Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Transfus Apher Sci ; 62(1): 103625, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36585276

RESUMO

The latest WHO report determined the increasing diversity within the CoV-2 omicron and its descendent lineages. Some heavily mutated offshoots of BA.5 and BA.2, such as BA.4.6, BF.7, BQ.1.1, and BA.2.75, are responsible for about 20% of infections and are spreading rapidly in multiple countries. It is a sign that Omicron subvariants are now developing a capacity to be more immune escaping and may contribute to a new wave of COVID-19. Covid-19 infections often induce many alterations in human physiological defense and the natural control systems, with exacerbated activation of the inflammatory and homeostatic response, as for any infectious diseases. Severe activation of the early phase of hemostatic components, often occurs, leading to thrombotic complications and often contributing to a lethal outcome selectively in certain populations. Development of autoimmune complications increases the disease burden and lowers its prognosis. While the true mechanism still remains unclear, it is believed to mainly be related to the host autoimmune responses as demonstrated, only in some patients suffering from the presence of autoantibodies that worsens the disease evolution. In fact in some studies the development of autoantibodies to angiotensin converting enzyme 2 (ACE2) was identified, and in other studies autoantibodies, thought to be targeting interferon or binding to annexin A1, or autoantibodies to phospholipids were seen. Moreover, the occurrence of autoimmune heparin induced thrombocytopenia has also been described in infected patients treated with heparin for controlling thrombogenicity. This commentary focuses on the presence of various autoantibodies reported so far in Covid-19 diseases, exploring their association with the disease course and the durability of some related symptoms. Attempts are also made to further analyze the potential mechanism of actions and link the presence of antibodies with pathological complications.


Assuntos
COVID-19 , Hemostáticos , Humanos , COVID-19/complicações , Síndrome , Autoanticorpos , Progressão da Doença
2.
Transfus Apher Sci ; 60(4): 103209, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34244081

RESUMO

There is an increasing interest in Extracellular Vesicles released by many cells through membrane shedding. In addition to cell signaling, these particles are true messenger cargos, which can carry cell surface proteins, miRNAs and non-coding RNAs to other and distant cells. They are part of the inter-cellular crosstalk and they contribute to transferring biological messages far away from the triggering event. EVs are biomarkers of many diseases, including thrombo-embolic pathology, infections, neurological or metabolic disorders, and malignancy. Their role and significance are presented and discussed in this short review, as consequences of disease and causes of its progression. But they can also be beneficial for tissue healing or repair, and they can be prepared in vitro to be used for cell- targeted treatments. Many identification and measurement methods for EV's are sophisticated, which restricts their use to research studies, but they have, nevertheless, a high laboratory potential for diagnosis, prognosis and evolution as follow-up of many pathologies. New emerging laboratory tools offer more friendly and easy applications for characterizing EVs and testing their associated activity, especially for the procoagulant ones.


Assuntos
Biomarcadores Tumorais/sangue , Vesículas Extracelulares/metabolismo , Infecções/sangue , Doenças Metabólicas/sangue , Neoplasias/sangue , Doenças do Sistema Nervoso/sangue , Tromboembolia/sangue , Animais , Comunicação Celular , MicroRNA Circulante/sangue , Humanos , RNA Neoplásico/sangue
3.
Transfus Apher Sci ; 59(4): 102861, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32636115

RESUMO

Culture of blood cells, mainly erythrocytes, at industrial levels complying with cGMP regulations, aim to make them available, at large scale, any time and everywhere, when needed for transfusion, or laboratory applications. Understanding how blood cells differentiate and develop in-vivo, and mechanisms of differentiation and growth factors, has opened newer strategies for in-vitro culture from multipotent stem cells or immortalized lines. This offers interesting perspectives for obtaining such cultured bioproduct cells for medical applications. In addition, many attempts for preparing platelets in-vitro from megakaryocyte culture have been reported. Nevertheless, the quantities of functional viable platelets obtained are still not sufficient to envisage transfusion applications. Other strategic approaches concern culture of organoids, which can synthesize functional blood proteins, but still significant scale-up of yield needs to be addressed. Finally, considerable advances have been made in culturing specific lymphocytes for personalized immunotherapy of some cancer patients with highly promising results in certain applications. This concise mini report focuses on the progress made in these directions, and attempts are made to describe some newer perspectives.


Assuntos
Técnicas de Cultura de Células/métodos , Eritrócitos/metabolismo , Organoides/metabolismo , Humanos
4.
Transfus Apher Sci ; 59(3): 102804, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32387238

RESUMO

Covid-19 is characterized by weak symptoms in most affected patients whilst severe clinical complications, with frequent fatal issues, occur in others. Disease severity is associated with age and comorbidities. Understanding of viral infectious mechanisms, and antibody immune response, can help to better control disease progression. SARS-CoV-2 has a major impact on the Renin Angiotensin Aldosterone System (RAAS), through its binding to the membrane cellular glycoprotein, Angiotensin Converting Enzyme-2 (ACE-2), then infecting cells for replication. This report hypothesizes the possible implication of an autoimmune response, induced by generation of allo- or autoantibodies to ACE-2, or to its complexes with viral spike protein. This could contribute to some delayed severe complications occurring in affected patients. We also propose a strategy for investigating this eventuality.


Assuntos
Anticorpos Antivirais/imunologia , Autoanticorpos/imunologia , Autoimunidade , Betacoronavirus/imunologia , Coagulação Sanguínea , Infecções por Coronavirus/sangue , Isoanticorpos/imunologia , Peptidil Dipeptidase A/imunologia , Pneumonia Viral/sangue , Sistema Renina-Angiotensina/fisiologia , Trombofilia/etiologia , Enzima de Conversão de Angiotensina 2 , Especificidade de Anticorpos , COVID-19 , Infecções por Coronavirus/complicações , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/imunologia , Progressão da Doença , Coagulação Intravascular Disseminada/etiologia , Humanos , Pandemias , Peptidil Dipeptidase A/sangue , Peptidil Dipeptidase A/fisiologia , Pneumonia Viral/complicações , Pneumonia Viral/imunologia , SARS-CoV-2 , Serina Endopeptidases/fisiologia , Índice de Gravidade de Doença , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Trombofilia/sangue , Trombofilia/imunologia , Fatores de Tempo
5.
Eur J Haematol ; 103(2): 137-139, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31102471

RESUMO

We report a very high factor V inhibitor affecting the measurement of all coagulation factors besides fibrinogen, all these factors being dramatically decreased. This inhibitor could be linked to antibiotic use. The patient died of massive hemorrhage before a plasma exchange could be initiated.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/sangue , Coagulação Sanguínea , Fator V/antagonistas & inibidores , Hemorragia/sangue , Hemorragia/etiologia , Idoso de 80 Anos ou mais , Biomarcadores , Testes de Coagulação Sanguínea , Suscetibilidade a Doenças , Evolução Fatal , Feminino , Hemorragia/diagnóstico , Humanos
6.
Transfus Apher Sci ; 58(2): 216-222, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30954379

RESUMO

The contact system initiates the intrinsic pathway of coagulation and is started by Factor XII activation, which then activates prekallicrein to kallicrein and Factor XI to Factor XIa and, in the presence of high molecular weight kininogen, forms a "contact phase activation loop", that amplifies Factor XII activation. FXII deficiency is not associated with bleeding tendencies, but when the blood clots, the thrombus is less dense, thus favoring antithrombotic protection. Activated Factor XII inhibition emerges as an efficient target for preventing thrombo-embolic diseases without inducing a hemorrhagic risk. Activated Factor XII exhibits other activities, in that it can activate complement and provoke inflammation, contributing to innate immunity. It also stimulates fibrinolysis through uPA activation from scu-PA. Among the other components of the contact phase, Factor XI has a more important role in coagulation pathways and can directly activate FX, FVIII and FV, in a FIX independent pathway. Its deficiency is associated with a mild bleeding diathesis ("pseudo-hemophilia" or hemophilia C), with a variable incidence among kindreds. Recently, the occurrence of thrombotic events the same day following infusion of immunoglobulin concentrates has been demonstrated to be caused by the presence of trace amounts of activated Factor XI, pointing out the key role of this factor for thrombogenicity. Prekallicrein can be activated at the endothelial surface in the presence of high molecular weight kininogen, whose cleavage generates bradykinins and contributes to vessel tonicity and inflammation. The contact phase, through its activation loop, is then an important physiological system, which can initiate and regulate various biological functions and is at the crossroads of various biological activities. Many of the body's physiological functions are intimately linked between them, making the global approach of special usefulness for understanding the interactions which can result from any abnormality of one of them. New pharmaceutical drugs targeting a defined activity need to be investigated for all the possible interferences or side effects. In this article we aim to present and summarize the present understanding of contact phase system activation and regulation, its involvement in various physiological functions, and the laboratory tools for its exploration.


Assuntos
Transtornos da Coagulação Sanguínea/fisiopatologia , Coagulação Sanguínea/fisiologia , Fibrinólise/fisiologia , Humanos
7.
Transfus Apher Sci ; 58(4): 538-544, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31256946

RESUMO

Although suspected conceptually in the 60 s, Protein C and Protein S activities in hemostasis were investigated and reported from the mid-80 s, followed by the discovery of Thrombomodulin, an endothelial cell membrane associated protein, playing the most important heamostatic role. These 3 proteins act in regulating thrombogenesis and protecting against thrombo-embolic events. When blood is activated, any trace of circulating thrombin is captured by Thrombomodulin in the microcirculation, making thrombin become an anticoagulant through its capacity to activate Protein C to Activated Protein C, which operates as a sentinel in blood coagulation, in the form of a complex with free Protein S, to block any new blood activation site, and more especially circulating activated Factors V and VIII. Protein S not only acts as the Activated Protein C cofactor, but also as the cofactor of Tissue Factor Pathway Inhibitor. In addition, it has some functions in the complement pathway through its binding to C4b-BP. Another capability of activated protein C is to lower fibrinolytic activity, as the Activated Protein C Inhibitor is also known as Plasminogen Activator Inhibitor 3. The Protein C-Protein S system becomes less efficient in the presence of mutated Factor V (Factor V-Leiden or other variants), which is resistant to its inactivating effect. Other pathologies linked to this system concern the development of allo- or auto-antibodies to Protein S or to thrombin, which can generate severe thrombotic complications in affected patients. Some antithrombotic drugs have originated from this regulatory system. Protein C or Protein S concentrates are used for treating deficient patients. Activated Protein C (especially in patients with sepsis) or Thrombomodulin are proposed as antithrombotic medications. Most importantly, congenital or acquired Protein C or Protein S deficiencies are associated with severe recurrent thrombotic events. From the clinical standpoint most of the patients are heterozygous, as homozygosity is almost incompatible with life in the absence of a continuous and efficient treatment. Laboratory investigation of this highly complex system involves many different specialized assays for measuring these 3 proteins' activities, their antigenic content or their genetic sequence. The Protein S in-vitro anticoagulant activity is weak and contrasts with its high antithrombotic role in-vivo, showing that diagnostic assays have not yet succeeded in reproducing all the natural mechanisms for evidencing the anticoagulant role of Protein S. This paradoxal notion is discussed and illustrated in this manuscript as well is a revisit of the major characteristics and pathophysiological functions of the Protein C-Protein S-Thrombomodulin system; the associated pathologies; and the main laboratory tools available for clinical diagnosis. In respect to future perspectives, we also focused on developing more significant and relevant assays, especially for Protein S, thanks to the understanding of its biological roles.


Assuntos
Proteína C/metabolismo , Proteína S/metabolismo , Transdução de Sinais , Trombomodulina/metabolismo , Animais , Coagulação Sanguínea , Humanos
8.
Transfus Apher Sci ; 57(6): 804-811, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30401517

RESUMO

This manuscript aims to provide a concise review on current diagnostic/ confirmatory strategies of Heparin Induced Thrombocytopenia (HIT) with the combined use of immunological / functional assays in addition to the clinical probability. Laboratory diagnosis of HIT is of primordial importance as the related complications could become rapidly severe and life-threatening and can provoke limb amputation in some cases. The first action in the presence of HIT suspicion is to withdraw heparin and to initiate an alternative anticoagulant. Whilst vitamin K antagonists are not appropriate, anticoagulant options include Fondaparinux, Sodium Danaparoid, DOACs, Argatroban, and Bivalirudin. However, if HIT is excluded, patients can benefit again from the high therapeutic and antithrombotic efficacy of this drug, which remains superior to all the substitutive anticoagulant treatments. HIT is suspected in the presence of a platelet count drop > 50% on 2 successive counts, or a platelet count < 100 G/L, and of a significant clinical probability (4 Ts score). Testing patients' plasma is required for establishing the diagnosis. Laboratory investigation involves first the immunological measurement of heparin dependent IgG antibodies (mainly targeted to Heparin-Platelet Factor 4 complexes). When positive, a functional assay for platelet activation, performed at a low and high heparin concentration, allows confirming this disease. In any case, if the immuno-assay is negative, HIT can be excluded with a high probability, and heparin can be continued (if clinical examination favors this decision). Conversely, the higher the IgG antibody concentration is (and affinity), the higher is the probability of developing HIT. The functional assay has now become for confirming the platelet activation capacity of antibodies, and therefore confirming the presence of HIT. Up to now, the gold reference method for testing antibody-dependent platelet activation is the C14-Serotonin Release Assay, available only in very few laboratories working with radio-isotopes. A simple, sensitive, and accurate flow cytometry assay becomes now available to all clinical sites, and it can be easily used for testing the capacity of heparin dependent-antibodies to activate platelets, at low heparin concentration. This technique can be performed in any laboratory equipped with a flow cytometer and can make the HIT confirmation diagnosis rapidly available, which introduces a great improvement for management of patients with HIT. We believe that an evidence-based update on this topic is timely and well warranted.


Assuntos
Heparina/efeitos adversos , Imunoensaio/métodos , Trombocitopenia/induzido quimicamente , Trombocitopenia/diagnóstico , Anticorpos/imunologia , Humanos , Probabilidade , Reprodutibilidade dos Testes , Trombocitopenia/imunologia
9.
Transfus Apher Sci ; 57(3): 363-369, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29895509

RESUMO

On demand and prophylaxis usage of FVIII/ FIX concentrates for the therapeutic management of hemophilia has greatly changed quality of life, and healthy life span of affected patients. Availability of recombinant therapeutic FVIII and FIX products, and of their long-acting variants, further improves the treatment constraints, and progressively permits to hemophiliacs to have an almost normal way of life. Unlimited amounts of recombinant or engineered substitutive products become available, and open new avenues for extending the benefits of prophylaxis to all hemophiliac patients, not only in economically advanced territories, but also in emerging and developing countries, worldwide. Pharmacokinetics of injected products can be variable among treated patients, and dependent on age. In addition, patient medical status, existing diseases, and the nature of joint damages can impact protective effect of substitutive products, and risks associated to way of life and activity. Product requirements and half-life of infused products are therefore patient specific. Monitoring recoveries of injected products thus provide useful information for the most appropriate treatment adjustment. FVIII and FIX measurements in plasma of treated patients helps to establish the optimal interval between injections for each treated patient, and the overall therapeutic cost. Due to the high variability from reagent to reagent, and the different behavior from plasma derived products, clotting methods are not ideal for recombinant and long-acting products. They require to be performed only in association with a drug specific calibrator. They are not recommended for patients' survey, due to the high variety of reagents available. Chromogenic assays (2-stage methods) offer a standard reactivity to all available FVIII or FIX products in drugs, whether the way they are obtained, or in plasma. In a subset of treated patients, inhibitory antibodies to FVIII or FIX develop and can be measured with inhibition assays (Bethesda units), or by Elisa. Unfortunately, FVIII or FIX substitutive therapies cannot be used in patients with inhibitors, and alternative clinical management is requested, such as the use of FEIBA or FVIIa for their bypassing activity. A new treatment is being introduced in the form of a bispecific antibody (Emicizumab) targeted to both FIXa and FX, and which allows activating FX by FIXa without the need for FVIII. Some chromogenic assays (Biophen FVIII), designed with human proteins, offer the possibility to measure the activity and recovery of this new drug. Chromogenic methods are then useful for establishing potency of therapeutic products or monitoring recovery and kinetics in treated patients, through plasma measurements. Availability of International Standards for FVIII and FIX, in concentrates or plasma, allows harmonization of assay results.


Assuntos
Testes de Coagulação Sanguínea/métodos , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Bioensaio , Fator VIII/farmacologia , Humanos
10.
Transfus Apher Sci ; 57(2): 291-297, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29784539

RESUMO

Antithrombin [AT] is the main inhibitor for activated plasma coagulation serine esterases, inhibiting thrombin, Factors Xa and IXa, but also Factors XIIa, XIa, VIIa, kallicrein, and plasmin. Its activity is highly enhanced by heparin, through binding to the pentasaccharide sequences, for inhibition of all coagulation proteases, except thrombin, which inhibition requires its additional binding to the heparin polysaccharide chain. However, AT is the major inhibitor of thrombin in the blood circulation. Congenital or acquired deficiencies of AT expose affected patients to an increased risk of developing unprovoked and recurrent thrombo-embolic diseases. Antithrombin can be measured with various laboratory techniques, by either immunological or functional methods. Earlier, a radial immunodiffusion immunoassay allowed measurement of the protein antigenic content. Functional assays are mainly designed with Anti-Thrombin or Anti-Factor Xa chromogenic methods and are useful for detecting genetic molecular mutations with decreased inhibitory activity and contributed to study the conformational changes of antithrombin and its variants, which potentially regulate the activity of this serine protease inhibitor. These assays are not equivalent in terms of diagnosing protein abnormalities, associated with increased thrombotic incidence, and they have variable performance for reflecting impaired antithrombin binding capacity for heparin, reduced progressive inhibition of serine proteases, or accelerated switch rates to the latent and less active forms. A small proportion of AT (<10%) is present in blood in the ß-form, with a lower oligosaccharide content, a lower Molecular Weight, a higher binding rate to endothelial glycosaminoglycans, and a higher anticoagulant activity, hence requiring specific laboratory methods for its measurement. The ß-AT form is then of critical importance for controlling blood activation by tissue injury and preventing development of thrombo-embolic diseases. This article reviews the performance characteristics of the currently available assays, and their usefulness for monitoring the use of AT concentrates in intensive care units, disseminated intravascular coagulation or severe infections, to restore the anticoagulant protective effect of heparin by supplementing the requested AT concentration. The issues of automation, harmonization and standardization are also revisited and discussed.


Assuntos
Deficiência de Antitrombina III/diagnóstico , Antitrombina III/análise , Técnicas de Laboratório Clínico/métodos , Antitrombina III/administração & dosagem , Antitrombina III/genética , Antitrombina III/metabolismo , Deficiência de Antitrombina III/congênito , Deficiência de Antitrombina III/tratamento farmacológico , Deficiência de Antitrombina III/genética , Variação Genética , Humanos , Isoformas de Proteínas
11.
Transfus Apher Sci ; 57(1): 118-126, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29525568

RESUMO

For many years, the importance of fibrinolysis has been recognized, first for its intravascular antithrombotic action, and more recently for its many extravascular activities, associated with matrix degradation and tissue remodeling. In the blood circulation system, fibrinolysis prevents thrombosis, and is associated with various biological and clinical situations: risk factors for cardio-vascular diseases in high risk clinical situations (type II diabetes, hypertension, triglycerides, high BMI, elevated glucose, etc.), probably resulting from a significant reduction of the fibrinolysis potential, and elevation of PAI-1. Noteworthy, t-PA is mainly present as an inactive complex with PAI-1, and its concentration in plasma tends to follow that of PAI-1, but in a lesser extent. Hypofibrinolysis can favor the occurrence of thrombotic events, and possibly other biological dysfunctions. Fibrinolysis activity is however difficult to evaluate as it has a delayed activity after clot formation, is initiated and regulated after fibrin generation, and conversely to clotting, its action is delayed (long lag phase) and slow, before being dramatically amplified leading to rapid clot dissolution. We have designed a new assay for evaluating the global fibrinolytic capacity (GFC) in the body. Reagents are used in association with a specific instrument, which can be connected to any computer, and dedicated software is used for analyzing clot lysis kinetics. The assay is performed in a micro-cuvette, introduced into one of the instrument wells at 37 °C, and light transmittance is continuously measured. Assayed plasma is first supplemented with a limited and constant amount of t-PA with silica and is then clotted with thrombin and calcium. Clot dissolution (measurement of turbidity change) is recorded over time using the dedicated instrument (Lysis Timer), and clot lysis kinetics are analyzed with the associated software: primary and secondary derivatives of the light transmission curve give information on kinetics and completion of clot dissolution. Total assay time is about 1 h (but in the presence of hypofibrinolysis it can be prolonged). The concentration of t-PA used for the assay has been adjusted (100 ng/ml) to obtain an optimal sensitivity to hypofibrinolysis within a short time interval, and clot dissolution occurs within about 45 min for normal individuals, with a broad range from 30 min to 60 min, with some samples presenting a clot dissolution time >60 min (hypofibrinolysis). This new assay is performed with the tested plasma intrinsic factors, especially its own fibrinogen, and only exogeneous t-PA is added. GFC is highly sensitive to PAI-1 activity, but other factors regulating fibrinolysis contribute to the clot dissolution kinetics. Freshly prepared or frozen and thawed citrated plasma can be used. The usefulness of this assay for clinical applications is under investigation. Although fibrinolysis is mainly initiated in the body upon stimulation or blood clotting, and rapidly diluted and inhibited in the circulation, evaluation of its "residual" activity in plasma is expected to reflect its global body potential.


Assuntos
Fibrinólise , Hemostasia , Trombose/sangue , Testes de Coagulação Sanguínea/métodos , Humanos , Fatores de Risco
12.
Platelets ; 29(1): 95-97, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28960123

RESUMO

BACKGROUND: Light transmission aggregometry (LTA) is considered as the gold standard for testing platelet function in the setting of both platelet disorders suspicion and response to antiplatelet therapy evaluation. LTA requires however specialized equipment, substantial blood sample volumes, is technically challenging and time-consuming. AIM: To evaluate an automated platelet aggregation method performed on a routine coagulation analyzer Sysmex CS-2000i. METHODS: 46 patients presenting a bleeding syndrome and 62 patients with acute coronary syndrome receiving dual antiplatelet therapy were studied in total. Platelet aggregations were performed on CS-2000i equipped with a dedicated software and on APACT-4004 (Elitech, France) as the reference instrument. Aggregation was measured by monitoring the changes in light absorbance occurring in response to ADP 2.5, 5 and 10µM, collagen 3.3 µg/mL, epinephrin 10µM, ristocetin 1.25 mg/mL and arachidonic acid 0.5 mg/mL in platelet rich plasma (PRP). PRP were tested simultaneously on both CS-2000i and APACT-4004 devices. Platelet stirred speed were 800 rpm for both instruments. RESULTS: Significant correlations were observed between CS-2000i and LTA after all stimulations (p< 0.001). Patients presenting a bleeding syndrome had similar aggregation profiles with both methods. A single patient presented a severe platelet disorder (Glanzmann Thrombasthenia) and its PRP showed defective aggregation in response to all agonists except ristocetin with both instruments. Finally, the inter-agreement rates for CS-2000i and APACT-4004 to detect low responders to thienopyridines or aspirine were strong (weighted kappa> 0.70). CONCLUSION: Platelet aggregation on the routine coagulation analyzer CS-2000i is an easily accessible, handy, reliable, standardized, and rapid tool to assess platelet function which allows to skirt most of the LTA limitations.


Assuntos
Testes de Coagulação Sanguínea/métodos , Testes de Função Plaquetária/métodos , Biomarcadores/sangue , Testes de Coagulação Sanguínea/instrumentação , Transtornos Plaquetários/sangue , Transtornos Plaquetários/diagnóstico , Transtornos Plaquetários/tratamento farmacológico , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Humanos , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Testes de Função Plaquetária/instrumentação , Reprodutibilidade dos Testes
13.
Transfus Apher Sci ; 56(3): 279-286, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28552490

RESUMO

Thrombotic diseases caused by cancer progression have been reported as one of the major causes of cancer associated morbidity and mortality along with cancer invasiveness and infectious complications. Moreover, anticoagulant therapy with heparin and heparin-like drugs, or vitamin K antagonists, or the Direct Oral Anticoagulants, is seeing an extended application in cancer patients and offers prolonged life expectancy to oncology patients for whom blood activation and thrombotic events have a variable incidence, depending on cancer type. Laboratory tools are highly useful for identifying patients at thrombotic risk through the measurement of blood activation markers and selecting those appropriate for anticoagulant therapy. Among the pathological markers, DDimer or Extracellular Vesicles have the highest diagnostic value in these pathological conditions. Global assays are useful for dosage adjustment, such as assessing either an induced anticoagulant effect or the measurement of drug activity. Various assays are also developed such as platelet aggregometry techniques for evaluating drug induced- aggregates or methods allowing measurement of the drug activity to its targeted coagulation factors such as: heparin to thrombin or Factor Xa; DOACs to Thrombin or Factor Xa (Dabigatran to thrombin and DiXaIs, Rivaroxaban, Apixaban, and Edoxaban, to Factor Xa). Such explorative techniques help to find the right dosage adjustment to protect patients from developing thrombosis without exposing them bleeding. It also permits exploration of unexpected drug behavior in treated patients, to check the right adherence to therapy in long-term anticoagulant protocols, and prevention of bleeding in patients with impaired renal or hepatic function. Complementary use of blood activation markers brings additional information on the curative effects of the anticoagulant therapy, and allows identification of pro-thrombotic activity in the clinically silent state. These issues are concisely addressed below.


Assuntos
Anticoagulantes/uso terapêutico , Biomarcadores/sangue , Neoplasias/tratamento farmacológico , Trombose/tratamento farmacológico , Anticoagulantes/farmacologia , Humanos , Trombose/sangue
14.
Transfus Apher Sci ; 56(2): 107-117, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28343935

RESUMO

Neonates and children can develop rare bleeding disorders due to congenital/acquired coagulation Factor deficiencies, or allo-immune/autoimmune complications, or can undergo surgeries at high haemorrhagic risk. They then need specialized transfusion of blood components/products, or purified blood extracted products or recombinant proteins. Blood-derived therapies conventionally used for management of affected infants with genetic/acquired deficiencies, bleeding problems (coagulation Factor reduced or missing) or thrombotic disorders (reduced or missing anticoagulant proteins) pose some additional risks. These remedial therapies can cause tolerance when used very early in life and, sometimes needed, repeatedly. The introduction of recombinant proteins has allowed manufacturers to produce large amounts of the proteins usually present at very low concentration in blood. This has also changed the risk pattern of plasma-extracted products, especially in terms of continual reduction of viral transmission. Many efforts have been made over these past decades to reduce the risks associated with the use of all these products in terms of viral and bacterial safety, as well as immune disorders but they are not the objective of this article. Other associated side effects are the presence of undesired activities in blood products, which can produce thrombotic events or adverse reactions. The progressive introduction of blood derived products has greatly improved the prognosis and quality of life of affected patients. This concerns whole blood, but also blood cell concentrates, mainly platelets and red blood cells, plasma, while the blood extracted products are increasingly replaced by recombinant proteins. All these therapeutic products, i.e. blood extracted drugs, improve health and quality of life for hemophiliac's A or B, or patients with auto/allo-immune thrombocytopenias or with rare bleeding disorders, and those with thrombotic events occurring in childhood, which are mainly due to Protein C or Protein S deficiencies (congenital or acquired). Progress in analytical methods and biotechnology allow better control of the manufacturing processes for all blood derived or plasma extracted products and recombinant proteins, and contribute to improved manufacturing processes to minimize the occurrence of side effects. These adverse events can be due to the aging of the blood cell concentrate with release of their granule content, and generation of EVs, which can produce anaphylactic reactions and risk of thrombosis, but also to the presence of activated coagulation Factors in purified products, such as Factor Xia as recently identified in immunoglobulin concentrates. Characterization and measurement of contaminant products is of special usefulness during product preparation and for optimization of manufacturing processes for purified extracted products, but also for recombinant proteins. The pharmaceutical industry introduces these new methods for validating manufacturing processes, or for quality control assessments. The objective is first to warrant the full quality and safety of the lots produced, and assure the highest efficacy with the lowest risks when used in patients. For cell concentrates and fresh blood, storage conditions are critical and measurement of analytes such as EVs or Annexin V allows evaluation of quality of each individual transfused pouch. In addition to all the rules around viral and bacterial transmission risk, and immune tolerance, our available laboratory methods contribute to reducing the side effects of blood cell concentrates and derived plasma products, as well as those of the therapeutic recombinant proteins.


Assuntos
Transfusão de Componentes Sanguíneos , Pediatria , Reação Transfusional/prevenção & controle , Transfusão de Componentes Sanguíneos/instrumentação , Transfusão de Componentes Sanguíneos/métodos , Transfusão de Componentes Sanguíneos/tendências , Humanos , Pediatria/instrumentação , Pediatria/métodos
15.
Transfus Apher Sci ; 56(6): 906-913, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29162399

RESUMO

Activated Protein C Resistance is mainly associated to a factor V mutation (RQ506), which induces a deficient inactivation of activated factor V by activated protein C, and is associated to an increased risk of venous and arterial thrombosis in affected individuals, caused by the prolonged activated factor V survival. Its prevalence is mainly in Caucasians (about 5%), and this mutation is absent in Africans and Asians. Presence of Factor V-Leiden is usually evidenced with clotting methods, using a two-step APTT assay performed without or with APC: prolongation of blood coagulation time is decreased if this factor is present. The R506Q Factor V-Leiden mutation is now usually characterized using molecular biology, and this technique tends to become the first intention assay for characterization of patients. Both techniques are qualitative, and allow classifying tested individuals as heterozygotes or homozygotes for the mutation, when present. A new quantitative assay for Factor V-Leiden, using a one-step clotting method, has been developed, and designed with highly purified human coagulation proteins. Clotting is triggered with human Factor Xa, in presence of calcium and phospholipids (mixture which favours APC action over clotting process). Diluted tested plasma, is supplemented with a clotting mixture containing human fibrinogen, prothrombin, and protein S at a constant concentration. APC is added, and clotting is initiated with calcium. Calibration is performed with a pool of plasmas from patients carrying the R506Q Factor V mutation, and its mixtures with normal plasma. Homozygous patients have clotting times of about <40sec; heterozygous patients have clotting times of about 40-60sec and normal individuals yield clotting times >70sec. Factor V-Leiden concentration is usually >75% in homozygous patients, 30-60% in heterozygous patients and below 5% in normal. The assay is insensitive to clotting factor deficiencies (II, VII, VIII: C, IX, X), dicoumarol or heparin therapies, and has no interference with lupus anticoagulant (LA). This new assay for Factor V-Leiden can be easily used in any coagulation laboratory, is performed as a single test, and is quantitative. This assay has a high robustness, is accurate and presents a good intra- (<3%) and inter-assay (<5%) variability. It contributes solving most of the laboratory issues faced when testing factor V-Leiden. Quantitation of Factor V-L could contribute to a better assessment of thrombotic risk in affected patients, as this complication is first associated to and caused by the presence of a defined amount of FVa.


Assuntos
Resistência à Proteína C Ativada/metabolismo , Fator V/metabolismo , Trombose/tratamento farmacológico , Humanos , Fatores de Risco
16.
Transfus Apher Sci ; 56(4): 612-625, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28803708

RESUMO

Anti-phospholipid syndrome is a complex and severe clinical situation, associated with symptoms such as recurrent thrombosis, arterial or venous, at any site, pregnancy loss, and other related syndromes. These clinical burdens, are highly variable from patient to patient, and are associated with biological abnormalities, such as the presence of the Lupus Anticoagulant or phospholipid dependent antibodies, confirmed on two occasions at least 12 weeks apart. From the diagnosis standpoint, both, functional (clotting) or immunological assays, are difficult to standardize and to optimize, due to the absence of reference material, or a characteristic clinical group, and international reference preparations. Large cohort studies are necessary for defining the usefulness of each assay, in terms of specificity, sensitivity, accuracy and for following-up the disease evolution. Clotting assays are based on Activated Partial Thromboplastin Time (APTT) and diluted Russell Viper Venom Time (dRVVT), performed at low and high phospholipid concentration, or on 1:1 mixtures of tested sample and a normal plasma pool. They allow evaluation of the paradoxal effects of LAs, which are pro-thrombotic in vivo, and anticoagulant in vivo. Use of synthetic phospholipids improves assay specificities and sensitivities, especially in patients treated with anticoagulants. Immunoassays can also be used for testing phospholipid dependent antibodies, first identified and measured as anti-cardiolipin antibodies, but now characterized as targeted to phospholipid cofactor proteins: mainly ß2GP1 (which exposes cryptic epitopes upon binding to phospholipids), and in some cases prothrombin, and more rarely Protein S, Factor XIII, Protein Z or Annexin V. Use of optimized assays designed with well-characterized anionic phospholipids, then complexed with highly purified phospholipid cofactor protein (mainly ß2GP1), offers a better link between reactivity and clinical associations, than the former assays which were empirically designed with cardiolipin. Standardization also remains complicated due to the absence of international standards and harmonized quantitation units. Validation on large cohorts of negative and positive patients remains the key approach for defining assay performance and clinical usefulness. Laboratory practice for all these methods is now greatly facilitated thanks to the use of automated instruments and dedicated software. Along with clinical criteria, laboratory assays are of great usefulness for identification and confirmation of the anti-phospholipid syndrome and they allow disease follow-up when appropriate patient management is in place.


Assuntos
Síndrome Antifosfolipídica/sangue , Síndrome Antifosfolipídica/diagnóstico , Anexina A5/metabolismo , Anticorpos Anticardiolipina/sangue , Síndrome Antifosfolipídica/terapia , Proteínas Sanguíneas/metabolismo , Fator XIII/metabolismo , Humanos , Inibidor de Coagulação do Lúpus/sangue , Tempo de Tromboplastina Parcial/métodos , Proteína S/metabolismo , Protrombina/metabolismo , Tempo de Protrombina/métodos
17.
Transfus Apher Sci ; 56(1): 91-97, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28089408

RESUMO

The key coagulation factor FVII, and its activated form FVIIa, present a major interest for their role at the initiation phase of blood coagulation, and because they can activate all blood coagulation cascade, through the extrinsic, but also the intrinsic pathway. Blood activation initiated through FVII is first presented, as it is understood nowadays. Measurement of FVII and FVIIa were of main interest for epidemiological studies, but FVIIa contribution to assay results was only deduced. The introduction of specific FVIIa assays, functional or immunoassays, allowed measuring directly FVIIa without any interference of non-activated FVII, or other coagulation factors or their activated forms. The various methods available, and their characteristics are presented, with a special focus on two assays developed by our group for FVIIa (a clotting one and a chromogenic one). The FVIIa clotting assay shows evident superiority for measuring its activity in plasma, in pathophysiological conditions. The normal range is <2.5ng/ml, which represents less than 0.5% of the FVII protein. FVIIa is elevated in some pathological states. The chromogenic assay is of interest for assigning the potency of FVIIa concentrates, as it has a higher dynamic range. Both assays are fully automatable on laboratory instruments, and standardized in a satisfactory manner thanks to the use of the FVIIa concentrate WHO International Standard (NIBSC). The various applications and usefulness of FVIIa laboratory assays are discussed, for the measurement of therapeutic products, or for following recoveries in treated patients, including hemophiliacs with inhibitors, patients with severe bleeding risk (liver diseases, surgery, trauma, …), and lastly for measurement of its activity in therapeutic products.


Assuntos
Coagulação Sanguínea/fisiologia , Fator VIIa/metabolismo , Plasma/química , Fator VIIa/análise , Humanos
18.
Platelets ; 28(3): 235-241, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28102740

RESUMO

Functional and genetic assays for measuring platelet microvesicles (PMVs) are presented and discussed. Functional assays concern two groups of methods: a) homogeneous assays using the cofactor activity of phospholipids (PPLs) contained in PMVs and present in assayed plasmas, and a coagulation or a thrombin generation assay (TGA) as "end points"; b) capture-based assays, in which PMVs bind to an immobilized ligand, such as Annexin V in the presence of calcium, or monoclonal antibodies (MoAbs) specific for membrane proteins. Genetic assays aim to detect micro-RNA (miRNA) present in PMVs: miRNA must be extracted from plasma, and the expression pattern can be determined by various methods such as quantitative real-time PCR, microarray or sequencing. All these technical approaches introduce new exploration tools for measuring or quantitating PMVs or their associated activities, as biomarkers for disease evolution, their diagnosis or prognosis, and for monitoring of some antithrombotic or anti-inflammatory therapies. They offer invaluable analytical tools for research, drug discovery and epidemiological studies and have a strong potential as diagnostic tests.


Assuntos
Bioensaio/normas , Testes de Coagulação Sanguínea/normas , Plaquetas/metabolismo , Vesículas Extracelulares/metabolismo , Trombina/metabolismo , Anexina A5/química , Biomarcadores/metabolismo , Coagulação Sanguínea/fisiologia , Plaquetas/citologia , Comunicação Celular , Endocitose , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática , Vesículas Extracelulares/química , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Ativação Plaquetária , Reação em Cadeia da Polimerase em Tempo Real
19.
Transfus Apher Sci ; 55(1): 10-22, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27522103

RESUMO

Blood cells generate heterogeneous populations of vesicles that are delivered, as small-specialized packages of highly active cell fragments in blood circulation, having almost similar functional activities, as the mother cells. These so called extracellular vesicles are the essential part of an energy-dependent natural apoptotic process; hence their beneficial and harmful biological functions cannot be ignored. Evidence is accumulating, that cellular derived vesicles, originate from all viable cells including: megakaryocytes, platelets, red blood cells, white blood cells and endothelial cells, the highest in proportions from platelets. Shedding can also be triggered by pathological activation of inflammatory processes and activation of coagulation or complement pathways, or even by shear stress in the circulation. Structurally, so called MV/EV appear to be, sometimes inside-out and sometimes outside-in cell fragments having a bilayered phospholipid structure exposing coagulant-active phosphatidylserine, expressing various membrane receptors, and they serve as cell-to-cell shuttles for bioactive molecules such as lipids, growth factors, microRNAs, and mitochondria. Ex vivo processing of blood into its components, embodying centrifugation, processing by various apheresis procedures, leukoreduction, pathogen reduction, and finally storage in different media and different types of blood bags, also have major impacts on the generation and retention of MV content. These artificially generated small, but highly liable packages, together with the original pool of MVs collected from the donor, do exhibit differing biological activities, and are not inert elements and should be considered as a parameter of blood safety in haemovigilance programmes. Harmonization and consensus in sampling protocols, sample handling, processing, and assessment methods, in particular converting to full automation, are needed to achieve consensual interpretations. This review focuses on some of our past personal studies on the role of MV/EV focusing on characterization of platelet storage lesion and platelet therapy that shows the highest transfusion hazards [up to 25%], and loss of 25% platelet efficacy after various leukoreduction and validated platelet pathogen reduction treatments. The planned paths for the future of EV/MV involvement in immunological and viral/ non-viral transfusion hazards are also discussed. Whilst considerable advances made on the characterization of EV/MV, but disparity still exists between various surrogate markers, showing some subtle differences in the levels of MV/ EV & BRMs in platelet preparations, and the clinical outcome showing platelets derived by all current technologies are equivalents in vivo. One possible reason for such a disparity may be relatedto the fact that MVs, being the end products of apoptotic cells, have little specificity and clear rapidly from circulation [<6 h in thrombocytopoenia]. This makes their clinical usefulness rather short lived. The recent findings that pegylating smaller subsets of EV increases its circulatory life from <15 minutes to approximately about one hour is highly promising, in particular, for drug delivery on specific sides. Hence a promising clinical utility of EV/MV continues, as a journey without end, indeed. This manuscript is based mainly on the selected key readings listed below.


Assuntos
Plaquetas/metabolismo , Preservação de Sangue/métodos , Segurança do Sangue/métodos , Biomarcadores/metabolismo , Plaquetas/citologia , Humanos
20.
Transfus Apher Sci ; 55(1): 23-34, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27475803

RESUMO

Utility of EVs, as biomarkers of cause or consequence of various pathological complications, and prognosis of blood components' therapy in terms of safety/efficacy and their potential associated hazards, primed by EVs involvements in pro-inflammatory, immunomodulatory and activations of both pro/anti-coagulatory and others associated pathways, as well as various cellular cross talks, are highlighted as the fundamental. Today EVs are becoming the "buzz" words of the current diagnosis, development and research [DDR] strategies, with the aim of ensuring safer therapeutic approaches in the current clinical practices, also incorporating their potential in long term cost effectiveness in health care systems. The main focus of this manuscript is to review the current opinions in some fundamental areas of EVs involvements in health and diseases. Firstly, our goal is highlighting what are EVs/MVs/MPs and how are they generated in physiology, pathology or blood products; classification and significance of EVs generated in vivo; followed by consequences and physiological/pathological induced effects of EVs generation in vivo. Secondly, specific cell origin EVs and association with malignancy; focus on EVs carrying TF and annexin V as a protective protein for harmful effects of EVs, and associations with LA; and incidence of anti-annexin V antibodies are also discussed. Thirdly, utility of EVs is presented: as diagnostic tools of disease markers; prognosis and follow-up of clinical states; evaluation of therapy efficacy; quality and risk assessment of blood products; followed by the laboratory tools for exploring, characterizing and measuring EVs, and/or their associated activity, using our own experiences of capture based assays. Finally, in perspective, the upcoming low volume sampling, fast, reliable and reproducibility and friendly use laboratory tools and the standardization of measurement methods are highlighted with the beneficial effects that we are witnessing in both wound healing and tissue remodeling, with an expected blockbuster status EVs as future therapeutic directions.


Assuntos
Transfusão de Componentes Sanguíneos/efeitos adversos , Segurança do Sangue/métodos , Micropartículas Derivadas de Células/metabolismo , Biomarcadores/sangue , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA