Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 533
Filtrar
1.
Cells ; 12(6)2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36980195

RESUMO

INTRODUCTION: Serum lipoproteins, with the exception of high-density lipoprotein cholesterol (HDL-C), are increased in polycystic ovary syndrome (PCOS) and their levels may reflect the associated obesity and insulin resistance, but the nature of this association is not fully explained. Therefore, proteomic analysis of key proteins in lipoprotein metabolism was performed. METHODS: In this cohort study, plasma was collected from 234 women (137 with PCOS and 97 controls without PCOS). Somalogic proteomic analysis was undertaken for the following 19 proteins involved in lipoprotein, and particularly HDL, metabolism: alpha-1-antichymotrypsin; alpha-1-antitrypsin; apolipoproteins A-1, B, D, E, E2, E3, E4, L1, and M; clusterin; complement C3; hemopexin; heparin cofactor II; kininogen-1; serum amyloid A-1; amyloid beta A-4; and paraoxonase-1. RESULTS: The levels of apolipoprotein E were higher in PCOS (p = 0.012). However, the other isoforms of ApoE, ApoE2, E3, and E4, did not differ when compared with controls. ApoM was lower in PCOS (p = 0.000002). Complement C3 was higher in PCOS (p = 0.037), as was heparin cofactor II (HCFII) (p = 0.0004). The levels of the other proteins associated with lipoprotein metabolism did not differ between PCOS and controls. CONCLUSIONS: These data contribute to the concern of the deleterious dyslipidemia found in PCOS, with the novel combination reported here of higher levels of ApoE, C3 and HCFII together with lower ApoM. The dysregulation of these proteins could circumvent the protective effect of HDL-C and contribute to a more atherogenic profile that may increase cardiovascular risk.


Assuntos
Síndrome do Ovário Policístico , Feminino , Humanos , Complemento C3 , Cofator II da Heparina , Estudos de Coortes , Peptídeos beta-Amiloides , Proteômica , HDL-Colesterol , Apolipoproteínas E
3.
J Atheroscler Thromb ; 30(8): 871-883, 2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-36244745

RESUMO

AIMS: Thrombin exerts various pathophysiological functions by activating protease-activated receptors (PARs), and thrombin-induced activation of PARs promotes the development of non-alcoholic fatty liver disease (NAFLD). Since heparin cofactor II (HCII) specifically inactivates thrombin action, we hypothesized that plasma HCII activity correlates with the severity of NAFLD. METHODS: A cross-sectional study was conducted. Plasma HCII activity and noninvasive clinical markers of hepatic fibrosis including fibrosis-4 (FIB-4) index, NAFLD fibrosis score (NFS) and aspartate aminotransferase-to-platelet ratio index (APRI) were determined in 305 Japanese patients with type 2 diabetes mellitus (T2DM). The relationships between plasma HCII activity and the clinical markers were statistically evaluated. RESULTS: Multiple regression analysis including confounding factors showed that plasma HCII activity independently contributed to decreases in FIB-4 index (p<0.001), NFS (p<0.001) and APRI (p=0.004). In addition, logistic regression analysis for the prevalence of advanced hepatic fibrosis defined by the cutoff points of the clinical scores showed that plasma HCII activity was the sole and common negative factor for prevalence of advanced hepatic fibrosis (FIB-4 index: p=0.002, NFS: p=0.026 and APRI: p=0.012). CONCLUSIONS: Plasma HCII activity was inversely associated with clinical hepatic fibrosis indices including FIB-4 index, NFS and APRI and with the prevalence of advanced hepatic fibrosis in patients with T2DM. The results suggest that HCII can serve as a novel biomarker for assessment of hepatic fibrosis of NAFLD in patients with T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Hepatopatia Gordurosa não Alcoólica , Humanos , Diabetes Mellitus Tipo 2/complicações , Cofator II da Heparina , Estudos Transversais , Trombina , Cirrose Hepática/diagnóstico , Cirrose Hepática/complicações , Biomarcadores , Índice de Gravidade de Doença
4.
J Biomol Struct Dyn ; 41(9): 3717-3727, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35343865

RESUMO

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.


Assuntos
Cofator II da Heparina , Manose , Humanos , Cofator II da Heparina/química , Cofator II da Heparina/metabolismo , Manose/farmacologia , Coagulação Sanguínea , Anticoagulantes/farmacologia , Anticoagulantes/química , Heparina/farmacologia , Antitrombina III/farmacologia , Antitrombina III/fisiologia , Antitrombinas/farmacologia , Trombina/química
5.
Front Endocrinol (Lausanne) ; 12: 658304, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248840

RESUMO

Objective: Detailed proteomic analysis in a cohort of patients with differing severity of COVID-19 disease identified biomarkers within the complement and coagulation cascades as biomarkers for disease severity has been reported; however, it is unclear if these proteins differ sufficiently from other conditions to be considered as biomarkers. Methods: A prospective, parallel study in T2D (n = 23) and controls (n = 23). A hyperinsulinemic clamp was performed and normoglycemia induced in T2D [4.5 ± 0.07 mmol/L (81 ± 1.2 mg/dl)] for 1-h, following which blood glucose was decreased to ≤2.0 mmol/L (36 mg/dl). Proteomic analysis for the complement and coagulation cascades were measured using Slow Off-rate Modified Aptamer (SOMA)-scan. Results: Thirty-four proteins were measured. At baseline, 4 of 18 were found to differ in T2D versus controls for platelet degranulation [Neutrophil-activating peptide-2 (p = 0.014), Thrombospondin-1 (p = 0.012), Platelet factor-4 (p = 0.007), and Kininogen-1 (p = 0.05)], whilst 3 of 16 proteins differed for complement and coagulation cascades [Coagulation factor IX (p < 0.05), Kininogen-1 (p = 0.05), and Heparin cofactor-2 (p = 0.007)]; STRING analysis demonstrated the close relationship of these proteins to one another. Induced euglycemia in T2D showed no protein changes versus baseline. At hypoglycemia, however, four proteins changed in controls from baseline [Thrombospondin-1 (p < 0.014), platelet factor-4 (p < 0.01), Platelet basic protein (p < 0.008), and Vitamin K-dependent protein-C (p < 0.00003)], and one protein changed in T2D [Vitamin K-dependent protein-C, (p < 0.0002)]. Conclusion: Seven of 34 proteins suggested to be biomarkers of COVID-19 severity within the platelet degranulation and complement and coagulation cascades differed in T2D versus controls, with further changes occurring at hypoglycemia, suggesting that validation of these biomarkers is critical. It is unclear if these protein changes in T2D may predict worse COVID-19 disease for these patients. Clinical Trial Registration: https://clinicaltrials.gov/, identifier NCT03102801.


Assuntos
Fatores de Coagulação Sanguínea/metabolismo , COVID-19/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hipoglicemia/metabolismo , Idoso , Biomarcadores/metabolismo , Coagulação Sanguínea , Estudos de Casos e Controles , Ativação do Complemento , Fator IX/metabolismo , Feminino , Técnica Clamp de Glucose , Cofator II da Heparina/metabolismo , Humanos , Cininogênios/metabolismo , Masculino , Pessoa de Meia-Idade , Peptídeos/metabolismo , Ativação Plaquetária , Fator Plaquetário 4/metabolismo , Estudos Prospectivos , Proteína C/metabolismo , Proteômica , SARS-CoV-2 , Índice de Gravidade de Doença , Trombospondina 1/metabolismo , beta-Tromboglobulina/metabolismo
6.
Environ Mol Mutagen ; 62(7): 409-421, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34331478

RESUMO

Polycyclic aromatic hydrocarbons (PAHs) are considered as an external factor that induces atherosclerotic cardiovascular disease. Although miR-155 is known to be involved in cardiovascular disease, whether it is involved in PAH-induced arteriosclerosis remains unclear. We evaluated the effects of PAHs on vascularization, permeability, and miR-155 expression in HUVECs. We found that PAHs-induced sclerosis of HUVECs was characterized by increasing permeability, decreasing proliferation, and vascular lumen number. The expression of miR-155 was upregulated by PAHs treatment, and transfection with miR-155 inhibitor could reverse above effect of PAHs-induced sclerosis. Meanwhile, transcriptome sequencing revealed that 63 genes were downregulated in the group of PAHs treatment alone, and were then upregulated in the miR-155 inhibitor group. These genes were mainly involved in complement and coagulation cascades, cytokine-cytokine receptor interaction, TNF signaling pathway, and NF-kappa B signaling pathway. Among these 63 genes, SERPIND1 was directly targeted and regulated by miR-155. Further in vivo experiments in ApoE-/- mice confirmed that PAH accelerates the development of arteriosclerosis by promoting the expression of miR-155 to downregulate the SERPIND1. Therefore, PAH exaggerates atherosclerosis by activating miR-155-dependent endothelial injury. This study provides a fundamental insight on the miR-155 mechanism for PAHs enhancing atherosclerosis and miR-155 potentially serving as a novel drug target.


Assuntos
Aterosclerose/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Cofator II da Heparina/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , MicroRNAs/genética , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Animais , Aterosclerose/induzido quimicamente , Aterosclerose/genética , Aterosclerose/metabolismo , Cofator II da Heparina/genética , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout para ApoE
7.
J Diabetes Investig ; 12(12): 2172-2182, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34043882

RESUMO

AIMS/INTRODUCTION: Thrombin exerts various pathophysiological functions by activating protease-activated receptors (PARs). Recent data have shown that PARs influence the development of glomerular diseases including diabetic kidney disease (DKD) by regulating inflammation. Heparin cofactor II (HCII) specifically inactivates thrombin; thus, we hypothesized that low plasma HCII activity correlates with DKD development, as represented by albuminuria. MATERIALS AND METHODS: Plasma HCII activity and spot urine biomarkers, including albumin and liver-type fatty acid-binding protein (L-FABP), were determined as the urine albumin-to-creatinine ratio (uACR) and the urine L-FABP-to-creatinine ratio (uL-FABPCR) in 310 Japanese patients with diabetes mellitus (176 males and 134 females). The relationships between plasma HCII activities and those DKD urine biomarkers were statistically evaluated. In addition, the relationship between plasma HCII activities and annual uACR changes was statistically evaluated for 201/310 patients (115 males and 86 females). RESULTS: The mean plasma HCII activity of all participants was 93.8 ± 17.7%. Multivariate-regression analysis including confounding factors showed that plasma HCII activity independently contributed to the suppression of the uACR and log-transformed uACR values (P = 0.036 and P = 0.006, respectively) but not uL-FABPCR (P = 0.541). In addition, plasma HCII activity significantly and inversely correlated with annual uACR and log-transformed uACR increments after adjusting for confounding factors (P = 0.001 and P = 0.014, respectively). CONCLUSIONS: The plasma HCII activity was inversely and specifically associated with glomerular injury in patients with diabetes. The results suggest that HCII can serve as a novel predictive factor for early-stage DKD development, as represented by albuminuria.


Assuntos
Albuminúria/sangue , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/urina , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/urina , Cofator II da Heparina/análise , Adulto , Idoso , Albuminas/metabolismo , Albuminúria/urina , Biomarcadores/sangue , Biomarcadores/urina , Creatinina/urina , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/etiologia , Proteínas de Ligação a Ácido Graxo/urina , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Receptores Ativados por Proteinase/sangue , Análise de Regressão , Trombina/metabolismo
8.
Carbohydr Polym ; 256: 117513, 2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-33483034

RESUMO

Marine sulfated polysaccharides have aroused widespread concern for their various structures and bioactivities. Peroxide depolymerization is a common strategy in analysis of structures and structure-activity relationships of polysaccharides. However, confirming the depolymerization process and exact structures of the degradation products is still a considerable challenge. This study reported the structures of a fucan sulfate (FS) from sea cucumber Stichopus herrmanni and its depolymerized products (dFS) prepared by peroxide degradation. The FS was elucidated with a highly regular structure, {-3)-L-Fuc2S-(α1-}n. Structure analysis of oligosaccharides purified from dFS suggested that peroxide degradation involved in cleavage of glycosidic bonds and oxidative modification of reducing end of sugar residue, while no break in sugar ring was observed. Both FS and series of dFSs exhibited significant anticoagulant activities due to their anti-thrombin effects in presence of heparin cofactor II and their potencies were related to their molecular sizes, dFS with ∼ 20 kDa showed the strongest activity.


Assuntos
Anticoagulantes/química , Coagulação Sanguínea/efeitos dos fármacos , Peróxidos/química , Polissacarídeos/química , Stichopus/química , Animais , Anticoagulantes/isolamento & purificação , Anticoagulantes/farmacologia , Organismos Aquáticos , Testes de Coagulação Sanguínea , Sequência de Carboidratos , Cofator II da Heparina/farmacologia , Humanos , Hidrólise , Peso Molecular , Polissacarídeos/isolamento & purificação , Polissacarídeos/farmacologia , Stichopus/fisiologia , Relação Estrutura-Atividade , Trombina/antagonistas & inibidores , Trombina/metabolismo
9.
IUBMB Life ; 72(10): 2180-2193, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32827448

RESUMO

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.


Assuntos
Cofator II da Heparina/química , Cofator II da Heparina/genética , Cofator II da Heparina/metabolismo , Fígado/metabolismo , Processamento Alternativo , Fator Xa/metabolismo , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Isoformas de Proteínas , Espectrometria de Fluorescência , Trombina/metabolismo , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Ativador de Plasminogênio Tecidual/metabolismo
10.
Int J Biol Macromol ; 164: 87-94, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32663563

RESUMO

A sulfated fucan was extracted, purified, and characterized from Acaudina leucoprocta (a low value sea cucumber) to better understand and utilize this species. The structure of the sulfated fucan was elucidated using chemical and modern spectroscopic analyses including HPGPC, IR, AFM, GC-MS, and NMR, and its bioactivity was investigated. Our results showed that the sulfated fucan was mainly composed of → 3)-α-L-Fucp-(1→ linkage, and that the sulfate groups were substituted at the O-2 and/or O-4 positions of the fucose ring. In detail, the sulfated fucan consisted of Fuc0S (40%), Fuc2S4S (24%), Fuc2S (24%), and Fuc4S (12%). On average, there were seven sulfate groups on every eight fucose residues. Assay for anticoagulant activity indicated that the sulfated fucan displayed intrinsic anticoagulant activity and specific anti-thrombin activity through heparin cofactor II. Our results showed that this bioactive sulfated fucan could enable the high-value utilization of this low-value sea cucumber.


Assuntos
Anticoagulantes/isolamento & purificação , Polissacarídeos/isolamento & purificação , Animais , Anticoagulantes/química , Anticoagulantes/farmacologia , Testes de Coagulação Sanguínea , Configuração de Carboidratos , Cromatografia por Troca Iônica , Cromatografia Gasosa-Espectrometria de Massas , Cofator II da Heparina/farmacologia , Humanos , Microscopia de Força Atômica , Estrutura Molecular , Peso Molecular , Polissacarídeos/química , Polissacarídeos/farmacologia , Pepinos-do-Mar/química , Análise Espectral , Sulfatos/análise
11.
Nat Chem Biol ; 16(4): 379-382, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32198493

RESUMO

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.


Assuntos
Engenharia de Proteínas/métodos , Somatomedinas/química , Tirosina/análogos & derivados , Animais , Códon de Terminação/metabolismo , Escherichia coli/metabolismo , Código Genético , Cofator II da Heparina/metabolismo , Humanos , Mamíferos , Processamento de Proteína Pós-Traducional , Proteínas/química , Tirosina/química , Tirosina-tRNA Ligase/metabolismo
12.
Cytokine ; 126: 154876, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31629109

RESUMO

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.


Assuntos
Biomarcadores/sangue , Menopausa Precoce/sangue , Insuficiência Ovariana Primária/sangue , Adulto , Anticorpos , Proteínas de Transporte/sangue , Regulação para Baixo , Estradiol/sangue , Feminino , Receptores Frizzled/sangue , Glicoproteínas/sangue , Cofator II da Heparina/metabolismo , Humanos , Molécula 3 de Adesão Intercelular/sangue , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Interferon gama/sangue , Interferons/sangue , Interleucina-17/sangue , Interleucinas/sangue , Metaloproteinase 7 da Matriz/sangue , Pessoa de Meia-Idade , Neurturina/sangue , Insuficiência Ovariana Primária/imunologia , Insuficiência Ovariana Primária/patologia , Análise Serial de Proteínas , Receptores de Interleucina-17/sangue , Albumina Sérica Humana , Regulação para Cima
13.
PLoS Genet ; 15(3): e1007948, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30870413

RESUMO

Glial cells regulate multiple aspects of synaptogenesis. In the absence of Schwann cells, a peripheral glial cell, motor neurons initially innervate muscle but then degenerate. Here, using a genetic approach, we show that neural activity-regulated negative factors produced by muscle drive neurodegeneration in Schwann cell-deficient mice. We find that thrombin, the hepatic serine protease central to the hemostatic coagulation cascade, is one such negative factor. Trancriptomic analysis shows that expression of the antithrombins serpin C1 and D1 is significantly reduced in Schwann cell-deficient mice. In the absence of peripheral neuromuscular activity, neurodegeneration is completely blocked, and expression of prothrombin in muscle is markedly reduced. In the absence of muscle-derived prothrombin, neurodegeneration is also markedly reduced. Together, these results suggest that Schwann cells regulate NMJs by opposing the effects of activity-regulated, muscle-derived negative factors and provide the first genetic evidence that thrombin plays a central role outside of the coagulation system.


Assuntos
Antitrombina III/genética , Cofator II da Heparina/genética , Junção Neuromuscular/genética , Protrombina/genética , Sinapses/genética , Animais , Perfilação da Expressão Gênica , Camundongos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Músculo Esquelético/metabolismo , Degeneração Neural/genética , Neuroglia , Junção Neuromuscular/crescimento & desenvolvimento , Células de Schwann/metabolismo , Trombina/genética
14.
Glycobiology ; 29(6): 446-451, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30869126

RESUMO

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.


Assuntos
Dermatan Sulfato/farmacologia , Cofator II da Heparina/metabolismo , Inibidores de Serina Proteinase/farmacologia , Trombina/antagonistas & inibidores , Configuração de Carboidratos , Dermatan Sulfato/síntese química , Dermatan Sulfato/química , Humanos , Modelos Moleculares , Inibidores de Serina Proteinase/síntese química , Inibidores de Serina Proteinase/química , Trombina/metabolismo
15.
Psychiatry Res ; 272: 404-410, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30611956

RESUMO

Globally, depression is one of the most serious debilitating psychiatric mental disorders. In this study, we validated the expression levels of fibrinogen alpha (FGA), fibrinogen beta (FGB), fibrinogen gamma (FGG), Complement factor B (CFB) and serpin family D member 1(SERPIND1) in the acute phase response signaling pathway in plasma samples using enzyme-linked immunosorbent assay (ELISA).Then illuminate the roles of FGA, FGB, FGG, CFB, SERPIND1 in depression using microarray data. Gene expression dataset GSE98793 was downloaded from the Gene Expression Omnibus database. There were 128 whole blood samples included 64 patients with major depressed patients and 64 healthy controls. Differentially expressed genes (DEGs) were identified, and then protein-protein interaction (PPI) network was constructed to screen crucial genes associated with FGA, FGB, FGG, CFB and SERPIND1. Moreover, gene ontology (GO) biological processes analyses was performed. The ELISA data showed that the expression levels of FGA, FGB, FGG, CFB and SERPIND1 were up-regulated in depressed patients. The enriched GO terms were predominantly associated with the biological processes including more genes were inflammation related. The PPI network was found these five genes interacted with 11 genes. FGA, FGB, FGG, CFB and SERPIND1 may be important in the pathogenesis of depression.


Assuntos
Reação de Fase Aguda/sangue , Reação de Fase Aguda/diagnóstico , Fator B do Complemento/metabolismo , Depressão/sangue , Depressão/diagnóstico , Cofator II da Heparina/metabolismo , Reação de Fase Aguda/psicologia , Adulto , Biomarcadores/sangue , Depressão/psicologia , Feminino , Fibrinogênio/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade
16.
Eur J Med Chem ; 154: 133-143, 2018 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-29787913

RESUMO

Fucosylated chondroitin sulfate (FCS), an unusual glycosaminoglycan with fucose side chains, is a promising anticoagulant agent. To assess the effect of its structure on anticoagulant activity, its derivatives with various degrees of fucosylation (DF), molecular weights (Mw) and sulfation patterns were prepared and characterized. Biological tests showed that their APTT (activated partial thromboplastin time) prolonging activity and intrinsic factor Xase complex (factor IXa-VIIIa-Ca2+-PL complex) inhibitory activity were both reduced in FCS derivatives with lower Mw and DF. However, FCSs with DF at least 16% resulted in greater heparin cofactor II (HCII)-dependent thrombin inhibitory activity in response to decreasing DF, and these activities did not depend on Mw (Mw > 5.2 kDa). Solution competition binding assay further suggested that modulating the DF of FCS derivatives might enhance inhibition of thrombin by activating HCII. These findings imply that FCS derivatives with suitable chain length and DF value may be novel anticoagulants by activating HCII.


Assuntos
Anticoagulantes/farmacologia , Sulfatos de Condroitina/farmacologia , Cofator II da Heparina/metabolismo , Trombina/antagonistas & inibidores , Anticoagulantes/química , Anticoagulantes/isolamento & purificação , Sulfatos de Condroitina/química , Sulfatos de Condroitina/isolamento & purificação , Relação Dose-Resposta a Droga , Cofator II da Heparina/química , Humanos , Estrutura Molecular , Relação Estrutura-Atividade , Trombina/metabolismo
17.
Chin J Nat Med ; 16(4): 241-251, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29703324

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of acute lower respiratory tract infections. Qingfei oral liquid (QFOL), a traditional Chinese medicine, is widely used in clinical treatment for RSV-induced pneumonia. The present study was designed to reveal the potential targets and mechanism of action for QFOL by exploring its influence on the host cellular network following RSV infection. We investigated the serum proteomic changes and potential biomarkers in an RSV-infected mouse pneumonia model treated with QFOL. Eighteen BALB/c mice were randomly divided into three groups: RSV pneumonia model group (M), QFOL-treated group (Q) and the control group (C). Serum proteomes were analyzed and compared using a label-free quantitative LC-MS/MS approach. A total of 172 protein groups, 1009 proteins, and 1073 unique peptides were successfully identified. 51 differentially expressed proteins (DEPs) were identified (15 DEPs when M/C and 43 DEPs when Q/M; 7 DEPs in common). Classification and interaction network showed that these proteins participated in various biological processes including immune response, blood coagulation, complement activation, and so forth. Particularly, fibrinopeptide B (FpB) and heparin cofactor II (HCII) were evaluated as important nodes in the interaction network, which was closely involved in coagulation and inflammation. Further, the FpB level was increased in Group M but decreased in Group Q, while the HCII level exhibited the opposite trend. These findings not only indicated FpB and HCII as potential biomarkers and targets of QFOL in the treatment of RSV pneumonia, but also suggested a regulatory role of QFOL in the RSV-induced disturbance of coagulation and inflammation-coagulation interactions.


Assuntos
Biomarcadores/sangue , Medicamentos de Ervas Chinesas/farmacologia , Fibrinopeptídeo B/análise , Cofator II da Heparina/análise , Proteoma/efeitos dos fármacos , Proteômica , Infecções por Vírus Respiratório Sincicial/sangue , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Animais , Cromatografia Líquida , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Fibrinopeptídeo B/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Cofator II da Heparina/genética , Pulmão/patologia , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Espectrometria de Massas em Tandem
18.
PLoS One ; 13(3): e0193482, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29494632

RESUMO

Oversulfated chondroitin sulfate (OSCS) was identified as a contaminant in certain heparin preparations as the cause of adverse reactions in patients. OSCS was found to possess both plasma anticoagulant activity and the ability to activate prekallikrein to kallikrein. Differentially sulfated chondroitin sulfates were prepared by synthetic modification of chondroitin sulfate and were compared to the activity of OSCS purified from contaminated heparin. Whilst chondroitin sulfate was found to have minimal anticoagulant activity, increasing sulfation levels produced an anticoagulant response which we directly show for the first time is mediated through heparin cofactor II. However, the tetra-sulfated preparations did not possess any higher anticoagulant activity than several tri-sulfated variants, and also had lower heparin cofactor II mediated activity. Activation of prekallikrein was concentration dependent for all samples, and broadly increased with the degree of sulfation, though the di-sulfated preparation was able to form more kallikrein than some of the tri-sulfated preparations. The ability of the samples to activate the kinin system, as measured by bradykinin, was observed to be through kallikrein generation. These results show that whilst an increase in sulfation of chondroitin sulfate did cause an increase in anticoagulant activity and activation of the kinin system, there may be subtler structural interactions other than sulfation at play given the different responses observed.


Assuntos
Anticoagulantes/síntese química , Bradicinina/metabolismo , Sulfatos de Condroitina/síntese química , Heparina/química , Calicreínas/metabolismo , Animais , Anticoagulantes/química , Anticoagulantes/farmacologia , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Relação Dose-Resposta a Droga , Contaminação de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Cofator II da Heparina/metabolismo , Humanos , Relação Estrutura-Atividade
20.
J Atheroscler Thromb ; 24(12): 1215-1230, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28502917

RESUMO

AIM: Accelerated thrombin action is associated with insulin resistance. It is known that upon activation by binding to dermatan sulfate proteoglycans, heparin cofactor Ⅱ(HCⅡ) inactivates thrombin in tissues. Because HCⅡ may be involved in glucose metabolism, we investigated the relationship between plasma HCⅡ activity and insulin resistance. METHODS AND RESULTS: In a clinical study, statistical analysis was performed to examine the relationships between plasma HCⅡ activity, glycosylated hemoglobin (HbA1c), fasting plasma glucose (FPG), and homeostasis model assessment-insulin resistance (HOMA-IR) in elderly Japanese individuals with lifestyle-related diseases. Multiple regression analysis showed significant inverse relationships between plasma HCⅡ activity and HbA1c (p=0.014), FPG (p=0.007), and HOMA-IR (p= 0.041) in elderly Japanese subjects. In an animal study, HCⅡ+/+ mice and HCⅡ+/- mice were fed with a normal diet or high-fat diet (HFD) until 25 weeks of age. HFD-fed HCⅡ+/- mice exhibited larger adipocyte size, higher FPG level, hyperinsulinemia, compared to HFD-fed HCⅡ+/+ mice. In addition, HFD-fed HCⅡ+/- mice exhibited augmented expression of monocyte chemoattractant protein-1 and tumor necrosis factor, and impaired phosphorylation of the serine/threonine kinase Akt and AMP-activated protein kinase in adipose tissue compared to HFD-fed HCⅡ+/+ mice. The expression of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase was also enhanced in the hepatic tissues of HFD-fed HCⅡ+/- mice. CONCLUSIONS: The present studies provide evidence to support the idea that HCⅡ plays an important role in the maintenance of glucose homeostasis by regulating insulin sensitivity in both humans and mice. Stimulators of HCⅡ production may serve as novel therapeutic tools for the treatment of type 2 diabetes.


Assuntos
Glicemia/metabolismo , Cofator II da Heparina/fisiologia , Homeostase , Resistência à Insulina , Insulina/sangue , Animais , Biomarcadores/sangue , Dieta Hiperlipídica/efeitos adversos , Feminino , Humanos , Hiperglicemia/sangue , Hiperglicemia/etiologia , Hiperglicemia/patologia , Hiperinsulinismo/sangue , Hiperinsulinismo/etiologia , Hiperinsulinismo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Obesidade/complicações , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA