Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Medicinas Complementares
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
MAbs ; 14(1): 2122957, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36151884

RESUMO

Biotherapeutics are exposed to common transition metal ions such as Cu(II) and Fe(II) during manufacturing processes and storage. IgG1 biotherapeutics are vulnerable to reactive oxygen species (ROS) generated via the metal-catalyzed oxidation reactions. Exposure to these metal ions can lead to potential changes to structure and function, ultimately influencing efficacy, potency, and potential immunogenicity of the molecules. Here, we stress four biotherapeutics of the IgG1 subclass (trastuzumab, trastuzumab emtansine, anti-NaPi2b, and anti-NaPi2b-vc-MMAE) with two common pharmaceutically relevant metal-induced oxidizing systems, Cu(II)/ ascorbic acid and Fe(II)/ H2O2, and evaluated oxidation, size distribution, carbonylation, Fc effector functions, antibody-dependent cellular cytotoxicity (ADCC) activity, cell anti-proliferation and autophaghic flux. Our study demonstrates that the extent of oxidation was metal ion-dependent and site-specific, leading to decreased FcγRIIIa and FcRn receptor binding and subsequently potentially reduced bioactivity, though antigen binding was not affected to a great extent. In general, the monoclonal antibody (mAb) and corresponding antibody-drug conjugate (ADC) showed similar impacts to product quality when exposed to the same metal ion, either Cu(II) or Fe(II). Our study clearly demonstrates that transition metal ion binding to therapeutic IgG1 mAbs and ADCs is not random and that oxidation products show unique structural and functional ramifications. A critical outcome from this study is our highlighting of key process parameters, route of degradation, especially oxidation (metal catalyzed or via ROS), on the CH1 and Fc region of full-length mAbs and ADCs.Abbreviations: DNPH 2,4-dinitrophenylhydrazine; ADC Antibody drug conjugate; ADCC Antibody-dependent cellular cytotoxicity; CDR Complementary determining region; DTT Dithiothreitol; HMWF high molecular weight form; LC-MS Liquid chromatography-mass spectrometry; LMWF low molecular weight forms; MOA Mechanism of action; MCO Metal-catalyzed oxidation; MetO Methionine sulfoxide; mAbs Monoclonal antibodies; MyBPC Myosin binding protein C; ROS Reactive oxygen species; SEC Size exclusion chromatography.


Assuntos
Antineoplásicos Imunológicos , Imunoconjugados , Ado-Trastuzumab Emtansina , Anticorpos Monoclonais/química , Ácido Ascórbico , Catálise , Ditiotreitol , Compostos Ferrosos , Peróxido de Hidrogênio , Imunoglobulina G/química , Miosinas/metabolismo , Oxirredução , Proteína C/metabolismo , Espécies Reativas de Oxigênio , Trastuzumab/metabolismo , Trastuzumab/farmacologia
2.
Biochim Biophys Acta Gen Subj ; 1865(6): 129892, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33722640

RESUMO

We previously demonstrated that heterozygous Gly197 to Arg mutation in PROC is associated with venous thrombosis due to the mutation abrogating both zymogenic and enzymatic activities of protein C and activated protein C (APC). In this study, we investigated the role of Gly197 on the structure and function of protein C by replacing it with Ala, Lys and Glu in separate constructs. Characterization of protein C mutants indicated their activation by thrombin is improved ~5-20-fold with the order of PC-G197K > PC-G197E > PC-G197A > PC-WT. Interestingly, the cofactor function of thrombomodulin (TM) in promoting the activation of zymogens by thrombin followed the reverse order of PC-WT > PC-G197A > PC-G197E > PC-G197K. The thrombin-generation inhibitory profiles of zymogens in a tissue factor-mediated thrombin generation assay using protein C-deficient plasma with or without supplementation with TM followed the same order of zymogen activation in the purified system. Evaluation of anticoagulant activities of APC derivatives by prothrombinase and aPTT assays revealed a normal activity for APC-G197A but dramatically impaired activity for the other two mutants. In the endothelial cell permeability assay, APC-G197A exhibited normal antiinflammatory activity, but the other two mutants were nearly inactive. These results suggest that Gly197 plays a key role in TM cofactor-dependent protein C activation by thrombin. It facilitates the recognition of protein C by thrombin in the presence of TM but impedes it in the absence of the cofactor. In APC, a small residue at this position is required for the proper folding/reactivity of the active-site pocket of the protease, a hypothesis supported by structural modeling.


Assuntos
Anti-Inflamatórios/farmacologia , Anticoagulantes/farmacologia , Glicina/genética , Mutação , Proteína C/química , Proteína C/metabolismo , Fator V/metabolismo , Glicina/química , Glicina/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Proteína C/genética , Conformação Proteica , Relação Estrutura-Atividade , Trombina/metabolismo , Trombomodulina/metabolismo
3.
Medicine (Baltimore) ; 99(12): e19650, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32195974

RESUMO

RATIONALE: Mutations of the NKX2-1 gene are associated with brain-lung-thyroid syndrome, which is characterized by benign hereditary chorea, hypothyroidism, and pulmonary disease with variable presentation. Surfactant protein C (SFTPC) gene mutations result in chronic interstitial lung disease in adults or severe neonatal respiratory distress syndrome. PATIENT CONCERNS: Recurrent hypoxemia was observed shortly after birth in a baby at a gestational age of 40 weeks and birth weight of 3150 g. The need for respiratory support gradually increased. He had hypothyroidism and experienced feeding difficulties and irritability. DIAGNOSIS: Genetic examination of the peripheral blood revealed combined mutations of the NKX2-1 and SFTPC genes. INTERVENTIONS: The patient was administered respiratory support, antibiotics, low-dose dexamethasone, supplementary thyroxine, venous nutrition, and other supportive measures. OUTCOMES: The patient's guardian stopped treatment 3 months after commencement of treatment, due to the seriousness of his condition and the patient died. LESSONS: Combined mutations of NKX2-1 and SFTPC genes are very rare. Thus, idiopathic interstitial pneumonia with hypothyroidism and neurological disorders require special attention.


Assuntos
Atetose/genética , Coreia/genética , Hipotireoidismo Congênito/genética , Proteína C/metabolismo , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Fator Nuclear 1 de Tireoide/genética , Atetose/sangue , Atetose/diagnóstico , Atetose/terapia , Coreia/sangue , Coreia/diagnóstico , Coreia/terapia , Hipotireoidismo Congênito/sangue , Hipotireoidismo Congênito/diagnóstico , Hipotireoidismo Congênito/terapia , Evolução Fatal , Transtornos da Alimentação e da Ingestão de Alimentos/diagnóstico , Transtornos da Alimentação e da Ingestão de Alimentos/etiologia , Humanos , Hipotireoidismo/diagnóstico , Hipotireoidismo/etiologia , Hipóxia/diagnóstico , Hipóxia/etiologia , Recém-Nascido , Cariotipagem , Doenças Pulmonares Intersticiais/diagnóstico , Doenças Pulmonares Intersticiais/etiologia , Masculino , Mutação , Cuidados Paliativos/métodos , Recidiva , Síndrome do Desconforto Respiratório do Recém-Nascido/sangue , Síndrome do Desconforto Respiratório do Recém-Nascido/diagnóstico , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia
4.
Microb Pathog ; 112: 30-37, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28942173

RESUMO

Manganese transport protein C (MntC) of Staphylococcus aureus represents an excellent vaccine-candidate antigen. The important role of CD4+ T cells in effective immunity against S. aureus infection was shown; however, CD4+ T cell-specific epitopes on S. aureus MntC have not been well identified. Here, we used bioinformatics prediction algorithms to evaluate and identify nine candidate epitopes within MntC. Our results showed that peptide M8 emulsified in Freund's adjuvant induced a much higher cell-proliferation rate as compared with controls. Additionally, CD4+ T cells stimulated with peptide M8 secreted significantly higher levels of interferon-γ and interleukin-17A. These results suggested that peptide M8 represented an H-2d (I-E)-restricted Th17-specific epitope.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/isolamento & purificação , Manganês/metabolismo , Proteína C/metabolismo , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Algoritmos , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Citocinas/metabolismo , Mapeamento de Epitopos , Escherichia coli/genética , Feminino , Interferon gama/metabolismo , Interleucina-17/metabolismo , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos BALB C , Proteína C/genética , Proteína C/imunologia , Estrutura Secundária de Proteína , Proteínas Recombinantes/imunologia , Infecções Estafilocócicas/imunologia , Células Th1/imunologia , Células Th17/imunologia
5.
J Immunol ; 199(8): 2930-2936, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28877991

RESUMO

The host injury involved in multiorgan system failure during severe inflammation is mediated, in part, by massive infiltration and sequestration of hyperactive neutrophils in the visceral organ. A recombinant form of human activated protein C (rhAPC) has shown cytoprotective and anti-inflammatory functions in some clinical and animal studies, but the direct mechanism is not fully understood. Recently, we reported that, during endotoxemia and severe polymicrobial peritonitis, integrin VLA-3 (CD49c/CD29) is specifically upregulated on hyperinflammatory neutrophils and that targeting the VLA-3high neutrophil subpopulation improved survival in mice. In this article, we report that rhAPC binds to human neutrophils via integrin VLA-3 (CD49c/CD29) with a higher affinity compared with other Arg-Gly-Asp binding integrins. Similarly, there is preferential binding of activated protein C (PC) to Gr1highCD11bhighVLA-3high cells isolated from the bone marrow of septic mice. Furthermore, specific binding of rhAPC to human neutrophils via VLA-3 was inhibited by an antagonistic peptide (LXY2). In addition, genetically modified mutant activated PC, with a high affinity for VLA-3, shows significantly improved binding to neutrophils compared with wild-type activated PC and significantly reduced neutrophil infiltration into the lungs of septic mice. These data indicate that variants of activated PC have a stronger affinity for integrin VLA-3, which reveals novel therapeutic possibilities.


Assuntos
Inflamação/imunologia , Integrina alfa3beta1/metabolismo , Pulmão/imunologia , Insuficiência de Múltiplos Órgãos/imunologia , Neutrófilos/imunologia , Peritonite/imunologia , Proteína C/metabolismo , Animais , Terapia Biológica , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Ativação de Neutrófilo , Ligação Proteica , Proteína C/genética , Proteínas Recombinantes/genética
6.
J Thromb Haemost ; 15(5): 950-960, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28211163

RESUMO

Essentials Protein S is a cofactor of activated protein C (APC) and tissue factor pathway inhibitor (TFPI). There are no assays to quantify separate APC and TFPI cofactor activities of protein S in plasma. We developed assays to measure the APC- and TFPI-cofactor activities of protein S in plasma. The assays were sensitive to protein S deficiency, and not affected by the Factor V Leiden mutation. SUMMARY: Background Protein S plays an important role in the down-regulation of coagulation as cofactor for activated protein C (APC) and tissue factor pathway inhibitor (TFPI). Aim To develop functional assays to quantify the APC- and TFPI-cofactor activities of protein S in plasma. Methods APC- and TFPI-cofactor activities of protein S in plasma were measured using calibrated automated thrombography in protein S-depleted plasma supplemented with a small amount of sample plasma either in the presence of anti-TFPI antibodies and APC (APC-cofactor activity) or at excess full-length TFPI without APC (TFPI-cofactor activity). Total and free protein S levels in plasma were measured by ELISAs. Results Average APC-cofactor activities of protein S were 113%, 108% and 89% in plasma from normal individuals (n = 15), FV Leiden heterozygotes (n = 14) and FV Leiden homozygotes (n = 7), respectively, whereas the average APC-cofactor activity of protein S in plasma from heterozygous protein S-deficient individuals (n = 21) was significantly lower (55%). Similar trends were observed for the TFPI-cofactor activity of protein S, with averages of 109%, 115% and 124% in plasma from individuals with normal protein S levels and different FV Leiden genotypes, and 64% in plasma from protein S-deficient patients. APC-cofactor activities of protein S correlated significantly with free and total protein S antigen levels, whereas TFPI-cofactor activities correlated less with protein S antigen levels. Conclusion We have developed functional protein S assays that measure both the APC- and TFPI-cofactor activities of protein S in plasma, which are hardly if at all affected by the FV Leiden mutation.


Assuntos
Testes de Coagulação Sanguínea/métodos , Coagulação Sanguínea , Lipoproteínas/sangue , Proteína C/metabolismo , Deficiência de Proteína S/diagnóstico , Proteína S/metabolismo , Trombina/metabolismo , Resistência à Proteína C Ativada/sangue , Resistência à Proteína C Ativada/diagnóstico , Resistência à Proteína C Ativada/genética , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática , Fator V/genética , Humanos , Mutação Puntual , Valor Preditivo dos Testes , Proteína S/genética , Deficiência de Proteína S/sangue , Deficiência de Proteína S/genética
7.
J Tradit Chin Med ; 36(6): 779-83, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-29949711

RESUMO

OBJECTIVE: To investigate the effects of electronically stimulating Tianshu (ST 25) and Dachangshu (BL 25), Quchi (LI 11) and Shangjuxu (ST 37) on the jejunum c-kit protein and c-kit mRNA in rats with functional diarrhea (FD). METHODS: FD models were established through intragastric administration with folium sennae. Experimental rats were then divided into 4 groups: blank group, model group, electroacupuncture group Ⅰ [Tianshu (ST 25) and Dachangshu (BL 25) of both sides] and electroacupuncture group Ⅱ [Quchi (Li 11) and Shangjuxu (ST 37) of both sides], 10 in each. After treatment with electroacupuncture for 10 days, The expressions of jejunum c-kit protein and c-kit mRNA in each group were detected with Western blot and Real-Time quantitative real-time polymerase chain reaction (PCR). RESULTS: The expressions of c-kit protein and c-kit mRNA in the model group increased significantly compared to those in the blank group (P < 0.01); the expressions in electroacupuncture group Ⅰsignificantly decreased compared to those in the model group (P < 0.01). CONCLUSION: Our findings suggest that electronicall stimulating both Tianshu (ST 25) and Dachangshu (BL 25) significantly increased the expressions of jejunum c-kit protein and c-kit mRNA in FD rats, which means the treatment might have better therapeutic effects on FD.


Assuntos
Diarreia/terapia , Eletroacupuntura , Jejuno/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Pontos de Acupuntura , Animais , Diarreia/genética , Diarreia/metabolismo , Humanos , Masculino , Proteína C/genética , Proteína C/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Metab Brain Dis ; 30(1): 57-65, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24810631

RESUMO

Multiple sclerosis (MS) is a neuroinflammatory disease characterized by demyelination and axonal damage of the central nervous system. The pathogenesis of MS has also been linked to vascular inflammation and local activation of the coagulation system, resulting in perivascular fibrin deposition. Treatment of experimental autoimmune encephalomyelitis (EAE), a model of human MS, with antithrombotic and antiinflammatory activated protein C (APC) reduces disease severity. Since recombinant APC (Drotecogin alfa), originally approved for the treatment of severe sepsis, is not available for human MS studies, we tested the hypothesis that pharmacologic activation of endogenous protein C could likewise improve the outcome of EAE. Mice were immunized with murine myelin oligodendrocyte glycoprotein (MOG) peptides and at the onset of EAE symptoms, were treated every other day with either WE thrombin (25 µg/kg; i.v.), a selective recombinant protein C activator thrombin analog, or saline control. Mice were monitored for changes in disease score until euthanized for ex vivo analysis of inflammation. Administration of WE thrombin significantly ameliorated clinical severity of EAE, reduced inflammatory cell infiltration and demyelination, suppressed the activation of macrophages comprising the CD11b + population and reduced accumulation of fibrin (ogen) in the spinal cord. These data suggest that symptomatic MS may respond to a treatment strategy that involves temporal pharmacological enhancement of endogenous APC generation.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Proteína C/agonistas , Trombina/uso terapêutico , Animais , Avaliação Pré-Clínica de Medicamentos , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Ativação Enzimática , Fibrina/análise , Fibrinogênio/análise , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Ativação de Macrófagos , Masculino , Camundongos , Esclerose Múltipla , Glicoproteína Mielina-Oligodendrócito/imunologia , Fragmentos de Peptídeos/imunologia , Mutação Puntual , Proteína C/metabolismo , Medula Espinal/patologia , Baço/imunologia , Baço/patologia , Trombina/genética , Resultado do Tratamento , Fator de Necrose Tumoral alfa/biossíntese , Substância Branca/patologia
9.
Thromb Haemost ; 112(5): 883-92, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25230930

RESUMO

Despite years of research and efforts to translate stroke research to clinical therapy, ischaemic stroke remains a major cause of death, disability, and diminished quality of life. Primary and secondary preventive measures combined with improved quality of care have made significant progress. However, no novel drug for ischaemic stroke therapy has been approved in the past decade. Numerous studies have shown beneficial effects of activated protein C (APC) in rodent stroke models. In addition to its natural anticoagulant functions, APC conveys multiple direct cytoprotective effects on many different cell types that involve multiple receptors including protease activated receptor (PAR) 1, PAR3, and the endothelial protein C receptor (EPCR). Application of molecular engineered APC variants with altered selectivity profiles to rodent stroke models demonstrated that the beneficial effects of APC primarily require its cytoprotective activities but not its anticoagulant activities. Extensive basic, preclinical, and clinical research provided a compelling rationale based on strong evidence for translation of APC therapy that has led to the clinical development of the cytoprotective-selective APC variant, 3K3A-APC, for ischaemic stroke. Recent identification of non-canonical PAR1 and PAR3 activation by APC that give rise to novel tethered-ligands capable of inducing biased cytoprotective signalling as opposed to the canonical signalling provides a mechanistic explanation for how APC-mediated PAR activation can selectively induce cytoprotective signalling pathways. Collectively, these paradigm-shifting discoveries provide detailed insights into the receptor targets and the molecular mechanisms for neuroprotection by cytoprotective-selective 3K3A-APC, which is currently a biologic drug in clinical trials for ischaemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Proteína C/uso terapêutico , Animais , Antígenos CD/fisiologia , Coagulação Sanguínea , Isquemia Encefálica/epidemiologia , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Receptor de Proteína C Endotelial , Endotélio Vascular/fisiopatologia , Ativação Enzimática , Hemorragia/prevenção & controle , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Moleculares , Proteína C/metabolismo , Proteína C/farmacologia , Conformação Proteica , Receptores de Superfície Celular/fisiologia , Receptores Ativados por Proteinase/fisiologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Trombina/farmacologia , Trombina/fisiologia , Ativador de Plasminogênio Tecidual/efeitos adversos , Ativador de Plasminogênio Tecidual/uso terapêutico
10.
Blood ; 123(25): 3979-87, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24740810

RESUMO

Protein S is a cofactor for tissue factor pathway inhibitor (TFPI), accelerating the inhibition of activated factor X (FXa). TFPI Kunitz domain 3 residue Glu226 is essential for enhancement of TFPI by protein S. To investigate the complementary functional interaction site on protein S, we screened 44 protein S point, composite or domain swap variants spanning the whole protein S molecule for their TFPI cofactor function using a thrombin generation assay. Of these variants, two protein S/growth arrest-specific 6 chimeras, with either the whole sex hormone-binding globulin (SHBG)-like domain (Val243-Ser635; chimera III) or the SHBG laminin G-type 1 subunit (Ser283-Val459; chimera I), respectively, substituted by the corresponding domain in growth arrest-specific 6, were unable to enhance TFPI. The importance of the protein S SHBG-like domain (and its laminin G-type 1 subunit) for binding and enhancement of TFPI was confirmed in FXa inhibition assays and using surface plasmon resonance. In addition, protein S bound to C4b binding protein showed greatly reduced enhancement of TFPI-mediated inhibition of FXa compared with free protein S. We show that binding of TFPI to the protein S SHBG-like domain enables TFPI to interact optimally with FXa on a phospholipid membrane.


Assuntos
Lipoproteínas/metabolismo , Proteína S/metabolismo , Sítios de Ligação/genética , Western Blotting , Proteína de Ligação ao Complemento C4b/metabolismo , Fator Xa/metabolismo , Células HEK293 , Humanos , Lipoproteínas/genética , Mutação , Fosfolipídeos/metabolismo , Proteína C/metabolismo , Proteína S/genética , Globulina de Ligação a Hormônio Sexual/metabolismo , Ressonância de Plasmônio de Superfície , Trombina/metabolismo , Tromboplastina/metabolismo
11.
J Thromb Haemost ; 12(7): 1054-65, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24766850

RESUMO

BACKGROUND: Increased hypercoagulability has been reported with low doses of direct thrombin inhibitors but not with direct factor Xa inhibitors. OBJECTIVES: To compare the effects of rivaroxaban with those of melagatran and dabigatran on thrombin generation (TG) and tissue factor-induced hypercoagulability and to explore the possible involvement of the thrombin-thrombomodulin/activated protein C system. METHODS: In normal human plasma and in protein C-deficient plasma, TG was investigated in vitro in the presence and absence of recombinant human soluble thrombomodulin (rhs-TM). TG was determined by calibrated automated thrombography and an ELISA for prothrombin fragments 1+2 (F1+2 ). In an in vivo rat model, hypercoagulability was induced by tissue factor; levels of thrombin-antithrombin (TAT) and fibrinogen and the platelet count were determined. RESULTS: Rivaroxaban inhibited TG in a concentration-dependent manner. In the absence of rhs-TM, melagatran and dabigatran also inhibited TG concentration dependently. However, in the presence of rhs-TM, lower concentrations of melagatran (119-474 nmol L(-1) ) and dabigatran (68-545 nmol L(-1) ) enhanced endogenous thrombin potential, peak TG, and F1+2 formation in normal plasma but not in protein C-deficient plasma. In vivo, rivaroxaban dose-dependently inhibited TAT generation, whereas melagatran showed a paradoxical effect, with an increase in TAT and a small decrease in fibrinogen and platelet count at lower doses. CONCLUSION: Low concentrations of the direct thrombin inhibitors melagatran and dabigatran enhanced TG and hypercoagulability, possibly via inhibition of the protein C system. In contrast, rivaroxaban reduced TG and hypercoagulability under all conditions studied, suggesting that it does not suppress this negative-feedback system.


Assuntos
Antitrombinas/uso terapêutico , Inibidores do Fator Xa/uso terapêutico , Morfolinas/uso terapêutico , Tiofenos/uso terapêutico , Trombofilia/tratamento farmacológico , Tromboplastina/química , Animais , Azetidinas/uso terapêutico , Benzilaminas/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/citologia , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Plasma/metabolismo , Contagem de Plaquetas , Proteína C/metabolismo , Protrombina/metabolismo , Ratos , Ratos Wistar , Rivaroxabana , Tromboelastografia , Trombina/metabolismo , Trombomodulina/metabolismo
12.
Thromb Res ; 133(6): 1105-14, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24513148

RESUMO

INTRODUCTION: Activated protein C (APC) is the central enzyme of the anticoagulant protein C pathway. Low concentrations of APC circulate in plasma and are believed to contribute to the maintenance of a normal haemostatic balance. MATERIALS AND METHODS: We have used a structure-based virtual screening approach to discover small drug-like molecules that inhibit the interaction between APC and its substrate FVa through inhibition of a predominant APC exosite, known to be involved in FVa substrate binding. We have combined in silico selection with functional screening and direct binding analysis to identify novel molecules and to ascertain and characterize the inhibition of the interaction between APC and FVa. RESULTS: We have identified a number of novel molecules that bind to APC and protein C with Kd values in the range of 10(-3)- 10(-5)M. Inhibition by these molecules is incomplete, which most likely reflects the extended surface that is involved in the interaction between APC and its substrates. Direct binding of hit molecules to variant APC molecules that were mutated in the targeted binding site revealed that several of the molecules presented a 100-500 fold lower affinity for the variant molecule, suggesting that these molecules indeed bind the exosite of APC. CONCLUSIONS: The protein-protein interaction inhibitors discovered here, could function as starting molecules for further development of small molecules with anti-APC properties. Such molecules may be of clinical interest, in particular in individuals where thrombin formation is compromised and the haemostatic balance is tipped towards bleeding tendencies, such as in haemophilia A.


Assuntos
Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Sítios de Ligação , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Modelos Moleculares , Proteína C/química , Proteína C/metabolismo , Inibidor da Proteína C/química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
13.
J Nat Med ; 68(1): 144-53, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23760613

RESUMO

6-Methylsulfinylhexyl isothiocyanate (6-MSITC) is an active compound in wasabi (Wasabia japonica Matsum.), which is one of the most popular spices in Japan. 6-MSITC suppresses lipopolysaccharide-induced macrophage activation, arachidonic- or adenosine diphosphate-induced platelet activation, and tumor cell proliferation. These data indicate that 6-MSITC has several biological activities involving anti-inflammatory, anti-coagulant, and anti-apoptosis properties. Endothelial cells (ECs) maintain vascular homeostasis and play crucial roles in crosstalk between blood coagulation and vascular inflammation. In this study, we determined the anti-coagulant and anti-inflammatory effects of 6-MSITC on human umbilical vein endothelial cells (HUVECs). 6-MSITC slightly reduced tissue factor expression, but did not alter von Willebrand factor release in activated HUVECs. 6-MSITC modulated the generation of activated protein C, which is essential for negative regulation of blood coagulation, on normal ECs. In addition, 6-MSITC reduced tumor necrosis factor-α (TNF-α)-induced interleukin-6 and monocyte chemoattractant protein-1 expression. 6-MSITC markedly attenuated TNF-α-induced adhesion of human monoblast U937 cells to HUVECs and reduced vascular cell adhesion molecule-1 and E-selectin mRNA expression in activated ECs. These results showed that 6-MSITC modulates EC function and suppresses cell adhesion. This study provides new insight into the mechanism of the anti-inflammatory effect of 6-MSITC, suggesting that 6-MSITC has therapeutic potential as a treatment for vasculitis and vascular inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Anticoagulantes/farmacologia , Adesão Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Isotiocianatos/farmacologia , Leucócitos/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Relação Dose-Resposta a Droga , Selectina E/genética , Selectina E/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Interleucina-6/metabolismo , Leucócitos/citologia , Leucócitos/metabolismo , Proteína C/metabolismo , RNA Mensageiro/metabolismo , Tromboplastina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Células U937 , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Fator de von Willebrand/metabolismo
14.
Thromb Res ; 127(1): e1-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20926118

RESUMO

UNLABELLED: INTRODUATION: Resveratrol, a phytoestrogen present at a high concentration in red wine, has been reported to possess many health benefit effects that are protective against age-related diseases. Protein S (PS), an important anticoagulant factor in the protein C (PC) anticoagulant pathway, is mainly synthesized by hepatocytes, and its plasma level is decreased in high-estrogen conditions such as pregnancy and oral contraceptive use. The aim of this study was to investigate whether resveratrol affects PS expression in HepG2 cells. MATERIALS AND METHODS: The secreted and intracellular levels of PS were determined by an enzyme-linked ligandsorbent assay and Western blotting. The mRNA expressions of PS, PC and ß chain of C4b-binding protein (C4BP-ß) were analyzed by reverse transcription-polymerase chain reaction. The PS gene promotor activities in HepG2 cells transiently expressing estrogen receptor (ER) α were examined by a luciferase reporter assay. RESULTS: Resveratrol dose- and time-dependently down-regulated the PS expression in HepG2 cells at a transcriptional level, resulting in a significant decrease in secreted PS; however, the PC and C4BP-ß mRNA expressions were not affected. This action of resveratrol was not mediated through either the ER signaling or those of mitogen-activated protein kinases and protein kinase C. Piceatannol, a hydroxylated metabolite of resveratrol, and genistein, an isoflavone found in soy products, also down-regulated the PS expression. CONCLUSIONS: Resveratrol down-regulates the PS expression in HepG2 cells in an ER-independent manner, and the two phenolic hydroxyls at carbon-3 and -5 of resveratrol may be involved in this function.


Assuntos
Proteínas Sanguíneas/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Fitoestrógenos/farmacologia , Proteína S/metabolismo , Estilbenos/farmacologia , Vinho , Proteínas Sanguíneas/genética , Western Blotting , Carcinoma Hepatocelular/genética , Relação Dose-Resposta a Droga , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genisteína/farmacologia , Células Hep G2 , Antígenos de Histocompatibilidade/metabolismo , Humanos , Neoplasias Hepáticas/genética , Estrutura Molecular , Fitoestrógenos/química , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteína C/metabolismo , Proteína S/genética , RNA Mensageiro/metabolismo , Resveratrol , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estilbenos/química , Relação Estrutura-Atividade , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Transfecção
15.
Inflammation ; 34(1): 10-21, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20237952

RESUMO

The present study was performed to investigate the anti-septic effects of Qi-Shao-Shuang-Gan (QSSG), a combination of Astragalus membranaceus saponins (SAM) and Paeonia lactiflora glycosides (GPL), in septic mice induced by cecal ligation and puncture. QSSG was shown to elevate the survival rate of mice, decrease infiltration of polymorphonuclear leukocytes into livers and lungs, lower serum levels of myeloperoxidase, nitric oxide, and lactate dehydrogenase, and decrease mRNA expressions of inducible nitric oxide synthase and interleukin-1ß in livers. It also restored the impaired expressions of protein C (PC) mRNA in mouse livers and expressions of thrombomodulin and endothelial PC receptor mRNA in endothelial cells. Neither SAM nor GPL alone could significantly increase the survival rate of septic mice. The findings indicate that QSSG exerts protective action against polymicrobial sepsis by inhibiting systemic inflammatory response and upregulating PC pathway, and there are synergistic effects between SAM and GPL.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Glicosídeos/uso terapêutico , Saponinas/uso terapêutico , Sepse/tratamento farmacológico , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Astragalus propinquus/química , Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/metabolismo , Ceco/cirurgia , Combinação de Medicamentos , Interleucina-1beta/genética , L-Lactato Desidrogenase/sangue , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Infiltração de Neutrófilos/efeitos dos fármacos , Óxido Nítrico/sangue , Óxido Nítrico Sintase/genética , Paeonia/química , Peroxidase/sangue , Proteína C/genética , Proteína C/metabolismo , Edema Pulmonar/prevenção & controle , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Aleatória , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Sepse/metabolismo , Sepse/patologia , Trombomodulina/genética , Trombomodulina/metabolismo
16.
Orphanet J Rare Dis ; 5: 21, 2010 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-20630065

RESUMO

Hereditary combined vitamin K-dependent clotting factors deficiency (VKCFD) is a rare congenital bleeding disorder resulting from variably decreased levels of coagulation factors II, VII, IX and X as well as natural anticoagulants protein C, protein S and protein Z. The spectrum of bleeding symptoms ranges from mild to severe with onset in the neonatal period in severe cases. The bleeding symptoms are often life-threatening, occur both spontaneously and in a surgical setting, and usually involve the skin and mucosae. A range of non-haemostatic symptoms are often present, including developmental and skeletal anomalies. VKCFD is an autosomal recessive disorder caused by mutations in the genes of either gamma-glutamyl carboxylase or vitamin K2,3-epoxide reductase complex. These two proteins are necessary for gamma-carboxylation, a post-synthetic modification that allows coagulation proteins to display their proper function. The developmental and skeletal anomalies seen in VKCFD are the result of defective gamma-carboxylation of a number of non-haemostatic proteins. Diagnostic differentiation from other conditions, both congenital and acquired, is mandatory and genotype analysis is needed to confirm the defect. Vitamin K administration is the mainstay of therapy in VKCFD, with plasma supplementation during surgery or severe bleeding episodes. In addition, prothrombin complex concentrates and combination therapy with recombinant activated FVII and vitamin K supplementation may constitute alternative treatment options. The overall prognosis is good and with the availability of several effective therapeutic options, VKCFD has only a small impact on the quality of life of affected patients.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/fisiopatologia , Fatores de Coagulação Sanguínea/metabolismo , Proteínas Sanguíneas/metabolismo , Proteína C/metabolismo , Proteína S/metabolismo , Deficiência de Vitamina K/congênito , Transtornos Herdados da Coagulação Sanguínea/diagnóstico , Carbono-Carbono Ligases/genética , Carbono-Carbono Ligases/metabolismo , Humanos , Recém-Nascido , Proteínas Recombinantes/uso terapêutico , Vitamina K/uso terapêutico , Deficiência de Vitamina K/genética
17.
Ter Arkh ; 82(3): 36-8, 2010.
Artigo em Russo | MEDLINE | ID: mdl-20564920

RESUMO

AIM: To study the time course of changes in the activity of the protein C system and other hemostatic parameters under intravascular laser irradiation of blood (ILIB) in patients with community-acquired pneumonia (CAP). SUBJECTS AND METHODS: One hundred and forty patients aged 17 to 62 years (mean 39.5 +/- 8.4 years) with CAP were examined. A control group (n = 40) received conventional drug therapy; the study group (n = 100) had a course of ILIB in addition to conventional therapy. RESULTS: Before treatment, the patients with CAP were observed to have a lower protein C system activity and the signs of hypercoagulation that were eliminated by ILIB. CONCLUSION: ILIB is an effective method in correcting hemocoagulative disorders in patients with CAP.


Assuntos
Coagulação Sanguínea/efeitos da radiação , Terapia com Luz de Baixa Intensidade/métodos , Pneumonia Bacteriana/radioterapia , Adolescente , Adulto , Antitrombina III/metabolismo , Infecções Comunitárias Adquiridas , Humanos , Masculino , Pessoa de Meia-Idade , Pneumonia Bacteriana/sangue , Proteína C/metabolismo , Resultado do Tratamento , Adulto Jovem
18.
Eur J Pharmacol ; 635(1-3): 103-8, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20123096

RESUMO

The present study was attempted to evaluate the therapeutic effects of activated protein C and/or hyperbaric oxygen in an animal model of heatstroke. Sixty-eight minutes heat stress (43 degrees C) initiated, the anesthetized rats were randomized to several groups and administered: 1) no resuscitation (vehicle solution plus normabaric air, 2) intravenous activated protein C (1mg in 1ml of normal saline per kg of body weight), 3) hyperbaric oxygen (100% oxygen at 202kpa for 17min), and 4) intravenous activated protein C plus hyperbaric oxygen. Another group of rats exposed to room temperature (26 degrees C) was used as normothermic controls. Blood sampling was 0min, 70min, and 85min after heat stress initiated. When the vehicle-treated rats underwent heat exposure, their survival time values found were to be 19-25min. Resuscitation with activated protein C or hyperbaric oxygen significantly and equally improved survival during heatstroke (134-159min). As compared with those of activated protein C or hyperbaric oxygen alone, combined activated protein C and hyperbaric oxygen significantly had higher survival time values (277-347min). All vehicle-treated heatstroke animals displayed systemic response, hypercoagulable state, and hepatic and renal dysfunction. Combined activated protein C and hyperbaric oxygen therapy reduced these heatstroke reactions better than activated protein C or hyperbaric oxygen alone. The results indicate consequently, combined activated protein C and hyperbaric oxygen therapy heightens benefit in combating heatstroke reactions.


Assuntos
Golpe de Calor/terapia , Oxigenoterapia Hiperbárica , Proteína C/metabolismo , Proteína C/uso terapêutico , Animais , Ativação Enzimática , Golpe de Calor/complicações , Golpe de Calor/tratamento farmacológico , Hipotensão/complicações , Hipotensão/tratamento farmacológico , Hipotensão/terapia , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/terapia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Fígado/efeitos dos fármacos , Fígado/fisiopatologia , Insuficiência de Múltiplos Órgãos/complicações , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Insuficiência de Múltiplos Órgãos/terapia , Proteína C/farmacologia , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Trombofilia/complicações , Trombofilia/tratamento farmacológico , Trombofilia/terapia
19.
Pathophysiol Haemost Thromb ; 37(2-4): 88-97, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21430357

RESUMO

Although thromboembolism is a problematic complication of chemotherapy, the pathogenic mechanisms by which chemotherapeutic agents exert prothrombotic effects in vivo are unclear.The objective of this study was to examine the effects of adjuvant chemotherapy on thrombin generation, the protein C anticoagulant pathway, and microparticle tissue factor (MP TF) activity in 26 breast cancer patients (stages I to III). The patients received cyclophosphamide, 5-fluorouracil, and methotrexate, epirubicin, or doxorubicin. Plasma samples were collected on day 1 (baseline), day 2, and day 8 for the first 2 cycles of chemotherapy. Levels of thrombin-antithrombin (TAT) complexes, MP TF activity, and components of the protein C anticoagulant pathway, including protein C, activated protein C (APC), soluble thrombomodulin (sTM), and soluble endothelial protein C receptor (sEPCR), were measured. Compared to prechemotherapy baseline levels, plasma TAT, protein C, and APC were significantly different following the administration of chemotherapy (p < 0.01 for each). Plasma TAT was higher in cycle 1, day 2, and cycle 2, day 8, compared to baseline. Plasma protein C levels were lower in cycle 2, day 8, whereas plasma APC levels were lower in cycle 2, day 1, and cycle 2, day 8. No significant changes were found in plasma sEPCR, sTM, or MP TF activity. This study suggests that adjuvant chemotherapy in women with breast cancer increases thrombin generation and impairs the endothelium-based protein C anticoagulant pathway.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias da Mama/sangue , Neoplasias da Mama/tratamento farmacológico , Proteína C/metabolismo , Trombina/metabolismo , Adulto , Antígenos CD/sangue , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Antitrombina III , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Receptor de Proteína C Endotelial , Epirubicina/administração & dosagem , Epirubicina/efeitos adversos , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/efeitos adversos , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Peptídeo Hidrolases/sangue , Receptores de Superfície Celular/sangue , Trombomodulina/sangue , Tromboplastina/metabolismo , Trombose/sangue , Trombose/induzido quimicamente , Fatores de Tempo
20.
Blood ; 114(8): 1658-65, 2009 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-19531655

RESUMO

Protamine sulfate is a positively charged polypeptide widely used to reverse heparin-induced anticoagulation. Paradoxically, prospective randomized trials have shown that protamine administration for heparin neutralization is associated with increased bleeding, particularly after cardiothoracic surgery with cardiopulmonary bypass. The molecular mechanism(s) through which protamine mediates this anticoagulant effect has not been defined. In vivo administration of pharmacologic doses of protamine to BALB/c mice significantly reduced plasma thrombin generation and prolonged tail-bleeding time (from 120 to 199 seconds). Similarly, in pooled normal human plasma, protamine caused significant dose-dependent prolongations of both prothrombin time and activated partial thromboplastin time. Protamine also markedly attenuated tissue factor-initiated thrombin generation in human plasma, causing a significant decrease in endogenous thrombin potential (41% +/- 7%). As expected, low-dose protamine effectively reversed the anticoagulant activity of unfractionated heparin in plasma. However, elevated protamine concentrations were associated with progressive dose-dependent reduction in thrombin generation. To assess the mechanism by which protamine mediates down-regulation of thrombin generation, the effect of protamine on factor V activation was assessed. Protamine was found to significantly reduce the rate of factor V activation by both thrombin and factor Xa. Protamine mediates its anticoagulant activity in plasma by down-regulation of thrombin generation via a novel mechanism, specifically inhibition of factor V activation.


Assuntos
Fator V/antagonistas & inibidores , Protaminas/farmacologia , Trombina/metabolismo , Animais , Anticoagulantes/farmacologia , Tempo de Sangramento , Coagulação Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Fator V/metabolismo , Fator VIIIa/metabolismo , Heparina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Proteína C/metabolismo , Proteína C/fisiologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA