Cofactor II de Heparina/inmunología , Cofactor II de Heparina/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/metabolismo , Western Blotting , Cromatografía Líquida de Alta Presión , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis
12.
Mar Drugs ; 13(4): 2063-84, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25871288

RESUMEN

Sulfated fucans, the complex polysaccharides, exhibit various biological activities. Herein, we purified two fucans from the sea cucumbers Holothuria edulis and Ludwigothurea grisea. Their structures were verified by means of HPGPC, FT-IR, GC-MS and NMR. As a result, a novel structural motif for this type of polymers is reported. The fucans have a unique structure composed of a central core of regular (1→2) and (1→3)-linked tetrasaccharide repeating units. Approximately 50% of the units from L. grisea (100% for H. edulis fucan) contain sides of oligosaccharides formed by nonsulfated fucose units linked to the O-4 position of the central core. Anticoagulant activity assays indicate that the sea cucumber fucans strongly inhibit human blood clotting through the intrinsic pathways of the coagulation cascade. Moreover, the mechanism of anticoagulant action of the fucans is selective inhibition of thrombin activity by heparin cofactor II. The distinctive tetrasaccharide repeating units contribute to the anticoagulant action. Additionally, unlike the fucans from marine alga, although the sea cucumber fucans have great molecular weights and affluent sulfates, they do not induce platelet aggregation. Overall, our results may be helpful in understanding the structure-function relationships of the well-defined polysaccharides from invertebrate as new types of safer anticoagulants.


Asunto(s)
Anticoagulantes/aislamiento & purificación , Descubrimiento de Drogas , Polisacáridos/aislamiento & purificación , Pepinos de Mar/química , Animales , Anticoagulantes/química , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Brasil , Secuencia de Carbohidratos , Fenómenos Químicos , China , Cofactor II de Heparina/antagonistas & inhibidores , Cofactor II de Heparina/metabolismo , Holothuria/química , Humanos , Cinética , Peso Molecular , Polisacáridos/química , Polisacáridos/farmacología , Pepinos de Mar/crecimiento & desarrollo , Especificidad de la Especie , Relación Estructura-Actividad
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 40(2): 177-81, 2015 Feb.
Artículo en Zh | MEDLINE | ID: mdl-25769326

RESUMEN

OBJECTIVE: To explore the relationship between activity of plasma heparin cofactor II (HC II) and the incidence of in-stent restenosis aft er the intervention of arteriosclerosis obliterans in lower extremity. METHODS: A total of 62 patients with arteriosclerosis obliterans in lower extremity underwent femoropopliteal stent implantation. They were divided into 2 groups: A high HC II activity group (≥100%, n=40) and a low HC II activity group (<100%, n=22). All patients filled in follow up tables and conducted body examination. Possible risk factors resulting in restenosis were collected. Patients were followed up for 6 months after femoropopliteal stent implantation. RESULTS: Baseline clinical characteristics were not significantly different between the 2 groups. The degree and incidence of angiographic restenosis at the end of the 6th month after the implantation in the high HC II activity group were all significantly lower than those in the low HC II activity group (P<0.05). Multivariate analysis demonstrated that high plasma HC II activity was an independent factor in reducing the incidence of angiographic restenosis (OR=0.982, P=0.048, 95%CI, 0.966, 0.998). CONCLUSION: High plasma HC II activity is an independent factor in reducing the degree of in-stent restenosis. The lower the plasma HC II activity, the severer the degree of in-stent restenosis.


Asunto(s)
Arteriosclerosis Obliterante/cirugía , Cofactor II de Heparina/metabolismo , Stents , Constricción Patológica , Humanos , Incidencia , Extremidad Inferior , Factores de Riesgo
14.
J Biol Chem ; 287(41): 34256-63, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22904320

RESUMEN

We previously clarified that heparin cofactor II (HCII), a serine proteinase inhibitor, exerts various protective actions on cardiovascular diseases in both experimental and clinical studies. In the present study, we aimed to clarify whether HCII participates in the regulation of angiogenesis. Male heterozygous HCII-deficient (HCII(+/-)) mice and male littermate wild-type (HCII(+/+)) mice at the age of 12-16 weeks were subjected to unilateral hindlimb ligation surgery. Laser speckle blood flow analysis showed that blood flow recovery in response to hindlimb ischemia was delayed in HCII(+/-) mice compared with that in HCII(+/+) mice. Capillary number, arteriole number, and endothelial nitric-oxide synthase (eNOS), AMP-activated protein kinase (AMPK), and liver kinase B1 (LKB1) phosphorylation in ischemic muscles were decreased in HCII(+/-) mice. Human purified HCII (h-HCII) administration almost restored blood flow recovery, capillary density, and arteriole number as well as phosphorylation levels of eNOS, AMPK, and LKB1 in ischemic muscles of HCII(+/-) mice. Although treatment with h-HCII increased phosphorylation levels of eNOS, AMPK, and LKB1 in human aortic endothelial cells (HAECs), the h-HCII-induced eNOS phosphorylation was abolished by compound C, an AMPK inhibitor, and by AMPK siRNA. In a similar fashion, tube formation, proliferation, and migration of HAECs were also promoted by h-HCII treatment and were abrogated by pretreatment with compound C. HCII potentiates the activation of vascular endothelial cells and the promotion of angiogenesis in response to hindlimb ischemia via an AMPK-eNOS signaling pathway. These findings suggest that HCII is a novel therapeutic target for treatment of patients with peripheral circulation insufficiency.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cofactor II de Heparina/metabolismo , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transducción de Señal/fisiología , Animales , Aorta/citología , Aorta/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Cofactor II de Heparina/farmacología , Humanos , Masculino , Ratones , Ratones Mutantes , Neovascularización Fisiológica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
BMC Biochem ; 14: 6, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23496873

RESUMEN

BACKGROUND: Heparin cofactor II (HCII) is a circulating protease inhibitor, one which contains an N-terminal acidic extension (HCII 1-75) unique within the serpin superfamily. Deletion of HCII 1-75 greatly reduces the ability of glycosaminoglycans (GAGs) to accelerate the inhibition of thrombin, and abrogates HCII binding to thrombin exosite 1. While a minor portion of HCII 1-75 can be visualized in a crystallized HCII-thrombin S195A complex, the role of the rest of the extension is not well understood and the affinity of the HCII 1-75 interaction has not been quantitatively characterized. To address these issues, we expressed HCII 1-75 as a small, N-terminally hexahistidine-tagged polypeptide in E. coli. RESULTS: Immobilized purified HCII 1-75 bound active α-thrombin and active-site inhibited FPR-ck- or S195A-thrombin, but not exosite-1-disrupted γT-thrombin, in microtiter plate assays. Biotinylated HCII 1-75 immobilized on streptavidin chips bound α-thrombin and FPR-ck-thrombin with similar KD values of 330-340 nM. HCII 1-75 competed thrombin binding to chip-immobilized HCII 1-75 more effectively than HCII 54-75 but less effectively than the C-terminal dodecapeptide of hirudin (mean Ki values of 2.6, 8.5, and 0.29 µM, respectively). This superiority over HCII 54-75 was also demonstrated in plasma clotting assays and in competing the heparin-catalysed inhibition of thrombin by plasma-derived HCII; HCII 1-53 had no effect in either assay. Molecular modelling of HCII 1-75 correctly predicted those portions of the acidic extension that had been previously visualized in crystal structures, and suggested that an α-helix found between residues 26 and 36 stabilizes one found between residues 61-67. The latter region has been previously shown by deletion mutagenesis and crystallography to play a crucial role in the binding of HCII to thrombin exosite 1. CONCLUSIONS: Assuming that the KD value for HCII 1-75 of 330-340 nM faithfully predicts that of this region in intact HCII, and that 1-75 binding to exosite 1 is GAG-dependent, our results support a model in which thrombin first binds to GAGs, followed by HCII addition to the ternary complex and release of HCII 1-75 for exosite 1 binding and serpin mechanism inhibition. They further suggest that, in isolated or transferred form, the entire HCII 1-75 region is required to ensure maximal binding of thrombin exosite 1.


Asunto(s)
Cofactor II de Heparina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Escherichia coli/metabolismo , Cofactor II de Heparina/química , Cofactor II de Heparina/genética , Hirudinas/síntesis química , Hirudinas/química , Hirudinas/metabolismo , Histidina/genética , Histidina/metabolismo , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Cinética , Ratones , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Oligopéptidos/genética , Oligopéptidos/metabolismo , Péptidos/síntesis química , Péptidos/química , Unión Proteica , Estructura Terciaria de Proteína , Conejos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Alineación de Secuencia , Serpinas/química , Serpinas/metabolismo , Trombina/química , Trombina/metabolismo
16.
J Biomol Struct Dyn ; 41(9): 3717-3727, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35343865

RESUMEN

Thromboembolic diseases are a major cause of mortality in human and the currently available anticoagulants are associated with various drawbacks, therefore the search for anticoagulants that have better safety profile is highly desirable. Compounds that are part of the dietary routine can be modified to possibly increase their anticoagulant potential. We show mannose 2,3,4,5,6-O-pentasulfate (MPS) as a synthetically modified form of mannose that has appreciable anticoagulation properties. An in silico study identified that mannose in sulfated form can bind effectively to the heparin-binding site of antithrombin (ATIII) and heparin cofactor II (HCII). Mannose was sulfated using a simple sulfation strategy-involving triethylamine-sulfur trioxide adduct. HCII and ATIII were purified from human plasma and the binding analysis using fluorometer and isothermal calorimetry showed that MPS binds at a unique site. A thrombin inhibition analysis using the chromogenic substrate showed that MPS partially enhances the activity of HCII. Further an assessment of in vitro blood coagulation assays using human plasma showed that the activated partial thromboplastin time (APTT) and prothrombin time (PT) were prolonged in the presence of MPS. A molecular dynamics simulation analysis of the HCII-MPS complex showed fluctuations in a N-terminal loop and the cofactor binding site of HCII. The results indicate that MPS is a promising lead due to its effect on the in vitro coagulation rate.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Cofactor II de Heparina , Manosa , Humanos , Cofactor II de Heparina/química , Cofactor II de Heparina/metabolismo , Manosa/farmacología , Coagulación Sanguínea , Anticoagulantes/farmacología , Anticoagulantes/química , Heparina/farmacología , Antitrombina III/farmacología , Antitrombina III/fisiología , Antitrombinas/farmacología , Trombina/química
17.
Anal Biochem ; 421(2): 489-98, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22206940

RESUMEN

Thrombin inactivation by heparin cofactor II (HCII) is accelerated by ternary complex formation with heparin. The novel active-site-labeled thrombins, [4'F]FPR-T and [6F]FFR-T, and the exosite I probe, Hir-(54-65)(SO3⁻), characterized thrombin exosite I and II interactions with HCII and heparin in the complex. HCII binding to exosite I of heparin-bound [4'F]FPR-T caused a saturable fluorescence increase, absent with antithrombin. Heparin binding to exosite II and a second weaker site caused fluorescence quenching of [6F]-FFR-T, attenuated by simultaneous Hir-(54-65)(SO3⁻) binding. Stopped-flow analysis demonstrated ordered assembly of HCII and the [6F]FFR-T·heparin complex, in agreement with tighter heparin binding to thrombin than to HCII. Saturating HCII dependences and bell-shaped heparin dependences of the fluorescence change reported ternary complex formation, consistent with a template mechanism in which the thrombin·heparin complex binds HCII and allowing for interaction of thrombin·(heparin)2 complexes with HCII. Hir-(54-65)(SO3⁻) displacement in reactions with FPR-blocked and active thrombin indicated a concerted action of the active site and exosite I during ternary complex formation. These studies demonstrate that binding of HCII to the thrombin·heparin complex is dramatically enhanced compared with heparin binding alone and that exosite I is still available for ligand or HCII binding when both heparin binding sites on thrombin are saturated.


Asunto(s)
Colorantes Fluorescentes/química , Cofactor II de Heparina/metabolismo , Heparina/metabolismo , Trombina/metabolismo , Cinética , Unión Proteica
18.
J Inherit Metab Dis ; 35(2): 355-62, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21732093

RESUMEN

Monitoring of therapeutic response in mucopolysaccharidosis (MPS) patients is problematic as most biomarkers are specific for either disease complications or specific organ system involvement. Recent studies have indicated that serum heparin-cofactor II-thrombin complex (HCII-T) may serve as an important biomarker in the group of MPSs where dermatan sulphate is stored. This complex forms when blood coagulates in the presence of glycosaminoglycans (GAGs) where the ultimate amount of HCII-T that forms reflects the concentration of circulating GAGs. We have studied serum HCII-T levels in 9 MPS I and 11 MPS II treated patients and have compared values to studies of urinary GAGs. In severe MPS I patients treated with either transplantation or enzyme replacement therapy (ERT), serum HCII-T levels never reach the range of normal despite normalization of uGAGs in some patients. Some attenuated MPS I patients have normalization of HCII-T but require a protracted exposure time relative to the drop in urinary GAGs. Treated MPS II patients show a clear correlation of serum HCII-T levels with the presence of antibodies to Idursulfase, with antibody positive patients showing an early drop in HCII-T levels with eventual increases in levels often to levels above those seen at baseline. This is contrasted by a robust and persistent drop in uGAGs. Antibody negative MPS II patients show a drop in HCII-T levels on treatment but levels never normalize despite normalization of uGAGs. This study highlights the utility and biologic relevance of serum HCII-T levels in monitoring therapy in these disorders.


Asunto(s)
Cofactor II de Heparina/metabolismo , Mucopolisacaridosis II/sangre , Mucopolisacaridosis I/sangre , Trombina/metabolismo , Adolescente , Adulto , Biomarcadores/sangre , Niño , Preescolar , Glicosaminoglicanos/sangre , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/orina , Humanos , Iduronato Sulfatasa/metabolismo , Lactante , Estudios Longitudinales , Mucopolisacaridosis I/orina , Mucopolisacaridosis II/orina
19.
J Biol Chem ; 285(11): 8278-89, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20053992

RESUMEN

Inactivation of thrombin (T) by the serpins heparin cofactor II (HCII) and antithrombin (AT) is accelerated by a heparin template between the serpin and thrombin exosite II. Unlike AT, HCII also uses an allosteric interaction of its NH(2)-terminal segment with exosite I. Sucrose octasulfate (SOS) accelerated thrombin inactivation by HCII but not AT by 2000-fold. SOS bound to two sites on thrombin, with dissociation constants (K(D)) of 10 +/- 4 microm and 400 +/- 300 microm that were not kinetically resolvable, as evidenced by single hyperbolic SOS concentration dependences of the inactivation rate (k(obs)). SOS bound HCII with K(D) 1.45 +/- 0.30 mm, and this binding was tightened in the T.SOS.HCII complex, characterized by K(complex) of approximately 0.20 microm. Inactivation data were incompatible with a model solely depending on HCII.SOS but fit an equilibrium linkage model employing T.SOS binding in the pathway to higher order complex formation. Hirudin-(54-65)(SO(3)(-)) caused a hyperbolic decrease of the inactivation rates, suggesting partial competitive binding of hirudin-(54-65)(SO(3)(-)) and HCII to exosite I. Meizothrombin(des-fragment 1), binding SOS with K(D) = 1600 +/- 300 microm, and thrombin were inactivated at comparable rates, and an exosite II aptamer had no effect on the inactivation, suggesting limited exosite II involvement. SOS accelerated inactivation of meizothrombin 1000-fold, reflecting the contribution of direct exosite I interaction with HCII. Thrombin generation in plasma was suppressed by SOS, both in HCII-dependent and -independent processes. The ex vivo HCII-dependent process may utilize the proposed model and suggests a potential for oversulfated disaccharides in controlling HCII-regulated thrombin generation.


Asunto(s)
Anticoagulantes/metabolismo , Coagulación Sanguínea/fisiología , Cofactor II de Heparina/metabolismo , Sacarosa/análogos & derivados , Trombina/metabolismo , Regulación Alostérica , Anticoagulantes/química , Anticoagulantes/farmacología , Unión Competitiva/efectos de los fármacos , Unión Competitiva/fisiología , Coagulación Sanguínea/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Precursores Enzimáticos/metabolismo , Precursores Enzimáticos/farmacología , Cofactor II de Heparina/química , Hirudinas/metabolismo , Hirudinas/farmacología , Humanos , Cinética , Modelos Químicos , Plasma , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Protrombina/metabolismo , Espectrometría de Fluorescencia , Sacarosa/química , Sacarosa/metabolismo , Sacarosa/farmacología , Trombina/química , Trombina/farmacología
20.
J Biol Chem ; 285(10): 7312-23, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20053999

RESUMEN

Heparin-like glycans with diverse disaccharide composition and high anticoagulant activity have been described in several families of marine mollusks. The present work focused on the structural characterization of a new heparan sulfate (HS)-like polymer isolated from the mollusk Nodipecten nodosus (Linnaeus, 1758) and on its anticoagulant and antithrombotic properties. Total glycans were extracted from the mollusk and fractionated by ethanol precipitation. The main component (>90%) was identified as HS-like glycosaminoglycan, representing approximately 4.6 mg g(-1) of dry tissue. The mollusk HS resists degradation with heparinase I but is cleaved by nitrous acid. Analysis of the mollusk glycan by one-dimensional (1)H, two-dimensional correlated spectroscopy, and heteronuclear single quantum coherence nuclear magnetic resonance revealed characteristic signals of glucuronic acid and glucosamine residues. Signals corresponding to anomeric protons of nonsulfated, 3- or 2-sulfated glucuronic acid as well as N-sulfated and/or 6-sulfated glucosamine were also observed. The mollusk HS has an anticoagulant activity of 36 IU mg(-1), 5-fold lower than porcine heparin (180 IU mg(-1)), as measured by the activated partial thromboplastin time assay. It also inhibits factor Xa (IC(50) = 0.835 microg ml(-1)) and thrombin (IC(50) = 9.3 microg ml(-1)) in the presence of antithrombin. In vivo assays demonstrated that at the dose of 1 mg kg(-1), the mollusk HS inhibited thrombus growth in photochemically injured arteries. No bleeding effect, factor XIIa-mediated kallikrein activity, or toxic effect on fibroblast cells was induced by the invertebrate HS at the antithrombotic dose.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/metabolismo , Arterias , Trombosis de las Arterias Carótidas/prevención & control , Endotelio Vascular , Matriz Extracelular/química , Proteoglicanos de Heparán Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/uso terapéutico , Animales , Anticoagulantes/aislamiento & purificación , Anticoagulantes/uso terapéutico , Antitrombinas/metabolismo , Arterias/efectos de los fármacos , Arterias/patología , Arterias/efectos de la radiación , Bivalvos/metabolismo , Conformación de Carbohidratos , Línea Celular , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/efectos de la radiación , Matriz Extracelular/metabolismo , Factor Xa/metabolismo , Inhibidores del Factor Xa , Femenino , Proteoglicanos de Heparán Sulfato/química , Proteoglicanos de Heparán Sulfato/aislamiento & purificación , Heparina/metabolismo , Heparina/uso terapéutico , Cofactor II de Heparina/metabolismo , Liasa de Heparina/metabolismo , Humanos , Masculino , Ácido Nitroso/metabolismo , Ratas , Análisis Espectral/métodos , Porcinos , Trombina/antagonistas & inhibidores , Trombina/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda