Coagulation

Last updated

Coagulation
Coagulation in vivo.png
Blood coagulation pathways in vivo showing the central role played by thrombin
HealthBeneficial

Coagulation, also known as clotting, is the process by which blood changes from a liquid to a gel, forming a blood clot. It results in hemostasis, the cessation of blood loss from a damaged vessel, followed by repair. The process of coagulation involves activation, adhesion and aggregation of platelets, as well as deposition and maturation of fibrin.

Contents

Coagulation begins almost instantly after an injury to the endothelium that lines a blood vessel. Exposure of blood to the subendothelial space initiates two processes: changes in platelets, and the exposure of subendothelial platelet tissue factor to coagulation factor VII, which ultimately leads to cross-linked fibrin formation. Platelets immediately form a plug at the site of injury; this is called primary hemostasis. Secondary hemostasis occurs simultaneously: additional coagulation factors beyond factor VII (listed below) respond in a cascade to form fibrin strands, which strengthen the platelet plug. [1]

Coagulation is highly conserved throughout biology. In all mammals, coagulation involves both cellular components (platelets) and proteinaceous components (coagulation or clotting factors). [2] [3] The pathway in humans has been the most extensively researched and is the best understood. [4] Disorders of coagulation can result in problems with hemorrhage, bruising, or thrombosis. [5]

List of coagulation factors

There are 13 traditional clotting factors, as named below, [6] and other substances necessary for coagulation:

Coagulation factors and related substances
Number/NameSynonym(s)FunctionAssociated genetic disordersType of moleculeSourcePathway(s)
Factor I Fibrinogen Forms fibrin threads in blood clotsPlasma proteinLiverCommon pathway; converted into fibrin
Factor II*ProthrombinIts active form (IIa) activates platelets, factors I, V, VII, VIII, XI, XIII, protein C Plasma proteinLiverCommon pathway; converted into thrombin
Factor III
  • Tissue factor
  • tissue thromboplastin
Co-factor of factor VIIa, which was formerly known as factor IIILipoprotein mixtureDamaged cells and plateletsExtrinsic
Factor IV
  • Calcium
  • Calcium ions
  • Ca2+ ions
Required for coagulation factors to bind to phospholipids, which were formerly known as factor IVInorganic ions in plasmaDiet, platelets, bone matrixEntire process of coagulation
Factor V
  • Proaccelerin
  • labile factor
  • Ac-globulin
Co-factor of factor X with which it forms the prothrombinase complex Activated protein C resistance Plasma proteinLiver, plateletsExtrinsic and intrinsic
Factor VI
  • Unassigned
    old name of factor Va
    (activated form of factor V)
  • accelerin (formerly)
N/AN/AN/A
Factor VII*
  • Proconvertin
  • Serum Prothrombin Conversion Accelerator (SPCA)
  • Stable factor
Activates factors IX, X; increases rate of catalytic conversion of prothrombin into thrombinCongenital factor VII deficiency Plasma proteinLiverExtrinsic
Factor VIII
  • Antihemophilic factor A
  • Antihemophilic factor (AHF)
  • Antihemophilic globulin (AHG)
Co-factor of factor IX with which it forms the tenase complex Hemophilia A Plasma protein factorPlatelets and endothelial cellsIntrinsic
Factor IX*
  • Antihemophilic factor B
  • Christmas factor
  • plasma thromboplastin component (PTC)
Activates factor X, forms tenase complex with factor VIII Hemophilia B Plasma proteinLiverIntrinsic
Factor X*
  • Stuart-Prower factor
  • Stuart factor
Activates factor II, forms prothrombinase complex with factor VCongenital Factor X deficiencyProteinLiverExtrinsic and intrinsic
Factor XI
  • Plasma thromboplastin antecedent (PTA)
  • Antihemophilic factor C
Activates factor IX Hemophilia C Plasma proteinLiverIntrinsic
Factor XII Hageman factorActivates XI, VII, prekallikrein and plasminogen Hereditary angioedema type IIIPlasma proteinLiverIntrinsic; initiates clotting in vitro; also activates plasmin
Factor XIII Fibrin-stabilizing factorCrosslinks fibrin threadsCongenital factor XIIIa/b deficiencyPlasma proteinLiver, plateletsCommon pathway; stabilizes fibrin; slows down fibrinolysis
Vitamin K Clotting vitaminEssential factor to the hepatic gamma-glutamyl carboxylase that adds a carboxyl group to glutamic acid residues on factors II, VII, IX and X, as well as Protein S, Protein C and Protein Z [8] Vitamin K deficiency Phytyl-substituted naphthoquinone derivative Gut microbiota
(e.g. E. coli [9] ),
dietary sources
Extrinsic [10]
von Willebrand factor Binds to VIII, mediates platelet adhesion von Willebrand disease Blood glycoproteinBlood vessels' endothelia,
bone marrow [11]
Prekallikrein Fletcher factorActivates XII and prekallikrein; cleaves HMWKPrekallikrein/Fletcher factor deficiency
Kallikrein Activates plasminogen
High-molecular-weight kininogen
  • Fitzgerald factor
  • HMWK
Supports reciprocal activation of factors XII, XI, and prekallikrein Kininogen deficiency
Fibronectin Mediates cell adhesion Glomerulopathy with fibronectin deposits
Antithrombin IIIInhibits factors IIa, Xa, IXa, XIa, and XIIa Antithrombin III deficiency
Heparin cofactor II Inhibits factor IIa, cofactor for heparin and dermatan sulfate ("minor antithrombin")Heparin cofactor II deficiency
Protein C Inactivates factors Va and VIIIa Protein C deficiency
Protein S Cofactor for activated protein C (APC, inactive when bound to C4b-binding protein Protein S deficiency
Protein Z Mediates thrombin adhesion to phospholipids and stimulates degradation of factor X by ZPI Protein Z deficiency
Protein Z-related protease inhibitor ZPIDegrades factors X (in presence of protein Z) and XI (independently
Plasminogen Converts to plasmin, lyses fibrin and other proteinsPlasminogen deficiency type I (ligneous conjunctivitis)
α2-Antiplasmin Inhibits plasminAntiplasmin deficiency
α2-Macroglobulin Inhibits plasmin, kallikrein, and thrombin
Tissue plasminogen activator t-PA or TPAActivates plasminogen
Urokinase Activates plasminogen Quebec platelet disorder
Plasminogen activator inhibitor-1 PAI-1Inactivates tPA and urokinase (endothelial PAIPlasminogen activator inhibitor-1 deficiency
Plasminogen activator inhibitor-2 PAI-2Inactivates tPA and urokinasePlasminogen activator inhibitor-1 deficiency
Cancer procoagulant Pathological activator of factor X; linked to thrombosis in various cancers [12]
* Vitamin K is required for biosynthesis of these clotting factors [8]

Physiology

The interaction of vWF and GP1b alpha. The GP1b receptor on the surface of platelets allows the platelet to bind to vWF, which is exposed upon damage to vasculature. The vWF A1 domain (yellow) interacts with the extracellular domain of GP1ba (blue). VWF-GP1ba.png
The interaction of vWF and GP1b alpha. The GP1b receptor on the surface of platelets allows the platelet to bind to vWF, which is exposed upon damage to vasculature. The vWF A1 domain (yellow) interacts with the extracellular domain of GP1ba (blue).

Physiology of blood coagulation is based on hemostasis, the normal bodily process that stops bleeding. Coagulation is a part of an integrated series of haemostatic reactions, involving plasma, platelet, and vascular components. [13]

Hemostasis consists of four main stages:

After the fibrin clot is formed, clot retraction occurs and then clot resolution starts, and these two process are together called "tertiary hemostasis". Activated platelets contract their internal actin and myosin fibrils in their cytoskeleton, which leads to shrinkage of the clot volume. Plasminogen activators, such as tissue plasminogen activator (t-PA), activate plasminogen into plasmin, which promotes lysis of the fibrin clot; this restores the flow of blood in the damaged/obstructed blood vessels. [22]

Vasoconstriction

When there is an injury to a blood vessel, the endothelial cells can release various vasoconstrictor substances, such as endothelin [23] and thromboxane, [24] to induce the constriction of the smooth muscles in the vessel wall. This helps reduce blood flow to the site of injury and limits bleeding.

Platelet activation and platelet plug formation

When the endothelium is damaged, the normally isolated underlying collagen is exposed to circulating platelets, which bind directly to collagen with collagen-specific glycoprotein Ia/IIa surface receptors. This adhesion is strengthened further by von Willebrand factor (vWF), which is released from the endothelium and from platelets; vWF forms additional links between the platelets' glycoprotein Ib/IX/V and A1 domain. This localization of platelets to the extracellular matrix promotes collagen interaction with platelet glycoprotein VI. Binding of collagen to glycoprotein VI triggers a signaling cascade that results in activation of platelet integrins. Activated integrins mediate tight binding of platelets to the extracellular matrix. This process adheres platelets to the site of injury. [25]

Activated platelets release the contents of stored granules into the blood plasma. The granules include ADP, serotonin, platelet-activating factor (PAF), vWF, platelet factor 4, and thromboxane A2 (TXA2), which, in turn, activate additional platelets. The granules' contents activate a Gq-linked protein receptor cascade, resulting in increased calcium concentration in the platelets' cytosol. The calcium activates protein kinase C, which, in turn, activates phospholipase A2 (PLA2). PLA2 then modifies the integrin membrane glycoprotein IIb/IIIa, increasing its affinity to bind fibrinogen. The activated platelets change shape from spherical to stellate, and the fibrinogen cross-links with glycoprotein IIb/IIIa aid in aggregation of adjacent platelets, forming a platelet plug and thereby completing primary hemostasis). [26]

Coagulation cascade

The classical blood coagulation pathway Classical blood coagulation pathway.png
The classical blood coagulation pathway
Modern coagulation pathway. Hand-drawn composite from similar drawings presented by Professor Dzung Le, MD, PhD, at UCSD Clinical Chemistry conferences on 14 and 21 October 2014. Original schema from Introduction to Hematology by Samuel I. Rapaport. 2nd ed.; Lippencott: 1987. Dr Le added the factor XI portion based on a paper from about year 2000. Dr. Le's similar drawings presented the development of this cascade over 6 frames, like a comic. Rapaport Coagulation Cascade.svg
Modern coagulation pathway. Hand-drawn composite from similar drawings presented by Professor Dzung Le, MD, PhD, at UCSD Clinical Chemistry conferences on 14 and 21 October 2014. Original schema from Introduction to Hematology by Samuel I. Rapaport. 2nd ed.; Lippencott: 1987. Dr Le added the factor XI portion based on a paper from about year 2000. Dr. Le's similar drawings presented the development of this cascade over 6 frames, like a comic.

The coagulation cascade of secondary hemostasis has two initial pathways which lead to fibrin formation. These are the contact activation pathway (also known as the intrinsic pathway), and the tissue factor pathway (also known as the extrinsic pathway), which both lead to the same fundamental reactions that produce fibrin. It was previously thought that the two pathways of coagulation cascade were of equal importance, but it is now known that the primary pathway for the initiation of blood coagulation is the tissue factor (extrinsic) pathway. The pathways are a series of reactions, in which a zymogen (inactive enzyme precursor) of a serine protease and its glycoprotein co-factor are activated to become active components that then catalyze the next reaction in the cascade, ultimately resulting in cross-linked fibrin. Coagulation factors are generally indicated by Roman numerals, with a lowercase a appended to indicate an active form. [27]

The coagulation factors are generally enzymes called serine proteases, which act by cleaving downstream proteins. The exceptions are tissue factor, FV, FVIII, FXIII. [28] Tissue factor, FV and FVIII are glycoproteins, and Factor XIII is a transglutaminase. [27] The coagulation factors circulate as inactive zymogens. The coagulation cascade is therefore classically divided into three pathways. The tissue factor and contact activation pathways both activate the "final common pathway" of factor X, thrombin and fibrin. [29]

Tissue factor pathway (extrinsic)

The main role of the tissue factor (TF) pathway is to generate a "thrombin burst", a process by which thrombin, the most important constituent of the coagulation cascade in terms of its feedback activation roles, is released very rapidly. FVIIa circulates in a higher amount than any other activated coagulation factor. The process includes the following steps: [27]

  1. Following damage to the blood vessel, FVII leaves the circulation and comes into contact with tissue factor expressed on tissue-factor-bearing cells (stromal fibroblasts and leukocytes), forming an activated complex (TF-FVIIa).
  2. TF-FVIIa activates FIX and FX.
  3. FVII is itself activated by thrombin, FXIa, FXII, and FXa.
  4. The activation of FX (to form FXa) by TF-FVIIa is almost immediately inhibited by tissue factor pathway inhibitor (TFPI).
  5. FXa and its co-factor FVa form the prothrombinase complex, which activates prothrombin to thrombin.
  6. Thrombin then activates other components of the coagulation cascade, including FV and FVIII (which forms a complex with FIX), and activates and releases FVIII from being bound to vWF.
  7. FVIIIa is the co-factor of FIXa, and together they form the "tenase" complex, which activates FX; and so the cycle continues. ("Tenase" is a contraction of "ten" and the suffix "-ase" used for enzymes.)

Contact activation pathway (intrinsic)

The contact activation pathway begins with formation of the primary complex on collagen by high-molecular-weight kininogen (HMWK), prekallikrein, and FXII (Hageman factor). Prekallikrein is converted to kallikrein and FXII becomes FXIIa. FXIIa converts FXI into FXIa. Factor XIa activates FIX, which with its co-factor FVIIIa form the tenase complex, which activates FX to FXa. The minor role that the contact activation pathway has in initiating blood clot formation can be illustrated by the fact that individuals with severe deficiencies of FXII, HMWK, and prekallikrein do not have a bleeding disorder. Instead, contact activation system seems to be more involved in inflammation, [27] and innate immunity. [30] Despite this, interference with the pathway may confer protection against thrombosis without a significant bleeding risk. [30]

Final common pathway

The division of coagulation in two pathways is arbitrary, originating from laboratory tests in which clotting times were measured either after the clotting was initiated by glass, the intrinsic pathway; or clotting was initiated by thromboplastin (a mix of tissue factor and phospholipids), the extrinsic pathway.[ citation needed ]

Further, the final common pathway scheme implies that prothrombin is converted to thrombin only when acted upon by the intrinsic or extrinsic pathways, which is an oversimplification. In fact, thrombin is generated by activated platelets at the initiation of the platelet plug, which in turn promotes more platelet activation.[ citation needed ]

Thrombin functions not only to convert fibrinogen to fibrin, it also activates Factors VIII and V and their inhibitor protein C (in the presence of thrombomodulin). By activating Factor XIII, covalent bonds are formed that crosslink the fibrin polymers that form from activated monomers. [27] This stabilizes the fibrin network.[ citation needed ]

The coagulation cascade is maintained in a prothrombotic state by the continued activation of FVIII and FIX to form the tenase complex until it is down-regulated by the anticoagulant pathways. [27]

Cell-based scheme of coagulation

A newer model of coagulation mechanism explains the intricate combination of cellular and biochemical events that occur during the coagulation process in vivo . Along with the procoagulant and anticoagulant plasma proteins, normal physiologic coagulation requires the presence of two cell types for formation of coagulation complexes: cells that express tissue factor (usually extravascular) and platelets.[ citation needed ]

The coagulation process occurs in two phases. First is the initiation phase, which occurs in tissue-factor-expressing cells. This is followed by the propagation phase, which occurs on activated platelets. The initiation phase, mediated by the tissue factor exposure, proceeds via the classic extrinsic pathway and contributes to about 5% of thrombin production. The amplified production of thrombin occurs via the classic intrinsic pathway in the propagation phase; about 95% of thrombin generated will be during this second phase. [31]

Fibrin clot formation

Fibrinolysis

Eventually, blood clots are reorganized and resorbed by a process termed fibrinolysis . The main enzyme responsible for this process is plasmin, which is regulated by plasmin activators and plasmin inhibitors. [32]

Role in immune system

The coagulation system overlaps with the immune system. Coagulation can physically trap invading microbes in blood clots. Also, some products of the coagulation system can contribute to the innate immune system by their ability to increase vascular permeability and act as chemotactic agents for phagocytic cells. In addition, some of the products of the coagulation system are directly antimicrobial. For example, beta-lysine, an amino acid produced by platelets during coagulation, can cause lysis of many Gram-positive bacteria by acting as a cationic detergent. [33] Many acute-phase proteins of inflammation are involved in the coagulation system. In addition, pathogenic bacteria may secrete agents that alter the coagulation system, e.g. coagulase and streptokinase.[ citation needed ]

Cofactors

Various substances are required for the proper functioning of the coagulation cascade:

Calcium and phospholipids

Calcium and phospholipids (constituents of platelet membrane) are required for the tenase and prothrombinase complexes to function. [34] Calcium mediates the binding of the complexes via the terminal gamma-carboxy residues on Factor Xa and Factor IXa to the phospholipid surfaces expressed by platelets, as well as procoagulant microparticles or microvesicles shed from them. [35] Calcium is also required at other points in the coagulation cascade. Calcium ions play a major role in the regulation of coagulation cascade that is paramount in the maintenance of hemostasis. Other than platelet activation, calcium ions are responsible for complete activation of several coagulation factors, including coagulation Factor XIII. [36]

Vitamin K

Vitamin K is an essential factor to the hepatic gamma-glutamyl carboxylase that adds a carboxyl group to glutamic acid residues on factors II, VII, IX and X, as well as Protein S, Protein C and Protein Z. In adding the gamma-carboxyl group to glutamate residues on the immature clotting factors, Vitamin K is itself oxidized. Another enzyme, Vitamin K epoxide reductase (VKORC), reduces vitamin K back to its active form. Vitamin K epoxide reductase is pharmacologically important as a target of anticoagulant drugs warfarin and related coumarins such as acenocoumarol, phenprocoumon, and dicumarol. These drugs create a deficiency of reduced vitamin K by blocking VKORC, thereby inhibiting maturation of clotting factors. Vitamin K deficiency from other causes (e.g., in malabsorption) or impaired vitamin K metabolism in disease (e.g., in liver failure) lead to the formation of PIVKAs (proteins formed in vitamin K absence), which are partially or totally non-gamma carboxylated, affecting the coagulation factors' ability to bind to phospholipid.[ citation needed ]

Regulators

Coagulation with arrows for negative and positive feedback. Coagulation full.svg
Coagulation with arrows for negative and positive feedback.

Five mechanisms keep platelet activation and the coagulation cascade in check.[ citation needed ] Abnormalities can lead to an increased tendency toward thrombosis:

Protein C

Protein C is a major physiological anticoagulant. It is a vitamin K-dependent serine protease enzyme that is activated by thrombin into activated protein C (APC). Protein C is activated in a sequence that starts with Protein C and thrombin binding to a cell surface protein thrombomodulin. Thrombomodulin binds these proteins in such a way that it activates Protein C. The activated form, along with protein S and a phospholipid as cofactors, degrades FVa and FVIIIa. Quantitative or qualitative deficiency of either (protein C or protein S) may lead to thrombophilia (a tendency to develop thrombosis). Impaired action of Protein C (activated Protein C resistance), for example by having the "Leiden" variant of Factor V or high levels of FVIII, also may lead to a thrombotic tendency.[ citation needed ]

Antithrombin

Antithrombin is a serine protease inhibitor (serpin) that degrades the serine proteases: thrombin, FIXa, FXa, FXIa, and FXIIa. It is constantly active, but its adhesion to these factors is increased by the presence of heparan sulfate (a glycosaminoglycan) or the administration of heparins (different heparinoids increase affinity to FXa, thrombin, or both). Quantitative or qualitative deficiency of antithrombin (inborn or acquired, e.g., in proteinuria) leads to thrombophilia.[ citation needed ]

Tissue factor pathway inhibitor (TFPI)

Tissue factor pathway inhibitor (TFPI) limits the action of tissue factor (TF). It also inhibits excessive TF-mediated activation of FVII and FX.[ citation needed ]

Plasmin

Plasmin is generated by proteolytic cleavage of plasminogen, a plasma protein synthesized in the liver. This cleavage is catalyzed by tissue plasminogen activator (t-PA), which is synthesized and secreted by endothelium. Plasmin proteolytically cleaves fibrin into fibrin degradation products that inhibit excessive fibrin formation.[ citation needed ]

Prostacyclin

Prostacyclin (PGI2) is released by endothelium and activates platelet Gs protein-linked receptors. This, in turn, activates adenylyl cyclase, which synthesizes cAMP. cAMP inhibits platelet activation by decreasing cytosolic levels of calcium and, by doing so, inhibits the release of granules that would lead to activation of additional platelets and the coagulation cascade. [32]

Medical assessment

Numerous medical tests are used to assess the function of the coagulation system: [3] [37]

The contact activation (intrinsic) pathway is initiated by activation of the "contact factors" of plasma, and can be measured by the activated partial thromboplastin time (aPTT) test.[ citation needed ]

The tissue factor (extrinsic) pathway is initiated by release of tissue factor (a specific cellular lipoprotein), and can be measured by the prothrombin time (PT) test. [39] PT results are often reported as ratio (INR value) to monitor dosing of oral anticoagulants such as warfarin.[ citation needed ]

The quantitative and qualitative screening of fibrinogen is measured by the thrombin clotting time (TCT). Measurement of the exact amount of fibrinogen present in the blood is generally done using the Clauss fibrinogen assay. [38] Many analysers are capable of measuring a "derived fibrinogen" level from the graph of the Prothrombin time clot.

If a coagulation factor is part of the contact activation or tissue factor pathway, a deficiency of that factor will affect only one of the tests: Thus hemophilia A, a deficiency of factor VIII, which is part of the contact activation pathway, results in an abnormally prolonged aPTT test but a normal PT test. The exceptions are prothrombin, fibrinogen, and some variants of FX that can be detected only by either aPTT or PT. If an abnormal PT or aPTT is present, additional testing will occur to determine which (if any) factor is present as aberrant concentrations.

Deficiencies of fibrinogen (quantitative or qualitative) will affect all screening tests.

Role in disease

Coagulation defects may cause hemorrhage or thrombosis, and occasionally both, depending on the nature of the defect. [40]

The GP1b-IX receptor complex. This protein receptor complex is found on the surface of platelets, and in conjunction with GPV allows for platelets to adhere to the site of injury. Mutations in the genes associated with the glycoprotein Ib-IX-V complex are characteristic of Bernard-Soulier syndrome. Gp1brec3.png
The GP1b-IX receptor complex. This protein receptor complex is found on the surface of platelets, and in conjunction with GPV allows for platelets to adhere to the site of injury. Mutations in the genes associated with the glycoprotein Ib-IX-V complex are characteristic of Bernard–Soulier syndrome.

Platelet disorders

Platelet disorders are either congenital or acquired. Examples of congenital platelet disorders are Glanzmann's thrombasthenia, Bernard–Soulier syndrome (abnormal glycoprotein Ib-IX-V complex), gray platelet syndrome (deficient alpha granules), and delta storage pool deficiency (deficient dense granules). Most are rare. They predispose to hemorrhage. Von Willebrand disease is due to deficiency or abnormal function of von Willebrand factor, and leads to a similar bleeding pattern; its milder forms are relatively common.[ citation needed ]

Decreased platelet numbers (thrombocytopenia) is due to insufficient production (e.g., myelodysplastic syndrome or other bone marrow disorders), destruction by the immune system (immune thrombocytopenic purpura), or consumption (e.g., thrombotic thrombocytopenic purpura, hemolytic-uremic syndrome, paroxysmal nocturnal hemoglobinuria, disseminated intravascular coagulation, heparin-induced thrombocytopenia). [41] An increase in platelet count leading to elevated risk of thrombosis is called thrombocytosis, which may lead to formation of thromboembolisms.[ citation needed ]

Coagulation factor disorders

The best-known coagulation factor disorders are the hemophilias. The three main forms are hemophilia A (factor VIII deficiency), hemophilia B (factor IX deficiency or "Christmas disease") and hemophilia C (factor XI deficiency, mild bleeding tendency).[ medical citation needed ]

Von Willebrand disease (which behaves more like a platelet disorder except in severe cases), is the most common hereditary bleeding disorder and is characterized as being inherited autosomal recessive or dominant. In this disease, there is a defect in von Willebrand factor (vWF), which mediates the binding of glycoprotein Ib (GPIb) to collagen. This binding helps mediate the activation of platelets and formation of primary hemostasis.[ medical citation needed ]

In acute or chronic liver failure, there is insufficient production of coagulation factors, possibly increasing risk of bleeding during surgery. [42]

Thrombosis is the pathological development of blood clots. These clots may break free and become mobile, forming an embolus or grow to such a size that occludes the vessel in which it developed. An embolism is said to occur when the thrombus (blood clot) becomes a mobile embolus and migrates to another part of the body, interfering with blood circulation and hence impairing organ function downstream of the occlusion. This causes ischemia and often leads to ischemic necrosis of tissue. Most cases of venous thrombosis are due to acquired states (older age, surgery, cancer, immobility) or inherited thrombophilias (e.g., antiphospholipid syndrome, factor V Leiden, and various other genetic deficiencies or variants).[ medical citation needed ]

Pharmacology

Procoagulants

The use of adsorbent chemicals, such as zeolites, and other hemostatic agents are also used for sealing severe injuries quickly (such as in traumatic bleeding secondary to gunshot wounds). Thrombin and fibrin glue are used surgically to treat bleeding and to thrombose aneurysms. Hemostatic Powder Spray TC-325 is used to treated gastrointestinal bleeding.[ citation needed ]

Desmopressin is used to improve platelet function by activating arginine vasopressin receptor 1A. [43]

Coagulation factor concentrates are used to treat hemophilia, to reverse the effects of anticoagulants, and to treat bleeding in people with impaired coagulation factor synthesis or increased consumption. Prothrombin complex concentrate, cryoprecipitate and fresh frozen plasma are commonly used coagulation factor products. Recombinant activated human factor VII is increasingly popular in the treatment of major bleeding.

Tranexamic acid and aminocaproic acid inhibit fibrinolysis and lead to a de facto reduced bleeding rate. Before its withdrawal, aprotinin was used in some forms of major surgery to decrease bleeding risk and the need for blood products.

Rivaroxaban drug bound to the coagulation factor Xa. The drug prevents this protein from activating the coagulation pathway by inhibiting its enzymatic activity. FXa-rivaroxaban.png
Rivaroxaban drug bound to the coagulation factor Xa. The drug prevents this protein from activating the coagulation pathway by inhibiting its enzymatic activity.

Anticoagulants

Anticoagulants and anti-platelet agents (together "antithrombotics") are amongst the most commonly used medications. Anti-platelet agents include aspirin, dipyridamole, ticlopidine, clopidogrel, ticagrelor and prasugrel; the parenteral glycoprotein IIb/IIIa inhibitors are used during angioplasty. Of the anticoagulants, warfarin (and related coumarins) and heparin are the most commonly used. Warfarin affects the vitamin K-dependent clotting factors (II, VII, IX, X) and protein C and protein S, whereas heparin and related compounds increase the action of antithrombin on thrombin and factor Xa. A newer class of drugs, the direct thrombin inhibitors, is under development; some members are already in clinical use (such as lepirudin). Also in clinical use are other small molecular compounds that interfere directly with the enzymatic action of particular coagulation factors (the directly acting oral anticoagulants: dabigatran, rivaroxaban, apixaban, and edoxaban). [44]

History

Initial discoveries

Theories on the coagulation of blood have existed since antiquity. Physiologist Johannes Müller (1801–1858) described fibrin, the substance of a thrombus. Its soluble precursor, fibrinogen, was thus named by Rudolf Virchow (1821–1902), and isolated chemically by Prosper Sylvain Denis (1799–1863). Alexander Schmidt suggested that the conversion from fibrinogen to fibrin is the result of an enzymatic process, and labeled the hypothetical enzyme "thrombin" and its precursor "prothrombin". [45] [46] Arthus discovered in 1890 that calcium was essential in coagulation. [47] [48] Platelets were identified in 1865, and their function was elucidated by Giulio Bizzozero in 1882. [49]

The theory that thrombin is generated by the presence of tissue factor was consolidated by Paul Morawitz in 1905. [50] At this stage, it was known that thrombokinase/thromboplastin (factor III) is released by damaged tissues, reacting with prothrombin (II), which, together with calcium (IV), forms thrombin, which converts fibrinogen into fibrin (I). [51]

Coagulation factors

The remainder of the biochemical factors in the process of coagulation were largely discovered in the 20th century.[ citation needed ]

A first clue as to the actual complexity of the system of coagulation was the discovery of proaccelerin (initially and later called Factor V) by Paul Owren  [ no ] (1905–1990) in 1947. He also postulated its function to be the generation of accelerin (Factor VI), which later turned out to be the activated form of V (or Va); hence, VI is not now in active use. [51]

Factor VII (also known as serum prothrombin conversion accelerator or proconvertin, precipitated by barium sulfate) was discovered in a young female patient in 1949 and 1951 by different groups.

Factor VIII turned out to be deficient in the clinically recognized but etiologically elusive hemophilia A; it was identified in the 1950s and is alternatively called antihemophilic globulin due to its capability to correct hemophilia A. [51]

Factor IX was discovered in 1952 in a young patient with hemophilia B named Stephen Christmas (1947–1993). His deficiency was described by Dr. Rosemary Biggs and Professor R.G. MacFarlane in Oxford, UK. The factor is, hence, called Christmas Factor. Christmas lived in Canada and campaigned for blood transfusion safety until succumbing to transfusion-related AIDS at age 46. An alternative name for the factor is plasma thromboplastin component, given by an independent group in California. [51]

Hageman factor, now known as factor XII, was identified in 1955 in an asymptomatic patient with a prolonged bleeding time named of John Hageman. Factor X, or Stuart-Prower factor, followed, in 1956. This protein was identified in a Ms. Audrey Prower of London, who had a lifelong bleeding tendency. In 1957, an American group identified the same factor in a Mr. Rufus Stuart. Factors XI and XIII were identified in 1953 and 1961, respectively. [51]

The view that the coagulation process is a "cascade" or "waterfall" was enunciated almost simultaneously by MacFarlane [52] in the UK and by Davie and Ratnoff [53] in the US, respectively.

Nomenclature

The usage of Roman numerals rather than eponyms or systematic names was agreed upon during annual conferences (starting in 1955) of hemostasis experts. In 1962, consensus was achieved on the numbering of factors I–XII. [54] This committee evolved into the present-day International Committee on Thrombosis and Hemostasis (ICTH). Assignment of numerals ceased in 1963 after the naming of Factor XIII. The names Fletcher Factor and Fitzgerald Factor were given to further coagulation-related proteins, namely prekallikrein and high-molecular-weight kininogen, respectively. [51]

Factor VI[ citation needed ] is unassigned, as accelerin was found to be activated Factor V.

Other species

All mammals have an extremely closely related blood coagulation process, using a combined cellular and serine protease process.[ citation needed ] In fact, it is possible for any mammalian coagulation factor to "cleave" its equivalent target in any other mammal.[ citation needed ] The only non-mammalian animal known to use serine proteases for blood coagulation is the horseshoe crab. [55]

See also

Related Research Articles

<span class="mw-page-title-main">Thrombus</span> Blood clot

A thrombus, colloquially called a blood clot, is the final product of the blood coagulation step in hemostasis. There are two components to a thrombus: aggregated platelets and red blood cells that form a plug, and a mesh of cross-linked fibrin protein. The substance making up a thrombus is sometimes called cruor. A thrombus is a healthy response to injury intended to stop and prevent further bleeding, but can be harmful in thrombosis, when a clot obstructs blood flow through healthy blood vessels in the circulatory system.

<span class="mw-page-title-main">Platelet</span> Component of blood aiding in coagulation

Platelets or thrombocytes are a component of blood whose function is to react to bleeding from blood vessel injury by clumping, thereby initiating a blood clot. Platelets have no cell nucleus; they are fragments of cytoplasm derived from the megakaryocytes of the bone marrow or lung, which then enter the circulation. Platelets are found only in mammals, whereas in other vertebrates, thrombocytes circulate as intact mononuclear cells.

<span class="mw-page-title-main">Disseminated intravascular coagulation</span> Medical condition where blood clots block small blood vessels

Disseminated intravascular coagulation (DIC) is a condition in which blood clots form throughout the body, blocking small blood vessels. Symptoms may include chest pain, shortness of breath, leg pain, problems speaking, or problems moving parts of the body. As clotting factors and platelets are used up, bleeding may occur. This may include blood in the urine, blood in the stool, or bleeding into the skin. Complications may include organ failure.

<span class="mw-page-title-main">Fibrinogen</span> Soluble protein complex in blood plasma and involved in clot formation

Fibrinogen is a glycoprotein complex, produced in the liver, that circulates in the blood of all vertebrates. During tissue and vascular injury, it is converted enzymatically by thrombin to fibrin and then to a fibrin-based blood clot. Fibrin clots function primarily to occlude blood vessels to stop bleeding. Fibrin also binds and reduces the activity of thrombin. This activity, sometimes referred to as antithrombin I, limits clotting. Fibrin also mediates blood platelet and endothelial cell spreading, tissue fibroblast proliferation, capillary tube formation, and angiogenesis and thereby promotes revascularization and wound healing.

<span class="mw-page-title-main">Thrombin</span> Enzyme involved in blood coagulation in humans

Thrombin is a serine protease, an enzyme that, in humans, is encoded by the F2 gene.

In biology, hemostasis or haemostasis is a process to prevent and stop bleeding, meaning to keep blood within a damaged blood vessel. It is the first stage of wound healing. Hemostasis involves three major steps:

<span class="mw-page-title-main">Bleeding diathesis</span> Medical condition

In medicine (hematology), bleeding diathesis is an unusual susceptibility to bleed (hemorrhage) mostly due to hypocoagulability, in turn caused by a coagulopathy. Therefore, this may result in the reduction of platelets being produced and leads to excessive bleeding. Several types of coagulopathy are distinguished, ranging from mild to lethal. Coagulopathy can be caused by thinning of the skin, such that the skin is weakened and is bruised easily and frequently without any trauma or injury to the body. Also, coagulopathy can be contributed by impaired wound healing or impaired clot formation.

<span class="mw-page-title-main">Partial thromboplastin time</span> Test for coagulation of blood

The partial thromboplastin time (PTT), also known as the activated partial thromboplastin time, is a blood test that characterizes coagulation of the blood. A historical name for this measure is the kaolin-cephalin clotting time (KCCT), reflecting kaolin and cephalin as materials historically used in the test. Apart from detecting abnormalities in blood clotting, partial thromboplastin time is also used to monitor the treatment effect of heparin, a widely prescribed drug that reduces blood's tendency to clot.

<span class="mw-page-title-main">Coagulation factor VII</span> Mammalian protein found in humans

Coagulation factor VII is one of the clotting factors in the coagulation cascade, and in humans is coded for by the gene F7. It is an enzyme of the serine protease class. Once bound to tissue factor released from damaged tissues, it is converted to factor VIIa, which in turn activates factor IX and factor X.

<span class="mw-page-title-main">Factor XIII</span> Class of enzymes

Factor XIII or fibrin stabilizing factor is a zymogen found in blood of humans and some other animals. It is activated by thrombin to factor XIIIa. Factor XIIIa is an enzyme of the blood coagulation system that crosslinks fibrin. Deficiency of XIII worsens clot stability and increases bleeding tendency.

<span class="mw-page-title-main">Thrombophilia</span> Abnormality of blood coagulation

Thrombophilia is an abnormality of blood coagulation that increases the risk of thrombosis. Such abnormalities can be identified in 50% of people who have an episode of thrombosis that was not provoked by other causes. A significant proportion of the population has a detectable thrombophilic abnormality, but most of these develop thrombosis only in the presence of an additional risk factor.

<span class="mw-page-title-main">Factor X</span> Mammalian protein found in Homo sapiens

Factor X, also known by the eponym Stuart–Prower factor, is an enzyme of the coagulation cascade. It is a serine endopeptidase. Factor X is synthesized in the liver and requires vitamin K for its synthesis.

An Error has occurred retrieving Wikidata item for infobox Coagulation factor V is a protein of the coagulation system, encoded for in the human by the F5 gene. It is rarely referred to as proaccelerin or labile factor. In contrast to most other coagulation factors, it is not enzymatically active but functions as a cofactor. Deficiency leads to predisposition for hemorrhage, while some mutations predispose for thrombosis.

<span class="mw-page-title-main">Hypoprothrombinemia</span> Medical condition

Hypoprothrombinemia is a rare blood disorder in which a deficiency in immunoreactive prothrombin, produced in the liver, results in an impaired blood clotting reaction, leading to an increased physiological risk for spontaneous bleeding. This condition can be observed in the gastrointestinal system, cranial vault, and superficial integumentary system, affecting both the male and female population. Prothrombin is a critical protein that is involved in the process of hemostasis, as well as illustrating procoagulant activities. This condition is characterized as an autosomal recessive inheritance congenital coagulation disorder affecting 1 per 2,000,000 of the population, worldwide, but is also attributed as acquired.

<span class="mw-page-title-main">Thrombin time</span>

The thrombin time (TT), also known as the thrombin clotting time (TCT), is a blood test that measures the time it takes for a clot to form in the plasma of a blood sample containing anticoagulant, after an excess of thrombin has been added. It is used to diagnose blood coagulation disorders and to assess the effectiveness of fibrinolytic therapy. This test is repeated with pooled plasma from normal patients. The difference in time between the test and the 'normal' indicates an abnormality in the conversion of fibrinogen to fibrin, an insoluble protein.

The dysfibrinogenemias consist of three types of fibrinogen disorders in which a critical blood clotting factor, fibrinogen, circulates at normal levels but is dysfunctional. Congenital dysfibrinogenemia is an inherited disorder in which one of the parental genes produces an abnormal fibrinogen. This fibrinogen interferes with normal blood clotting and/or lysis of blood clots. The condition therefore may cause pathological bleeding and/or thrombosis. Acquired dysfibrinogenemia is a non-hereditary disorder in which fibrinogen is dysfunctional due to the presence of liver disease, autoimmune disease, a plasma cell dyscrasias, or certain cancers. It is associated primarily with pathological bleeding. Hereditary fibrinogen Aα-Chain amyloidosis is a sub-category of congenital dysfibrinogenemia in which the dysfunctional fibrinogen does not cause bleeding or thrombosis but rather gradually accumulates in, and disrupts the function of, the kidney.

The fibrinolysis system is responsible for removing blood clots. Hyperfibrinolysis describes a situation with markedly enhanced fibrinolytic activity, resulting in increased, sometimes catastrophic bleeding. Hyperfibrinolysis can be caused by acquired or congenital reasons. Among the congenital conditions for hyperfibrinolysis, deficiency of alpha-2-antiplasmin or plasminogen activator inhibitor type 1 (PAI-1) are very rare. The affected individuals show a hemophilia-like bleeding phenotype. Acquired hyperfibrinolysis is found in liver disease, in patients with severe trauma, during major surgical procedures, and other conditions. A special situation with temporarily enhanced fibrinolysis is thrombolytic therapy with drugs which activate plasminogen, e.g. for use in acute ischemic events or in patients with stroke. In patients with severe trauma, hyperfibrinolysis is associated with poor outcome. Moreover, hyperfibrinolysis may be associated with blood brain barrier impairment, a plasmin-dependent effect due to an increased generation of bradykinin.

Thromboelastometry (TEM), previously named rotational thromboelastography (ROTEG) or rotational thromboelastometry (ROTEM), is an established viscoelastic method for hemostasis testing in whole blood. It is a modification of traditional thromboelastography (TEG).

Blood clotting tests are the tests used for diagnostics of the hemostasis system. Coagulometer is the medical laboratory analyzer used for testing of the hemostasis system. Modern coagulometers realize different methods of activation and observation of development of blood clots in blood or in blood plasma.

The platelet plug, also known as the hemostatic plug or platelet thrombus, is an aggregation of platelets formed during early stages of hemostasis in response to one or more injuries to blood vessel walls. After platelets are recruited and begin to accumulate around the breakage, their “sticky” nature allows them to adhere to each other. This forms a platelet plug, which prevents more blood from leaving the body as well as any outside contaminants from getting in. The plug provides a temporary blockage of the break in the vasculature. As such, platelet plug formation occurs after vasoconstriction of the blood vessels but before the creation of the fibrin mesh clot, which is the more permanent solution to the injury. The result of the platelet plug formation is the coagulation of blood. It can also be referred to as primary hemostasis.

References

  1. Furie, Barbara C.; Furie, Bruce (December 2005). "Thrombus formation in vivo". The Journal of Clinical Investigation. 115 (12): 3355–3362. doi:10.1172/JCI26987. PMC   1297262 . PMID   16322780.
  2. Michelson, Alan D. (2006). Platelets (2nd ed.). Elsevier. pp. 3–5. ISBN   978-0-08-046586-9. OCLC   909782638. Archived from the original on 10 May 2017. Retrieved 8 February 2022.
  3. 1 2 "Coagulation Factor Tests: MedlinePlus Medical Test". medlineplus.gov. Retrieved 27 April 2024.
  4. Schmaier, Alvin H.; Lazarus, Hillard M. (2011). Concise guide to hematology. Chichester, West Sussex, UK: Wiley-Blackwell. p. 91. ISBN   978-1-4051-9666-6. OCLC   779160978. Archived from the original on 8 February 2022. Retrieved 8 February 2022.
  5. Lillicrap, D.; Key, Nigel; Makris, Michael; Denise, O'Shaughnessy (2009). Practical Hemostasis and Thrombosis . Wiley-Blackwell. pp. 1–5. ISBN   978-1-4051-8460-1.
  6. Creative Commons by small.svg  This article incorporates text available under the CC BY 4.0 license.Betts, J Gordon; Desaix, Peter; Johnson, Eddie; Johnson, Jody E; Korol, Oksana; Kruse, Dean; Poe, Brandon; Wise, James; Womble, Mark D; Young, Kelly A (28 July 2023). Anatomy & Physiology. Houston: OpenStax CNX. 18.5 Homeostasis. ISBN   978-1-947172-04-3.
  7. "Prothrombin thrombophilia". MedlinePlus. Archived from the original on 12 September 2023. Retrieved 11 September 2023.
  8. 1 2 "Gamma-Glutamyl Carboxylase - an overview | ScienceDirect Topics". ScienceDirect. 9 September 2023. Archived from the original on 9 September 2023. Retrieved 9 September 2023.
  9. Blount, Zachary D. (25 March 2015). "The unexhausted potential of E. coli". eLife. 4: e05826. doi: 10.7554/eLife.05826 . ISSN   2050-084X. PMC   4373459 . PMID   25807083.
  10. "Coagulation Cascade: What Is It, Steps, and More | Osmosis". www.osmosis.org. Archived from the original on 8 September 2023. Retrieved 8 September 2023.
  11. "VWF gene: MedlinePlus Genetics". medlineplus.gov. Archived from the original on 11 May 2023. Retrieved 8 September 2023.
  12. Gordon, S. G.; Mielicki, W. P. (March 1997). "Cancer procoagulant: a factor X activator, tumor marker and growth factor from malignant tissue". Blood Coagulation & Fibrinolysis. 8 (2): 73–86. doi:10.1097/00001721-199703000-00001. ISSN   0957-5235. PMID   9518049.
  13. Bloom, A. L. (1990). "Physiology of blood coagulation". Haemostasis. 20 (Suppl 1): 14–29. doi:10.1159/000216159. ISSN   0301-0147. PMID   2083865. Archived from the original on 2 August 2022. Retrieved 15 October 2023.
  14. "Chapter 23 - Inflammation and its mediators". Rheumatology (6th edition) | Thromboxane A2 - an overview. Archived from the original on 9 September 2023. Retrieved 15 October 2023 via ScienceDirect.
  15. Charo, I. F.; Bekeart, L. S.; Phillips, D. R. (25 July 1987). "Platelet glycoprotein IIb-IIIa-like proteins mediate endothelial cell attachment to adhesive proteins and the extracellular matrix". The Journal of Biological Chemistry. 262 (21): 9935–9938. doi: 10.1016/S0021-9258(18)61053-1 . ISSN   0021-9258. PMID   2440865. Archived from the original on 15 October 2023.
  16. Watson, Steve P. (1 April 2009). "Platelet Activation by Extracellular Matrix Proteins in Haemostasis and Thrombosis". Current Pharmaceutical Design. 15 (12): 1358–1372. doi:10.2174/138161209787846702. PMID   19355974.
  17. Wagner, D. D.; Urban-Pickering, M.; Marder, V. J. (January 1984). "Von Willebrand protein binds to extracellular matrices independently of collagen". Proceedings of the National Academy of Sciences of the United States of America. 81 (2): 471–475. Bibcode:1984PNAS...81..471W. doi: 10.1073/pnas.81.2.471 . ISSN   0027-8424. PMC   344699 . PMID   6320190.
  18. Vermylen, Jos; Verstraete, Marc; Fuster, Valentin (1 December 1986). "Role of platelet activation and fibrin formation in thrombogenesis". Journal of the American College of Cardiology. Symposium on Thrombosis and Antithrombotic Therapy—1986. 8 (6, Supplement 2): 2B–9B. doi: 10.1016/S0735-1097(86)80002-X . ISSN   0735-1097. PMID   3537069. S2CID   23789418.
  19. Blanchette, V. S.; Brandão, L. R.; Breakey, V. R.; Revel-Vilk, S. (22 December 2016). "Primary and secondary hemostasis, regulators of coagulation, and fibrinolysis: Understanding the basics". SickKids Handbook of Pediatric Thrombosis and Hemostasis. Karger Medical and Scientific Publishers. ISBN   978-3-318-03026-6.
  20. "Coagulation Cascade: What Is It, Steps, and More". www.osmosis.org. Archived from the original on 8 September 2023. Retrieved 15 October 2023.
  21. 1 2 Weisel, John W.; Litvinov, Rustem I. (2017). "Fibrin Formation, Structure and Properties". Fibrous Proteins: Structures and Mechanisms. Subcellular Biochemistry. Vol. 82. pp. 405–456. doi:10.1007/978-3-319-49674-0_13. ISBN   978-3-319-49672-6. ISSN   0306-0225. PMC   5536120 . PMID   28101869.
  22. LaPelusa, Andrew; Dave, Heeransh D. (2023), "Physiology, Hemostasis", StatPearls, Treasure Island (FL): StatPearls Publishing, PMID   31424847, archived from the original on 12 March 2023, retrieved 15 October 2023
  23. Loscalzo, J. (1995). "Endothelial injury, vasoconstriction, and its prevention". Texas Heart Institute Journal. 22 (2): 180–184. ISSN   0730-2347. PMC   325239 . PMID   7647603.
  24. Yau, Jonathan W.; Teoh, Hwee; Verma, Subodh (19 October 2015). "Endothelial cell control of thrombosis". BMC Cardiovascular Disorders. 15: 130. doi: 10.1186/s12872-015-0124-z . ISSN   1471-2261. PMC   4617895 . PMID   26481314.
  25. Nigel Key; Michael Makris; et al. (2009). Practical Hemostasis and Thrombosis . Wiley-Blackwell. p. 2. ISBN   978-1-4051-8460-1.
  26. Watson, M. S.; Pallister, C. J. (2010). Haematology (2nd ed.). Scion Publishing Limited. pp. 334–336. ISBN   978-1-904842-39-2. OCLC   1023165019. Archived from the original on 8 February 2022. Retrieved 8 February 2022.
  27. 1 2 3 4 5 6 7 Pallister CJ, Watson MS (2010). Haematology. Scion Publishing. pp. 336–347. ISBN   978-1-904842-39-2.
  28. "Coagulation Factor". Clotbase.bicnirrh.res.in. Archived from the original on 11 December 2016. Retrieved 20 May 2018.
  29. Hoffbrand, A. V.; Pettit, J. E; Moss, P. A. H. (2002). Essential Haematology (4th ed.). London: Blackwell Science. pp. 241–243. ISBN   978-0-632-05153-3. OCLC   898998816. Archived from the original on 8 February 2022. Retrieved 8 February 2022.
  30. 1 2 Long AT, Kenne E, Jung R, Fuchs TA, Renné T (March 2016). "Contact system revisited: an interface between inflammation, coagulation, and innate immunity". Journal of Thrombosis and Haemostasis. 14 (3): 427–437. doi: 10.1111/jth.13235 . PMID   26707513.
  31. Hoffman, M. (August 2003). "Remodeling the blood coagulation cascade". Journal of Thrombosis and Thrombolysis. 16 (1–2): 17–20. doi:10.1023/B:THRO.0000014588.95061.28. PMID   14760207. S2CID   19974377. Archived from the original on 8 February 2022. Retrieved 8 February 2022.
  32. 1 2 Hoffbrand, A. V. (2002). Essential haematology. Oxford: Blackwell Science. pp. 243–245. ISBN   978-0-632-05153-3.
  33. Immunology – Chapter One: Innate (non-specific) immunity Archived 21 October 2014 at the Wayback Machine Gene Mayer, Ph.D. Immunology Section of Microbiology and Immunology On-line. University of South Carolina
  34. Palta, A.; Palta, S.; Saroa, R. (2014). "Overview of the coagulation system". Indian Journal of Anaesthesia. 58 (5): 515–523. doi: 10.4103/0019-5049.144643 . ISSN   0019-5049. PMC   4260295 . PMID   25535411.
  35. Signorelli, Salvatore Santo; Oliveri Conti, Gea; Fiore, Maria; Cangiano, Federica; Zuccarello, Pietro; Gaudio, Agostino; Ferrante, Margherita (26 November 2020). "Platelet-Derived Microparticles (MPs) and Thrombin Generation Velocity in Deep Vein Thrombosis (DVT): Results of a Case–Control Study". Vascular Health and Risk Management. 16: 489–495. doi: 10.2147/VHRM.S236286 . ISSN   1176-6344. PMC   7705281 . PMID   33273818.
  36. Singh, S.; Dodt, J; Volkers, P.; Hethershaw, E.; Philippou, H.; Ivaskevicius, V.; Imhof, D.; Oldenburg, J.; Biswas, A. (5 August 2019). "Structure functional insights into calcium binding during the activation of coagulation factor XIII A". Scientific Reports. 9 (1): 11324. Bibcode:2019NatSR...911324S. doi:10.1038/s41598-019-47815-z. ISSN   2045-2322. PMC   6683118 . PMID   31383913.
  37. David Lillicrap; Nigel Key; Michael Makris; Denise O'Shaughnessy (2009). Practical Hemostasis and Thrombosis . Wiley-Blackwell. pp.  7–16. ISBN   978-1-4051-8460-1.
  38. 1 2 Stang, LJ; Mitchell, LG (2013). "Fibrinogen". Methods in molecular biology (Clifton, N.J.). 992: 181–92. doi:10.1007/978-1-62703-339-8_14. PMID   23546714.
  39. "Prothrombin Time Test and INR (PT/INR): MedlinePlus Medical Test". medlineplus.gov. Retrieved 28 April 2024.
  40. Hughes-Jones, N. C.; Wickramasinghe, S. N.; Hatton, Chris (2008). Haematology (8th ed.). Oxford, England; Hoboken, New Jersey: Wiley-Blackwell Publishers. pp.  145–166. ISBN   978-1-4051-8050-4. OCLC   1058077604. Archived from the original on 8 February 2022. Retrieved 8 February 2022.
  41. "Disseminated Intravascular Coagulation". The Lecturio Medical Concept Library. Archived from the original on 12 July 2021. Retrieved 12 July 2021.
  42. Huber J, Stanworth SJ, Doree C, Fortin PM, Trivella M, Brunskill SJ, et al. (November 2019). Cochrane Haematology Group (ed.). "Prophylactic plasma transfusion for patients without inherited bleeding disorders or anticoagulant use undergoing non-cardiac surgery or invasive procedures". The Cochrane Database of Systematic Reviews. 2019 (11): CD012745. doi:10.1002/14651858.CD012745.pub2. PMC   6993082 . PMID   31778223.
  43. Kaufmann, J. E.; Vischer, U. M. (1 April 2003). "Cellular mechanisms of the hemostatic effects of desmopressin (DDAVP)". Journal of Thrombosis and Haemostasis. 1 (4): 682–689. doi:10.1046/j.1538-7836.2003.00190.x. ISSN   1538-7836. PMID   12871401. S2CID   30749769.
  44. Soff GA (March 2012). "A new generation of oral direct anticoagulants". Arteriosclerosis, Thrombosis, and Vascular Biology. 32 (3): 569–574. doi: 10.1161/ATVBAHA.111.242834 . PMID   22345595.
  45. Schmidt A (1872). "Neue Untersuchungen über die Faserstoffgerinnung". Pflügers Archiv für die gesamte Physiologie. 6: 413–538. doi:10.1007/BF01612263. S2CID   37273997.
  46. Schmidt A. Zur Blutlehre. Leipzig: Vogel, 1892.
  47. Arthus M, Pagès C (1890). "Nouvelle theorie chimique de la coagulation du sang". Arch Physiol Norm Pathol. 5: 739–746.
  48. Shapiro SS (October 2003). "Treating thrombosis in the 21st century". The New England Journal of Medicine. 349 (18): 1762–1764. doi:10.1056/NEJMe038152. PMID   14585945.
  49. Brewer DB (May 2006). "Max Schultze (1865), G. Bizzozero (1882) and the discovery of the platelet". British Journal of Haematology. 133 (3): 251–258. doi: 10.1111/j.1365-2141.2006.06036.x . PMID   16643426.
  50. Morawitz P (1905). "Die Chemie der Blutgerinnung". Ergebn Physiol. 4: 307–422. doi:10.1007/BF02321003. S2CID   84003009.
  51. 1 2 3 4 5 6 Giangrande PL (June 2003). "Six characters in search of an author: the history of the nomenclature of coagulation factors". British Journal of Haematology. 121 (5): 703–712. doi:10.1046/j.1365-2141.2003.04333.x. PMID   12780784. S2CID   22694905.
  52. Macfarlane RG (May 1964). "An enzyme cascade in the blood clotting mechanism, and its function as a biochemical amplifier". Nature. 202 (4931): 498–499. Bibcode:1964Natur.202..498M. doi:10.1038/202498a0. PMID   14167839. S2CID   4214940.
  53. Davie EW, Ratnoff OD (September 1964). "Waterfall sequence for intrinsic blood clotting". Science. 145 (3638): 1310–1312. Bibcode:1964Sci...145.1310D. doi:10.1126/science.145.3638.1310. PMID   14173416. S2CID   34111840.
  54. Wright IS (February 1962). "The nomenclature of blood clotting factors". Canadian Medical Association Journal. 86 (8): 373–374. PMC   1848865 . PMID   14008442.
  55. Osaki T, Kawabata S (June 2004). "Structure and function of coagulogen, a clottable protein in horseshoe crabs". Cellular and Molecular Life Sciences. 61 (11): 1257–1265. doi:10.1007/s00018-004-3396-5. PMID   15170505. S2CID   24537601.

Further reading