Factor XIII Deficiency

Back

Practice Essentials

Factor XIII (FXIII), which was initially termed fibrin stabilizing factor, is involved in clot preservation. FXIII also participates in other physiologic processes, including wound repair and healing. FXIII deficiency, an autosomal recessive disorder, is a rare but potentially life-threatening cause of a hemorrhagic diathesis. Paradoxically, alterations in FXIII may also predispose to thrombosis. 

Congenital FXIII deficiency is due principally to defects in the catalytic A subunit of FXIII, with more than 100 mutations throughout the factor XIII A gene identified.[1]  Acquired FXIII deficiencies, which result from autoantibodies against FXIII subunits, are extremely rare but may produce severe bleeding diatheses.[2]

Thrombin, generated by reactions initiated by activated tissue factor VII/factor IX pathways, leads to clot formation. See the image below.



View Image

Final steps in clot formation (from article: Factor XIII).

Signs and symptoms

The following symptoms should trigger an evaluation for FXIII deficiency:

Bleeding into joints

Physical findings

Physical findings depend on the site at which bleeding develops and include the following:

See Clinical Presentation for more detail.

Diagnosis

The following routine tests are the first step in the evaluation of any bleeding disorder:

However, these tests cannot be used to screen for FXIII deficiency because the results would be within reference ranges in a patient with isolated severe FXIII deficiency.[4]

Qualitative screening test for severe FXIII deficiency

Quantitative testing

If the 5M urea solubility test demonstrates positive results, this finding should be confirmed by quantitating FXIII activity using a monodansylcadaverine or putrescine incorporation assay.

A new sensitive assay used to quantitate FXIII activity is based on monitoring the amount of ammonia (NH3) released by using glutamate dehydrogenase and nicotinamide adenine dinucleotide phosphate during the transamidation reaction (cross-linking) by FXIII. Another new and sensitive colorimetric assay is based on incorporation of 5-(biotinamido) pentylamine into fibrin/fibrinogen.[5]

In addition, a2PI and plasminogen activator inhibitor-1 assays should be performed to exclude abnormalities in the fibrinolytic pathway, which accelerate clot lysis. Sodium dodecylsulfate polyacrylamide gel electrophoresis under reducing conditions has been used to assess the presence of cross-linked g or a chains of fibrin, which is a reflection of FXIII activity. The studies must be performed by laboratory personnel with special expertise.

Testing for inhibitors

Prenatal diagnosis

See Workup for more detail.

Management

FXIII replacement is used to treat bleeding, to prevent perioperative bleeding during elective surgical procedures or, prophylactically, to prevent recurrent bleeding, as in CNS or joint hemorrhages. Serial monitoring of achieved FXIII levels is essential to document the adequacy of any therapy.

FXIII concentrates for replacement are as follows:

Minor bleeding, as from cuts and abrasions, may respond to conservative measures, such as pressure, ice, and use of antifibrinolytic drugs. Avoidance of trauma and nonsteroidal anti-inflammatory drugs (NSAIDs) is helpful in reducing bleeding events.

Treatment of patients with inhibitors

See Treatment and Medication for more detail.

Background

The hemostatic system, consisting of blood vessels and blood, plays a crucial role in human survival. The importance of the plasma coagulation system in protecting life and preventing further blood loss following transection of a blood vessel has been understood for a long time. Blood normally is maintained in a fluid state, without evidence of bleeding or clotting. The presence of a bleeding diathesis in families with an X-linked pattern of inheritance of the disorder has been recognized for hundreds of years.

The recognition of factor deficiencies as the cause of hemophilias spurred investigations into the causes of other bleeding disorders and led to progress in understanding normal hemostasis. Knowledge of the fact that blood clots that are formed in the presence of calcium are stronger, insoluble in alkali, and resistant to proteolytic degradation led to the concept of insoluble clots in the earlier part of the last century.

In 1948, Laki and Lorand recognized that a serum factor, termed fibrin stabilizing factor, was responsible for the characteristics of insoluble fibrin clots.[6] In 1960, Duckert et al described the first case of an "undescribed congenital haemorrhagic diathesis probably due to fibrin stabilizing factor deficiency," which was a description of the consequences of severe factor XIII (FXIII) deficiency.[7, 8]

The importance of FXIII in the process of coagulation is underscored by symptoms borne by patients who are homozygously deficient in FXIII or who have an antibody that disrupts FXIII function. Paradoxically, alterations in FXIII may predispose patients to thrombosis. Based on all available data, FXIII is clearly involved in the clot preservation side of the delicate balance between clot formation and stability and clot degradation. FXIII participates in other physiologic processes, including wound repair and healing. The many functions of FXIII and the disruptions of those functions by mutations in the genes coding for FXIII are the subjects of on-going investigations.[9, 10, 11]

Gene polymorphisms are being evaluated for their influence on susceptibility to venous and arterial thromboembolism.[12] Variants of coagulation factors, including FXIII Val34Leu, have been implicated in influencing susceptibility to thromboembolic diseases.[13]

There is a question as to whether FXIII Val34Leu polymorphism is protective against idiopathic venous thromboembolism.The substitution of leucine for valine at amino acid position 34 of the FXIII gene, commonly referred to as FXIII Val34Leu polymorphism, has been reported to confer protection against venous thromboembolism. However, the results of a study in a white Canadian population did not support an independent association of the FXIII Val34Leu polymorphism with idiopathic venous thromboembolism.[14]

An association may exist between the FXIII Leu allele and a modest protective effect against acute myocardial infarction (MI) and may provide useful information in profiling susceptibility to MI.[15]

FXIII measurement has a variety of uses, potential and confirmed. Plasma levels of FXIII were found to be decreased in children with Henoch-Schönlein purpura having severe abdominal symptoms. Thus, it has been suggested that measurement of FXIII level may be of value to detect the vasculitic process of Henoch-Schönlein purpura before the rash occurs or long after it has disappeared in patients with isolated abdominal or scrotal problems.[16, 17] Immunohistochemistry studies have shown that FXIIIa-positive dermal dendritic cells were increased in a variety of skin tumors, including dermatofibromas.[18]

Severe FXIII deficiency, a rare autosomal recessive coagulation disorder, is associated with a relatively common prevalence of F13B gene defects, at least within the German population. The regions in and around the cysteine disulphide bonds in the FXIII-B protein are the sites of frequent mutations.19 It is relatively common in Iran, especially in Khash; most patients there have a unique mutation in the F13A gene (Trp187Arg), producing severe FXIII deficiency.[19]  

FXIIIs aids immobilization and killing of bacteria as well as phagocytosis by macrophages, likely functioning as part of the innate immune system.[20]

Use of relatively new specific FXIII assays are pivotal to avoid missing the diagnosis of FXIII deficiency, a rare but potentially life-threatening disorder.[21]

Pathophysiology

Structure, production, and half-life of FXIII

Plasma FXIII is a heterotetramer consisting of 2 identical proenzyme subunits (A2) and 2 identical carrier protein subunits (B2). Subunit A contains the catalytic site, the activation peptide, a calcium-binding site, and free sulfhydryl (SH) groups. Subunit B, a glycoprotein, acts as a carrier protein that stabilizes subunit A, binds the zymogen (subunit A) to fibrinogen, and acts as a brake on FXIII activation.[22, 23] Subunit B circulates in plasma as part of the tetramer A2 B2 and as a free B2 dimer; all of plasma subunit A is complexed with subunit B. The concentration of subunit A in plasma is 15 mg/mL, while that of subunit B is 21 mg/mL. Much of FXIII circulates in blood in association with fibrinogen.[24, 25]

Platelet FXIII (an A2 homodimer) constitutes approximately 50% of total FXIII activity in blood. Plasma FXIII has a long half-life of approximately 9-14 days. A similarity exists between a portion of the carboxy terminal (C terminal) domain of FXIII and the receptor-binding region of a2 -macroglobulin. The complex of a2 -macroglobulin and its substrate protease is removed from the circulation by binding to its receptor in the liver and other tissues; therefore, as has been suggested, FXIII also may be removed from the circulation by a similar mechanism.[26, 27] Some features of the A and B chains of FXIII are listed below. Monoclonal antibodies and naturally occurring inhibitors are used to elucidate structure-activity relationships.

Bone marrow cells, megakaryocytes, and monocytes/macrophages synthesize FXIII, with a possible role for hepatocytes in the synthesis of subunit A. Subunit B is synthesized by the liver. Tissue transglutaminase, the intracellular form of FXIII, consists of the A2 subunit (an A2 homodimer) and is present in a variety of cells including platelets, megakaryocytes, monocytes/macrophages, and in the liver, placenta, uterus, prostate, and dermal dendrocytes.[28] Red cells contain a transglutaminase that is activated by Ca2+ but is different from plasma transglutaminase in its cross-linking activity and can cross-link fibrinogen as well as fibrin. Trapped erythrocytes release FXIII when red cells lyse, providing additional cross-links to the aging thrombus.[22]

Table. Some Features of the A and B Chains of Factor XIII



View Table

See Table

Comparative biology shows that transglutaminases are distributed widely in nature and may represent the prototype for the evolution of clotting enzymes.[29] Partial homology of plasma FXIII exists with several proteins including tissue and keratinocyte transglutaminases, erythrocyte transglutaminase, and the hemocyte transglutaminase of the horseshoe crab and other zymogens of the same family.

A recent example is from the crystal structure of transglutaminase of the Red Sea bream, which shows that its active site and overall structure resemble that of human FXIII.[30] These homologies attest to conservation of the enzyme during evolution. Since the gene structures are similar, it is believed that they evolved from a common ancestor. Subunit B contains 10 repeating "sushi" units linked by disulfide bonds; the function of the sushi unit is unknown. Sushi structures are present in at least 26 proteins, including proteins in the horseshoe crab and in the vaccinia virus.

Activation

Thrombin, generated by reactions initiated by activated tissue factor VII/factor IX pathways (as illustrated in the first diagram below), leads to clot formation. Thrombin releases fibrinopeptide A from the a chain of fibrinogen, then fibrinopeptide B from the b chain of fibrinogen. Fibrin monomers (formed following the release of fibrinopeptides) polymerize spontaneously; this is followed by development of a complex branching clot as a result of the actions of activated FXIII (FXIIIa).[31] The sequence of these final steps is found in the second chart below.



View Image

Coagulation reactions leading to thrombin generation and activation of factor XIII.



View Image

Final steps in clot formation (from article: Factor XIII).

Thrombin starts the process of FXIII activation by cleaving an activation peptide from subunit A. The subsequent Ca2+ -dependent dissociation of subunit B allows FXIII activation to proceed. Calcium is important for activation of the zymogen (both FXIII and tissue transglutaminase require Ca2+), conformational changes, and opening of the catalytic site of FXIII to its substrate. Calcium also provides physical stability as determined by x-ray crystallography, computer modeling, and other studies; all of the changes allow the active subunit A to perform its functions optimally.[22, 32, 33]

When activated by thrombin, tissue FXIII functions in the same manner as plasma FXIIIa. Platelet FXIII undergoes nonproteolytic activation following the platelet activation-induced rise in cytosolic Ca2+. Activation of the red cell enzyme occurs upon exposure to Ca2+, and red cells that are present in the fibrin clot lyse and release their FXIII as the clot ages. Several controls in the complex activation process focus the actions of FXIIIa on fibrin rather than on fibrinogen. Cross-linking of polymerized soluble fibrin by FXIIIa is the final step in hemostasis, as illustrated in the image below. For extensive details of this activation process, the reader is referred to two recent reviews by Lorand.[22, 29]



View Image

Activation of factor XIII and generation of insoluble cross-linked fibrin. Adapted from Lorand L. Ann N Y Acad Sci. 2001;936:291-311.

Role of FXIII in cross-linking and resistance to lysis

FXIIIa cross-links the lysine of one g chain in the fibrin polymer with the glutamine of another g chain by transamidation, releasing ammonia in the process. Additional cross-links occur between a-a chains, a-g chains, a chains-a2 -plasmin inhibitor (a2 PI), and a chains-fibronectin. As a result of the extensive cross-linking actions of FXIIIa, the clot structure of fibrin polymers increases in complexity from dimers to trimers to tetramers.

The g chains of fibrinogen and fibrin normally bind to the platelet membrane glycoprotein IIb/IIIa complex. The same g chains are subject to cross-linking by FXIIIa; therefore, cross-linking also occurs between fibrin and the platelet membrane. Both plasma FXIIIa and platelet FXIIIa cross-link fibrin polymers, but under physiologic conditions, platelet FXIII is believed to play a minor role.[34] Red cell FXIII is responsible for hybrid cross-linking of a-g chains, in contrast to the actions of plasma FXIII.

Dysfibrinogenemias and dyshypofibrinogenemias result in alterations in fibrin (substrate for FXIIIa), which can interfere with the ability of FXIII to cross-link fibrin. A reduction in available fibrin resulting from afibrinogenemia can have the same effect. Conversely, increased fibrinogen levels have been identified as a risk factor for thrombosis.

Mechanisms of this risk were elucidated by a fibrinolysis assay containing purified components. The assay showed that lysis of fibrin decreased as fibrinogen levels increased, and the presence of a minor common variant of fibrin (g') is associated with accelerated cross-linking, which made the clot resistant to proteolysis by both plasmin and trypsin. Increased clot stability also was believed to result from increased concentration of FXIII in the clot. Non-cross-linked fibrin potentiates activation of FXIII by thrombin; thus, the substrate potentiates its enzyme, further contributing to clot stability.[25, 35, 36]

Cross-linking of a2 PI to a chains of fibrin by FXIIIa brings the principal inhibitor of plasmin to the site of the clot, ensuring resistance of the clot to proteolysis. Inhibition of a2 PI in in vitro systems leads to enhanced clot lysis. In humans, deficiency of a2 PI results in a bleeding disorder because of vulnerability of the fibrin clot to prompt degradation by plasmin. The formation of highly cross-linked a-fibrin polymers in the presence of high concentrations of FXIIIa produces clots that are highly resistant to fibrinolysis.[37]

Fibronectin, an adhesive protein, is a large component (approximately 4%) of the proteins in a fibrin clot, is present in plasma and cells, and is subject to cross-linking by both plasma and cellular FXIII. Cross-linking of fibronectin to fibronectin and fibronectin to fibrin is accomplished by FXIIIa, with fibronectin contributing to increased fiber size, density, and strength of the clot. FXIIIa also cross-links actin to fibrin and actin to myosin. Cross-linking of intracellular structural proteins is involved in clot retraction and cell migration. This complex gel network created by the actions of FXIII plays an important role in wound healing, cell adhesion, and cell migration. All of these cross-linking reactions impart increased mechanical strength to the clot, contributing to clot retraction and resistance of the clot to degradation by plasmin and providing an explanation for the known plasmin resistance of older clots.

Many other proteins function as substrates for FXIIIa, including von Willebrand factor (vWF), factor V (FV), thrombospondin, gelsolin, vitronectin, vinculin, lipoprotein (a), and collagen (FXIIIa cross-links collagen with fibronectin and vWF, attaches the clot to the vessel wall, impacts tissue repair, increases resistance of collagen to proteolysis, and modulates synthesis of collagen by fibroblasts). Thus, FXIII plays a role in a wide array of cross-linking reactions involving plasma proteins at the intracellular level, impacting many different functions.

Note that because of the lack of cross-linking in individuals with FXIII deficiency, their D-dimer level will remain low, even if they experience thrombosis, although levels of other fibrin degradation products will be increased. Consequently, the D-dimer assay is not reliable for screening in these patients.

Factors affecting level and activity of FXIII

When quantitative amine incorporation assays became available, healthy people were found to have an 8-fold spread in FXIIIa activity.[38] In recent studies of FXIII antigen and activity in humans, no correlation was found between these two parameters. During the search for an explanation, 23 unique FXIIIa genotypes were found. The Leu34 and Leu564 variants gave rise to increased specific activity; the Phe204 variant lowered specific activity. Other mutations gave rise to low, high, or median FXIII-specific activity, and some variants had no effect.[39, 40]

In a study of the variability of FXIII levels in racial groups, FXIII activity was found to be higher in Asian Indians (male and female) than in their Chinese counterparts, accounting for approximately one fourth of the variability. Common genetic polymorphisms in the A and B chains appeared to contribute to the differences.[41] An influence exerted by acquired factors was evident in the higher FXIII levels found in women who smoked 20 or more cigarettes per day during a normal pregnancy than was found in nonsmokers, with a lesser drop in the second half of pregnancy.[42]

Role of FXIII in pregnancy

In the latter half of pregnancy, some drop in FXIII levels is normal, but severe (homozygous) FXIII deficiency is a cause of recurrent miscarriages. In a study of gestational tissues, FXIII was found in the decidual layer of the placenta, while FXIII secretion was evident in cultures of round-shaped endometrial cells. A study of early (7-8 wk) gestational tissues obtained from women without FXIII deficiency and from a woman who was homozygous for FXIII deficiency showed poorly formed cytotrophoblastic shells and Nitabuch layers, along with absence of FXIIIa in tissues obtained from the woman with FXIII deficiency. Low plasma levels of FXIII appear to correlate with low placental levels of FXIII with poor trophoblastic development, which may be the cause of spontaneous miscarriages.

It has been suggested that preventing miscarriage in patients who are severely deficient requires FXIII supplementation beginning at approximately 5 weeks of gestation because FXIII, fibrinogen, and fibronectin are necessary to anchor cytotrophoblasts invading the endometrium.[43, 44] Reduced FXIII activity resulting from the Tyr204Phe mutation has been associated with repeated miscarriages.[45]

Role of FXIII in wound healing

Physiologically, hyperpermeability induced by severe metabolic inhibition of porcine aortic endothelial cells is prevented by FXIIIa, which is similar to the maintenance of endothelial barrier function by FXIIIa despite depletion of energy or during reperfusion of ischemic rat hearts.[46] In a different system, FXIII induced epithelial wound healing by increasing cell growth by approximately 2.5 fold, leading to replacement of damaged cells.[47] Smooth muscle cell migration, an integral part of the healing process, is facilitated by FXIII. Migration of smooth muscle cells in cross-linked fibrin gels was twice the migration seen in non–cross-linked gels, demonstrating the importance of the 3-dimensional clot structure created by cross-linking in smooth muscle cell migration.[48] In humans, Fibrogammin was shown to contribute to the healing of venous leg ulcers by reducing endothelial permeability.[49]

Effects of other agents on FXIII

Nitric oxide (NO), an important diffusible molecular messenger, is increasingly recognized as having an impact on coagulation proteins. Activity of plasma transglutaminase is inhibited by NO via nitrosylation of critical thiol groups (reactive cysteine residue), resulting in inhibition of both g-chain cross-linking and insoluble clot formation. NO donors and carriers inhibit FXIII activity in a dose-dependent manner, in a purified system and in plasma. Tissue transglutaminases are involved in apoptosis, and inhibition of their activity by NO prevents apoptosis.[50, 51]

Venoms and toxins can affect clot stability. Excessive bleeding resulting from envenomation can affect the functions of FXIII in different ways. Acuthrombin A, one of two proteases in the venom of Agkistrodon acutus (five-pace snake), activates FXIII.[52] Ancrod, obtained from the venom of Agkistrodon species, causes defibrination, thereby removing the substrate for FXIII.

A severe systemic bleeding disorder may develop several hours after initial contact with 2 types of caterpillars in the Saturniidae family (from Brazil and Venezuela). Intracranial and intracerebral bleeding and renal failure may follow. In this case, FXIII reduction results from generalized disseminated intravascular coagulation (DIC) induced by several activities directed against the hemostatic mechanism, including a FXIII proteolytic-urokinase–like activity.[53] Tridegin, a peptide inhibitor of FXIII present in the saliva of an Amazon leech (Haementeria ghilianii) accelerates fibrinolysis by inhibiting FXIIIa; tridegin is under investigation as a potential new antithrombotic agent. Destabilase, an enzyme present in the leech, hydrolyzes g-g fibrin cross-links and breaks down blood clots.[54]

Simvastatin is a commonly used cholesterol-lowering agent. A non–antibody-mediated drug-induced reduction in FXIII activity as part of a broader reduction in hemostatic activation has been suggested to be the reason for the proven antithrombotic efficacy of simvastatin in clinical trials.[55] Blood samples were obtained sequentially every 30 seconds from a bleeding time cut in patients with coronary artery disease, before and 3 months after simvastatin treatment. Samples were analyzed for the time-course drop in fibrinogen levels and activation of factors II, V, and XIII by quantitative Western blot analyses. Simvastatin, independent of its effects on cholesterol, significantly reduced the rate of blood clotting, as evidenced by reduced formation of several activation products including FXIIIa.

Several selective synthetic inhibitors have been shown to prevent the ability of FXIIIa to stabilize a clot, thereby reducing clot strength (clot stiffness, viscoelastic modulus) to approximately 20% of normal (values similar to those seen in patients with severe FXIII deficiency). Rapid lysis of these clots occurred following in vitro exposure to thrombolytic agents.[29] Imidazolium derivatives, a new class of compounds, specifically inhibit both FXIII-induced formation of a-chain polymers and the incorporation of a2 PI into the a chain of fibrin, resulting in accelerated clot lysis.[11, 56]

Specific monoclonal antibodies to FXIII have provided similar benefits by reducing the viscoelastic properties and by enhancing clot lysis. They also have been used to modify disease states. The beneficial effect of the absence of cross-linked fibrin on pathophysiologic processes was proven in an animal model of widespread thrombosis. FXIIIa deficiency induced in rabbits by pretreatment with a specific monoclonal antibody before induction of a generalized Schwartzman reaction protected them from the deleterious effects of widespread microvascular thrombosis. The protection resulted from the ability of the fibrinolytic system to effectively degrade non–cross-linked thrombi.[57] These data add support to the author's speculation many years ago of the potential use of drugs that inhibit cross-linking as a method of prophylaxis in venous thromboembolic disease.

The biochemical basis and potential for using modifiers of fibrin stabilization in improved thrombolytic therapies are discussed in a recent review by Lorand.[29] Similar ideas have been proposed by others, expanding on the importance of fibrin structure in thrombus formation and dissolution.[58] Prospective clinical trials must prove any thromboprophylactic efficacy of altering fibrin structure using specific drugs.

Other functions of FXIII

Plasma and tissue transglutaminases have been reported to promote cell adhesion through specific integrins for 2 different tumor cell types, MOLT-3 human lymphocyte–like leukemia and melanoma cells and SW480 colon cancer cells transfected with a ligand.[59] In contrast, FXIII did not stimulate growth of cultured human tumor cells.[60] An intriguing observation is the potential use of subunit A of FXIII and FXIII activity as a tumor marker in malignant brain tumors; its presence may distinguish benign from malignant brain tumors.[61] Recently, it was shown for the first time that intranuclear accumulation and cross-linking activity of FXIIIa occurred in maturing monocytes, supporting the hypothesis that FXIIIa may be involved in cell proliferation/differentiation, chromatin structure remodeling, and even cell death.[62] Further data are needed to unravel the role of FXIII in malignancies.

An unexpected role has been postulated for FXIII in degenerative brain disorders. In Alzheimer disease and spongiform encephalopathies, the brain contains fibrils that develop from native proteins containing a discordant a helix. Human FXIII was found to form fibrils in buffered saline, suggesting that FXIII, in addition to several other proteins, can be a source of this abnormal fibrillar protein.[63]

Possible interactions between deficiencies of FXIII and thrombin-activatable fibrinolytic inhibitor

Thrombin-activatable fibrinolytic inhibitor (TAFI), a single-chain carboxypeptidase B–like zymogen, is activated by thrombin to become activated TAFI (TAFIa).[64] The importance of TAFIa in fibrinolysis is emphasized by the fact that the conversion of only 1% of the zymogen to TAFIa is sufficient to suppress fibrinolysis by approximately 60%.

TAFIa suppresses fibrinolysis by removing C-terminal lysine and arginine residues exposed in the partially degraded fibrin clot produced by plasmin. Removal of C-terminal lysine residues from fibrin reduces the rate of plasminogen activation by a number of mechanisms, attenuating fibrinolysis. This effect is counterbalanced in normal plasma by activation of protein C, which has profibrinolytic properties because of its ability to suppress thrombin generation via its major effect of degrading activated factor V (FVa), and to a lesser extent, activated factor VIII (FVIIIa).[64, 65, 66]

As illustrated in the chart below, a delicate balance usually exists between thrombus formation and thrombus resolution; thrombin secures survival of the thrombus created by its action on fibrinogen by activating TAFI, thereby inhibiting fibrinolysis. Cross-linking of fibrin induced by FXIIIa (activated by thrombin) renders the clot insoluble. FXIII deficiency results in absence of cross-linked fibrin, leading to premature lysis of the clot by the fibrinolytic system; adverse consequences result, including bleeding. Theoretically, a deficiency of TAFI leading to decreased suppression of fibrinolysis (enhanced clot lysis) can potentiate bleeding resulting from FXIII deficiency (also associated with enhanced clot lysis). Note the image below.



View Image

Postulated interaction between factor XIII and thrombin-activatable fibrinolytic inhibitor.

Cell surface–directed hemostasis

The concept of coagulation as a waterfall or cascade effect has been acknowledged for a long time, with platelets and other cell surfaces providing the anionic phospholipids needed for complex formation, so that reactions can proceed efficiently. One review proposed that coagulation is essentially a cell surface–based event.[67] Platelet FXIII is positioned appropriately to influence the process. (See the diagram below.)



View Image

Cell surfaced–directed hemostasis. Initially, a small amount of thrombin is generated on the surface of the tissue factor–bearing (TF-bearing) cell. F....

Conclusion

Much work is needed, even in the clinical arena, to clarify the relationship between the exact levels of FXIII and hemorrhagic or thrombotic phenotypes. Establishing an international registry of patients deficient in FXIII would be of value in improving understanding of the protean manifestations of this uncommon disorder.

Frequency

United States

Overall estimated frequency of the autosomal recessive disorder involving a severe deficiency of subunit A is approximately 1 case per 2 million population. Previously, consanguinity was believed to be necessary, but the detection of compound heterozygotes by the application of molecular techniques is changing that perception. Approximately 200 cases of FXIII deficiency have been described thus far.[68] See Other Problems to be Considered for a discussion of acquired FXIII deficiency related to diseases or inhibitors.

International

FXIII deficiency has been reported in many ethnic groups around the world, including persons from Canada, Europe, India, Israel, Japan, Kuala Lumpur, Pakistan, Papua New Guinea, South America, Thailand, Turkey, and the United States. A high rate of consanguinity was documented in affected persons in Iran.[69]  In Khash, Iran  the prevalence is 1 homozygote per approximately 500 population, which is higher than its worldwide prevalence, with 3.5% heterozygotes.[19]

A prospective multicenter cohort project of inherited bleeding disorders in France identified 10,047 patients, with only 5.0% having a clotting factor deficiency of the uncommon varieties, specifically Factor I, II, V, combined V and VIII, VII, X, XI, and XIII.[70]

Diagnosis of disorders of FXIII inhibitors, which may have been missed in the past, is increasing as more laboratory support becomes available around the world. Increasing use of isoniazid (INH) to combat a worldwide rise in incidence of tuberculosis could contribute to an increased incidence of FXIII inhibitors in patients.

Variability in the distribution of mutations is exemplified by existing data. For example, significant ethnic heterogeneity was found in a Brazilian population in which the Val34Leu mutation was present in 51.2% of American Indians, 44% of whites, and 28.9% of Africans, but in only 2.5% of Japanese Asians.[71]

Mortality/Morbidity

Umbilical bleeding starting in the first few days after birth, recurrent intracranial bleeding, and recurrent early miscarriages are hallmarks of FXIII deficiency.

Approximately 30% of central nervous system (CNS) bleeding is recurrent, and approximately 50% of CNS bleeding may be fatal, but the severity of bleeding varies from family to family. Posttraumatic bleeding may be immediate, delayed, or recurrent. Traumatic joint bleeding may develop. Poor wound healing has been described, although this is not a universal finding.

Cryoprecipitate and fresh frozen plasma (FFP) provide a source of FXIII for most patients. All plasma-derived products carry risks of transmitting hepatitis, HIV, parvovirus B19, transfusion-transmitted virus (TTV), and prion-induced (new variant Creutzfeldt-Jacob disease [nvCJD]) illnesses (see Complications and the Medscape article Hemophilia A for more information). A recombinant factor XIII (rFXIII) subunit A concentrate is available for use in congenital FXIII A-subunit deficiency.[72]

Development of FXIII inhibitors (alloantibodies or autoantibodies) is associated with significant morbidity and mortality.

Pregnant women with FXIII deficiency have a significant risk of miscarriage, placental abruption, and postpartum hemorrhage without prophylaxis.[73]

Race-, sex-, and age-related demographics

No racial predilection exists for FXIII deficiency. FXIII deficiency has been reported widely. The restriction of certain polymorphisms to specific populations should be expected.

Since it is an autosomal disorder, homozygous FXIII deficiency occurs in either sex. Acquired inhibitors to FXIII can present in either males or females.

Physiologically, reduced levels of FXIII are found in healthy newborns, with a gradual rise in levels into the reference range. Premature infants have lower values than full-term neonates. FXIII levels drop in the latter half of a normal pregnancy.

Neonates with severe FXIII deficiency may present with bleeding from the umbilical cord. Easy bruising and delayed and recurrent bleeding after trauma begin in childhood. Oral bleeding can begin with teething and cuts or abrasions to the lips, tongue, and frenulum. Bleeding remains a problem throughout life and requires replacement therapy. FXIII deficiency acquired as a result of autoantibodies has been reported in the older population, as has acquired hemophilia A. Both drug-induced autoantibodies and alloantibodies have been reported in severely deficient patients who have been receiving replacement therapy.

History

The following symptoms should trigger an evaluation for factor XIII (FXIII) deficiency:

Physical

Physical findings depend on the site at which bleeding develops and include the following:

Causes

To date, most identified mutations leading to severe FXIII deficiency and a bleeding disorder involve subunit A, with very few mutations reported involving subunit B. The gene for subunit A is located on chromosome 6 bands p24-25. The gene is 160 kilobases in length and has 15 exons and 14 introns with specific structural and functional domains. Catalytic activity is encoded in the second exon, and the active cysteine is encoded by the seventh exon. The 2 Ca2+ -binding sites and a thrombin-inactivation site have been identified at other locations. The gene for subunit B is located on chromosome 1 bands q31-32.1, is 28 kilobases in length, and has 12 exons and 11 introns.[10, 75] (See the image below.)



View Image

Gene, messenger RNA, and protein for subunit A of factor XIII. Adapted from Reitsma PH. In: Hemostasis and Thrombosis: Basic Principles and Clinical P....

Detailed characteristics of complementary ribonucleic acid (cRNA) and messenger ribonucleic acid (mRNA) of the placental subunit A are known. The presence of an acetylated amino terminal end and the absence of glycosylation and disulfide bonds apparently are features typical of secreted cytoplasmic proteins. The presence of these characteristics makes it conducive for subunit A expressed in yeast systems to make a recombinant product.

Substitutions in the core domain of the enzyme, affecting highly conserved residues, result in a serious defect in structure and function. Missense mutations in the A chain are a common cause, accounting for approximately 50% of cases of severe FXIII deficiency. The defects result in an absence of subunit A protein but also are accompanied by a reduction in subunit B carrier protein (type II defect).

Nonsense mutations are an equally common cause of A chain defects, resulting in a frameshift-type, splice-type, or termination-type mutation. The few defects that have been reported in the B chain lead to a deficiency of the carrier protein (subunit B), which then leads to instability and reduction of plasma subunit A levels despite the presence of functional intracellular subunit A (type I defect).[76] Therefore, patients who are homozygous for subunit B mutations have a bleeding disorder. Most recently, impaired intracellular transport from the endoplasmic reticulum to the Golgi apparatus, with failure of secretion of the truncated FXIII subunit B produced by a single-base deletion, was reported to be the cause of severe FXIII deficiency in 3 unrelated patients.[77]

Many kinds of mutations have been (and continue to be) identified, with some mutations unique to certain families. The finding of compound heterozygotes has eliminated the mandatory search for consanguinity in all parents of patients with severe FXIII deficiency.[78, 79, 45]

An unusual mutation has been described in 2 Finnish sisters with a very mild bleeding disorder. One sister had 2 successful pregnancies without regular replacement therapy. The sisters had no detectable subunit A activity (< 1%) using plasma screening tests; however, using the 3H-putrescine incorporation assay, subunit A showed 0.35% of normal activity, with partial g-g dimerization of fibrin in clotted plasma. A full-length subunit A was detected in the patients' platelets using Western blot analysis.

The sisters had an Arg661→stop mutation on one allele and a T→C transition on the other allele. These data showed that a mutation in the splice donor site of intron C can result in different variant mRNA transcripts and that small amounts of correctly processed mRNA can produce a type of FXIII that can produce, at least, dimerization of fibrin, thus minimizing the clinical consequences.[74]

Various reported mutations are spread throughout the gene coding for FXIII without specific hot spots. In many patients, low steady-state mRNA levels have been found, which result in inefficient production of the abnormal protein.[11]

Data in the literature conflict regarding the impact of the common FXIII subunit A Val34Leu mutation (associated with higher plasma transglutaminase activity) on thrombotic disease. Note the following:

A possible cooperative interaction between the Val34Leu mutation and other known thrombophilic mutations also has been explored. Note the following:

Genetic polymorphisms affecting both the A and B subunits have been reported, but because they do not involve conserved amino acids or are not important for protein structure, they do not result in FXIII deficiency and bleeding. Based on an analysis of polymorphisms in the gene for FXIII subunit A and their products in a northern Portuguese population, it has been stated that the evolutionary order of appearance of the main protein alleles for FXIII is 1B-->2B-->1A-->2A and that intragenic combinations are likely to have played a role in the molecular diversity in the main FXIII subunit A alleles.[85]

Genetic polymorphisms and, particularly, intragenic polymorphisms are useful in genetic counseling of families with unknown mutations. For example, 80% of whites are heterozygous for a tetrameric repeat in intron 1 of subunit A, which can help differentiate defects in subunit A from defects in subunit B.[10, 11, 75, 5] Some polymorphisms are universal, while others appear to be restricted to particular ethnic groups. The latter situation will change as ethnic intermarriages increase in this global society. Families with severe FXIII deficiency associated with a serious disabling bleeding disorder have access to all of the genetic tools available to patients with hemophilia A and B.

Disorders of fibrin stabilization can affect the activity of FXIII or its substrates fibrin and fibrinogen. A proposed classification of disorders leading to a positive urea solubility test result is presented below.

Abnormalities of FXIII (enzyme) are as follows:

Abnormalities of the substrate for FXIII (fibrin/fibrinogen) are as follows:

Laboratory Studies

The following routine tests are the first step in the evaluation of any bleeding disorder:

However, these tests cannot be used to screen for FXIII deficiency because the results would be within reference ranges in a patient with isolated severe FXIII deficiency,[4] and blood collection, transport, and storage may alter results for these initial screening tests.[96]

The next test performed is a qualitative screening test for severe FXIII deficiency that assesses clot solubility in 5M urea or 1% monochloroacetic acid. If the thrombin and Ca2+ -induced clot lyses within a few hours, severe FXIII deficiency is suggested provided fibrinogen levels are qualitatively and quantitatively within reference range. Excluding hypofibrinogenemia and dysfibrinogenemia is important, since these conditions cause false-positive results on the 5M urea solubility test. The thrombin-clottable fibrinogen test can be used to exclude hypofibrinogenemia and dysfibrinogenemia.

If the 5M urea solubility test demonstrates positive results, this finding should be confirmed by quantitating FXIII activity using a monodansylcadaverine or putrescine incorporation assay, which must be performed by laboratory personnel with expertise.

Thromboelastography (TEG) is an old method used to assess clotting and lysis of fresh whole blood, and it has been used as an early tool in the initial evaluation, and as a simple laboratory test, of the mechanical strength (effect of FXIII) of fibrin sealants.[97] However, TEG cannot supplant any of the qualitative or quantitative tests discussed in this section.

A sensitive assay used to quantitate FXIII activity is based on monitoring the amount of ammonia (NH3) released by using glutamate dehydrogenase and nicotinamide adenine dinucleotide phosphate during the transamidation reaction (cross-linking) by FXIII. Note the following:

Another sensitive colorimetric assay based on incorporation of 5-(biotinamido) pentylamine into fibrin/fibrinogen was compared to a photometric method based on ammonia release and an ELISA of FXIII subunit A to quantitate FXIII activity. The test was shown to be sensitive to both reductions and increases in activity; the increases resulted from the FXIII Val34Leu mutation.[5]

In addition, a2 PI and plasminogen activator inhibitor-1 assays should be performed to exclude abnormalities in the fibrinolytic pathway, which accelerate clot lysis.

Sodium dodecylsulfate polyacrylamide gel electrophoresis under reducing conditions has been used to assess the presence of cross-linked g or a chains of fibrin, which is a reflection of FXIII activity. The studies must be performed by laboratory personnel with special expertise.

If the presence of an inhibitor is suspected in a patient with a positive urea solubility test result, the next step is to repeat the urea solubility test with mixtures containing varying proportions of patient and normal plasma to differentiate between a deficiency or an inhibitor as the cause of a positive result. Since FXIII activity is present in serum, serum also may be substituted for plasma in the test.

Semiquantitation of the susceptibility of the fibrin clot to fibrinolysis can be obtained by adding iodine-125-labeled fibrinogen, tissue plasminogen activator, thrombin, and Ca2+ to the patient's plasma, with measurement of the time to 50% clot lysis. This method is useful in evaluating inhibitors but must be performed by laboratory personnel with special expertise.

See Lorand for a recent review of further details of the sequence of necessary testing to confirm the presence of a FXIII inhibitor.[89]

Acquired systemic disorders, including decompensated DIC and liver disease, require standard tests to confirm the diagnosis.

Caution is warranted in obtaining blood samples for any coagulation assays from heparinized central lines because of the effect of large amounts of heparin on any coagulation test that depends on thrombin generation.

Prenatal diagnosis is as follows:

Perform liver function tests; kidney function tests; HIV-1 and HIV-2 antigen and antibody tests; hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D, and hepatitis E antigen/antibody levels; and other tests as needed.

Assess a-fetoprotein levels and other tumor markers as needed in patients with chronic hepatitis.

FXIII, which is involved in wound healing and angiogenesis, may be detectable by highly sensitive chemiluminescent ELISAs in tiny volumes of tear. This concept may provide a tool for monitoring FXIII subunit and complex levels in pathological conditions.[101]

Imaging Studies

MRI, CT scan, and ultrasound have been used to localize, quantify, and serially monitor the location and response of bleeding to specific therapy. Perform other imaging tests as needed to diagnose associated diseases.

Other Tests

Perform ECGs as needed.

Procedures

Diagnostic amniocentesis, chorionic villous sampling, or fetoscopy may be performed during pregnancy. Perform other routine procedures when indicated. Perform arthrocentesis only when infection is suggested. Any invasive procedure requires the appropriate factor replacement.

When indicated, perform other procedures, such as colonoscopy, in persons without hemophilia. Evaluate persistent GI tract bleeding without an apparent cause using endoscopy and colonoscopy to exclude underlying lesions. Persistent genitourinary tract bleeding requires evaluation for nephrolithiasis, tumors, or obstruction. If a biopsy is needed, patients require replacement therapy prior to and following the procedure until the biopsy site has healed.

Invasive lifesaving procedures should be performed in patients with inhibitors only in concert with appropriate treatment.

Approach Considerations

Factor XIII (FXIII) replacement is used to treat bleeding, to prevent perioperative bleeding during elective surgical procedures or, prophylactically, to prevent recurrent bleeding, as in central nervous system (CNS) or joint hemorrhages. Serial monitoring of achieved FXIII levels is essential to document the adequacy of any therapy.

Prompt and adequate therapy for acute bleeding is essential along with immobilization of the affected sites and pain relief. Most patients receive fresh frozen plasma (FFP) or cryoprecipitate to treat bleeding. Information regarding the amount of FXIII present in either of these products usually is not available; therefore, monitoring the adequacy of FXIII levels is essential.

Virus-inactivated FXIII concentrates made from human plasma or placenta are an improvement over traditional products. Human factor XIII concentrate (Corifact) is approved for prophylaxis in congenital FXIII deficiency by the US Food and Drug Administration (FDA). It is marketed under the brand name Fibrogammin P in Europe, South America, South Africa, and Japan. A second FXIII concentrate (Bio Products Laboratory, Elstree, Hertfordshire, UK) is available on a per-patient request.

Factor XIII A-subunit, recombinant (Tretten) was approved by the FDA in December 2013.[102] Approval was based on results from a clinical study that demonstrated the safety and efficacy of rFXIII A-subunit. The phase 3 trial included 41 patients and showed that preventive treatment with monthly 35 IU/kg rFXIII A-subunit injections significantly decreased the number of treatment-requiring bleeding episodes, compared with an historic control group of individuals who did not receive routine FXIII infusions, .[103]

The long half-life of FXIII of 6-19 days and the hemostatic efficacy of even small amounts of FXIII of approximately 5% allow replacement therapy to be administered every 4-6 weeks. An FFP dose of 2-3 mL/kg may be effective for up to 4 weeks.[104, 105] The dose of concentrate in adults with deficiency is 35 U/kg every 4 weeks.[106, 107, 108]

A paucity of data exists concerning the pediatric population. Hemostatic evaluation following a head trauma-induced large subcutaneous hematoma associated with recurrent postsurgical bleeding led to a diagnosis of severe FXIII deficiency in a 22-month-old boy. Following initial therapy, subsequent replacement with an FXIII concentrate dose of 50 U/kg every 5 weeks was sufficient to prevent rebleeding and allow healing.[109] Serial monitoring of actual levels achieved is important in children to determine adequacy of any therapy. For more information, see Pediatric Factor XIII Deficiency.

Minor bleeding, as from cuts and abrasions, may respond to conservative measures, such as pressure, ice, and use of antifibrinolytic drugs. Avoidance of trauma and nonsteroidal anti-inflammatory drugs (NSAIDs) is helpful in reducing bleeding events.

Several reports exist of the use of FXIII in unusual circumstances. Note the following:

Patients with acquired inhibitors to FXIII should be treated using well-established principles of therapy. Note the following:

To date, prophylactic factor replacement has been undertaken mainly in patients with intracranial bleeding or recurrent miscarriages caused by severe FXIII deficiency. Successful prevention of recurrent joint bleeds also has been accomplished using periodic transfusions of FFP and cryoprecipitate.[3] FFP can be administered in a dose of 2-3 mL/kg every 4 weeks.

A literature review of bleeding risks and reproduction among patients with severe FXIII deficiency suggests that patients with clinically significant bleeding should start receiving factor replacement therapy in childhood to reduce early mortality from hemorrhages and to allow patients to reach adulthood. During pregnancy, monthly replacement was found to be effective in preventing miscarriages.[114] However, both short-term benefits and potential long-term adverse consequences of prophylactic use of these products must be discussed, with full patient participation in all decision making.

Advances in the types of available products improve care. Addition of Tween 20 makes a reduction of the generation of soluble and insoluble aggregates of rFXIII possible when rFXIII is subjected to freezing and thawing or agitation.[115] Another advance in the technology relates to solving problems faced during freeze-drying and storing the dry solid. Improvement in storage stability of therapeutic proteins has obvious advantages for both storage and transport.[116]

Pooled plasma treated with solvent-detergent (PLAS+SD) is available to treat any condition in which FFP typically is used and for which no factor concentrate is available. Viral inactivation using the solvent-detergent (SD) process has been used in preparation of coagulation factor concentrates in the past. In vitro treatment of donor plasma with 1% of the solvent tri(n- butyl) phosphate (TNBP) and 1% of the detergent Triton X-100 leads to significant inactivation of a broad spectrum of lipid-enveloped viruses. Note the following:

Antifibrinolytic agents are not used commonly to treat patients with FXIII deficiency but may be used as ancillary therapy. The hemostatic plug formed in the presence of adequate levels of FXIII at the time of surgical trauma (as with dental procedures or with mucosal bleeding) can be preserved by inhibiting fibrinolysis with ε-aminocaproic acid (EACA; Amicar) or trans-p- aminomethyl-cyclohexane carboxylic acid (AMCA; also termed tranexamic acid; Cyklokapron) administered orally or, if needed, intravenously.

EACA has been administered in a dose of 5 g orally or intravenously slowly prior to the surgical procedure, along with a dose of the appropriate FXIII replacement. This is followed by a maintenance dose of 1 g/h postoperatively until it is appropriate to start tapering the dose over the next several days.

AMCA is administered in a dose of 1.5 g intravenously every 6-8 hours and tapered, as needed. Hhowever, it is not available in the United States.

Antifibrinolytic agents also can be used as a mouthwash for oral bleeding and have been used to stop local intracavitary oozing.

Antifibrinolytic agents are contraindicated in patients with hematuria originating from above the bladder, because of the possible risk of development of a firm occluding clot in the ureters when administered simultaneously with factor replacement (however, many urologists routinely use EACA irrigations after prostatic and bladder surgery)

Antifibrinolytic agents are not useful in the treatment of joint bleeding (see Hemophilia A for more information).

In recent years, the use of NSAIDs to relieve pain has increased in patients with bleeding disorders. Although they provide relief from inflammatory pain, patients experience increased GI tract or other bleeding because of the impact of the drugs on primary hemostasis, and they require additional FXIII replacement to control bleeding. The problem is magnified by the availability of over-the-counter NSAID pain relievers. Non-NSAIDs, such as acetaminophen and codeine-type analgesics, are much less effective, and some are addictive.

Routine dental care is of the utmost importance in maintaining dental hygiene. Other routine care, such as mammography in women older than 50 years or colonoscopy for patients older than 50 years, must be provided as in nonbleeding patients.

Gene therapy has not been used as a treatment modality in patients with FXIII deficiency thus far. However, the reader is referred to a review of gene therapy in the hemophilias and other blood diseases.[117]

Surgical Care

All elective procedures require proper perioperative management. Note the following:

Consultations

A hematologist, orthopedist, physical therapist, dentist, social worker, psychologist, infectious disease specialist, gastroenterologist/hepatologist, geneticist, and an appropriately equipped special laboratory all play important roles in providing optimal care for patients with FXIII deficiency and their families.

The efforts of the National Hemophilia Foundation and its regional chapters must be recognized in helping to educate patients, assist service providers, foster dialog regarding problems and solutions among patients with bleeding disorders, and improve conditions for the entire community through support of legislation.

Diet

A healthy and nutritional diet should be encouraged.

Activity

Appropriate physical activity and physical therapy must be encouraged to maintain and preserve muscle function.

Medication Summary

Factor XIII (FXIII) replacement therapy can be accomplished with FXIII concentrate; with fresh frozen plasma (FFP) or solvent/detergent-treated pooled plasma (Octaplas); or with cryoprecipitate. FXIII concentrate, human (Corifact) is commercially available in the United States. In December 2013, a recombinant FXIII A-subunit product (Tretten) was approved for preventing bleeding episodes in patients with congenital FXIII-a subunit deficiency.[72]

Dosing of cryoprecipitate is empiric, since no standardized amount of FXIII exists for cryoprecipitate. Repeat dosing should be guided by the adequacy of a prior dose as determined by FXIII assays.

Traditionally, FFP has been the source of factors for the treatment of coagulation factor deficiencies for which no concentrates are available, as was once the case with FXIII deficiency. An FFP dose of 2-3 mL/kg every 4 weeks has been used for replacement therapy under steady-state conditions. Higher risks of virally transmitted illnesses remain among patients who are recipients of multiple units of FFP. The greater degree of viral safety assured by this treatment has led to the exclusive use of solvent/detergent-treated pooled plasma instead of FFP in some countries (Norway and Belgium).

Solvent/detergent-treated pooled plasma is ABO blood type specific and offers more protection to patients than is found in standard FFP. As a result of treatment with 1% tri(n- butyl)phosphate (or TNBP as the solvent) and 1% Triton X-100 (as the detergent), lipid-enveloped viruses (eg, HIV, hepatitis B and C viruses, Hantavirus, Marburg virus, Ebola virus) are disrupted and killed in significant numbers. The resulting fragments are inactive and cannot replicate or cause disease. Patients with FXIII deficiency have been specifically treated successfully with this product.[119]

Adverse reactions include minor allergic reactions, which respond to antihistamines, and volume overload. Rarely, citrate toxicity, hypothermia, and other metabolic problems arise if large volumes are used rapidly. Noncardiogenic pulmonary edema can occur. Antibody-induced positive results to the direct antiglobulin test and hemolysis also may occur rarely. This product is contraindicated in patients with known IgA deficiency.

Careful screening of blood donors and viral testing of donated blood (HBV surface antigen, antibody to HBV core antigen, HCV, antibody to HIV-1 and HIV-2, HIV p24 antigen, antibodies to human T-cell leukemia virus [HTLV] types I and II, and screening for elevated levels of alanine aminotransferase [ALT]) have improved safety of blood products, but risks remain for a variety of reasons including failure to detect infections during the "window" or incubation period before currently available test results become positive.

Other types of infections in which screening currently is not performed, tests are not available, or the presence of infection is unknown continue to cause concerns. Some of the emerging pathogens previously referred to include HIV-2, HIV type O, hepatitis G, TTV, human herpesvirus 8, the SEN family of viruses, and prions causing Creutzfeldt Jacob disease [CJD] and nvCJD.[120, 121, 122]

Newer emerging technologies, such as those using nucleic acid chemistry, are being used to inactivate viruses, bacteria, and parasites and to attempt to remove prions, thus making blood and blood components safer than they are currently. These newer technologies attempt to preserve clinically useful components of blood while improving its safety. Potentially, these methodologies could be used to improve the safety of a wide variety of products.

Adjunctive role of inhibitors of fibrinolysis: Recognition of the importance of the lysine-binding sites in various interactions in the fibrinolytic pathway led to the synthesis of lysine analogs such as epsilon aminocaproic acid (EACA) and tranexamic acid (AMCA). These synthetic lysine analogs induce a conformational change in plasminogen when they bind to its lysine-binding site; plasminogen has the shape of a prolate ellipsoid after EACA binds to it. The bound plasminogen-EACA elongates into a long structure in which the interaction between the parts of plasminogen, as they existed, are lost. In vivo, the structures probably prevent plasminogen activation and, in large doses, bind plasmin, thereby preventing it from binding to its substrate fibrin. In the plasminogen-EACA binding sites, the tightest binding is to kringle 1 followed by kringles 4 and 5. The interaction with kringle 2 is weak, and kringle 3 does not interact at all.

A model of the structure of kringle 4 shows that the shallow trough formed by the hydrophobic amino acids is surrounded by positively and negatively charged amino acids at a distance ideal for EACA interaction. For further details regarding these interactions, please see Bachmann, 2001.[123] EACA is the most widely used antifibrinolytic drug in the United States. The minimal dose needed to inhibit either normal or excessive fibrinolysis is unknown. EACA is absorbed well orally, and 50% is excreted in the urine within 24 hours.

Generally, an initial loading dose is followed by a maintenance dose to adequately inhibit fibrinolysis until excess bleeding is controlled. Then, the maintenance dose is tapered gradually until it can be discontinued. Rarely, myopathy and muscle necrosis can develop. Lower doses are adequate when bleeding involves the urinary tract, since drug concentrations are 75- to 100-fold higher in urine than in plasma.

AMCA also is excreted rapidly in the urine, with more than 90% excreted within 24 hours. However, its antifibrinolytic effect lasts longer than EACA. AMCA inhibits fibrinolysis at lower plasma concentrations, although its serum half-life is similar to that of EACA. Therefore, AMCA can be administered less frequently and at lower doses. EACA and AMCA doses must be reduced when renal failure is present.

Aprotinin (Trasylol), a third antifibrinolytic drug obtained from bovine lung, is a nonhuman protein inhibitor of several serine proteases, including plasmin. It is approved by the FDA for use in patients undergoing open heart surgery to reduce operative blood loss. Aprotinin administration also has reduced blood loss and transfusion requirements in patients undergoing orthotopic liver transplantation or in patients undergoing elective resection of a solitary liver metastasis originating from colon cancer. Aprotinin is the most expensive of the 3 drugs discussed here. Aprotinin is now only available via a limited-access protocol. Fergusson et al reported an increased risk for death compared with tranexamic acid or aminocaproic acid in high-risk cardiac surgery.[124]

Factor XIII A-subunit, recombinant (Tretten)

Clinical Context:  FXIII A-subunit recombinant is a human factor XIII-A2 homodimer composed of 2 FXIII A-subunits. It is indicated for routine prevention of bleeding in patients with congenital Factor XIII A-subunit deficiency.

Factor XIII concentrate, human (Corifact)

Clinical Context:  Temporarily replaces missing clotting factor XIII which corrects and/or prevents bleeding. It is indicated for routine prophylactic treatment of congenital factor XIII (FXIII) deficiency.

Class Summary

FXIII is the terminal enzyme in the blood coagulation cascade; when activated by thrombin at the site of vessel wall injury, FXIII plays an important role in the maintenance of hemostasis through cross-linking of fibrin and other proteins in the fibrin clot. FXIII is a proenzyme that is activated, in the presence of calcium ion, by thrombin cleavage of the A-subunit to become activated FXIII (FXIIIa). It promotes cross-linking of fibrin during coagulation and is essential to the physiological protection of the clot against fibrinolysis.

Fresh frozen plasma (FFP, Octaplas)

Clinical Context:  Plasma is the fluid compartment of blood containing the soluble clotting factors. Octaplas is a solvent detergent treated, pooled FFP.

Class Summary

Use antifibrinolytic agents with fresh frozen plasma (FFP) replacement for minor surgical procedures (eg, dental extractions or sinus surgery) so that the surgery can be accomplished on an outpatient basis with the use of a single dose of product.

Concern remains regarding the possible relationship to acute thrombotic events, although a causal relationship is being questioned because the underlying disease state determines the site and extent of thrombosis.

Aminocaproic acid (Amicar)

Clinical Context:  EACA diminishes bleeding by inhibiting lysis of hemostatic plugs. Can be used PO or IV.

Tranexamic acid injection (Cyklokapron)

Clinical Context:  Diminishes bleeding by inhibiting fibrinolysis of hemostatic plug. A PO form is also available.

Class Summary

These agents are used as an ancillary measure and to diminish bleeding.

Further Outpatient Care

Clinic-supervised outpatient care is an extremely important part of treatment. Complete annual physical examinations and laboratory testing for inhibitors, hepatitis, and HIV infection, as well as other tests, should be performed as needed. As in persons without hemophilia, routine care should be provided to patients with FXIII deficiency, including examination of stool for blood, rectal examination, colonoscopy, prostate-specific antigen determinations, mammography, and dental care. Physical therapy may be needed for the long-term care of affected joints. Prophylactic care includes vaccination for HAV[125] and HBV and other routine vaccines.

Notify the local chapter of the National Hemophilia Foundation regarding the patient so that proper statistics can be provided to the appropriate agencies for adequate federal and state funding of patient care.

Provide counseling and classes to encourage questions and to help solve problems, such as possible ways to avoid transmission of HIV to an uninfected spouse and to children.[126]

An appointment with a psychosocial worker at the time of counseling ensures that other psychological, social, and economic support is provided.

Further Inpatient Care

Patients should be hospitalized for serious complications, such as severe bleeding, or for major surgical procedures, all which require complex interdisciplinary care including pharmacy and laboratory support. Constant clinical evaluation and laboratory monitoring ensure adequacy of product replacement, pain relief, and other supportive care. The hematologist must be centrally involved to coordinate care.

Inpatient & Outpatient Medications

Patients should avoid acetylsalicylic acid, nonsteroidal anti-inflammatory drugs (NSAIDs), and any over-the-counter herbal medications that can increase bleeding diathesis.

Transfer

If a qualified hematologist and laboratory personnel with expertise are available, patients may be cared for in a setting close to home. Laboratory testing provided by community hospitals has been improved by the existence of commercial referral laboratories.

Federal and state funding for programs may be available through a medical center. Costs of care are much higher at tertiary medical centers.

Deterrence/Prevention

Avoidance of high-risk activities (eg, boxing, motorbike riding) and NSAIDs reduces the frequency of bleeding.

Avoidance of alcohol helps protect liver function in patients with hepatitis.

Primary prophylaxis is the best way to prevent recurrent CNS bleeding, recurrent miscarriages or, rarely, recurrent joint bleeding. Joint replacement may be needed in the older patient with severe arthropathy. 

Carcao et al reported that in patients with congenital FXIII A-subunit deficiency, prophylaxis with recombinant factor XIII-A2 (rFXIII-A2) provided sufficient hemostatic coverage for minor surgery without the need for additional FXIII therapy. Minor surgery was performed as long as 10 to 21 days after the last dose of rFXIII-A2.[127]

HAV and HBV vaccines should be administered. Other routine vaccinations, such as those for influenza and pneumonia, should be provided as in other persons.

Complications

Recurrent CNS bleeding is a major problem requiring prophylactic transfusions. Infections, particularly HIV, AIDS, and chronic hepatitis, can lead to death. Interferon alfa has been used to treat chronic viral hepatitis. Multidrug cocktails are used to treat HIV/AIDS, but protease inhibitors can increase risk of bleeding. Some over-the-counter herbal remedies increase the risk of bleeding.

Viral safety in products derived from plasma is ensured through several techniques, ie, heating, pasteurization, SD treatment, and monoclonal antibody purification. These procedures currently free products from HIV and HCV (lipid-enveloped viruses) but do not solve the problem of transmission of non–lipid-enveloped viruses such as HAV, parvovirus B19, and other transfusion-transmitted viruses (TTV).

Even with recombinant products, a possibility exists of contamination with pathogens previously unknown, including new murine viruses. One report shows the presence of TTV in first-generation recombinant products, due to the use of human serum albumin that is contaminated with TTV.[122] Thus, virus-induced illnesses of concern include hepatitis viruses A-E, GB virus C (or hepatitis G virus), the SEN family of viruses,[121] and human herpesvirus 8,[120] all of which constitute emerging pathogens related to transfusion-transmitted illnesses.

Potential transmission of prions causing Creutzfeldt-Jakob disease (CJD) or its variant form (vCJD) in recipients of blood products was a serious concern early in this century. However, no individual with hemophilia nor any other blood product recipient in the United States is known to have developed CJD. A United Kingdom study found that as of May 2015, no new cases of transfusion-associated vCJD had occurred since 2007 and there was no evidence of transfusion transmission of sporadic CJD.[128] A sensitive and specific blood test for vCJD has been developed and has entered clinical use; it could be used to screen blood supplies.[129]

The presence of inhibitors adds another layer of complexity when alloantibodies develop as a consequence of transfusion of blood products. Spontaneous disappearance is a typical feature of autoantibodies, presumably as a response to removal of the antigenic stimulus. Bleeding associated with inhibitors can be life threatening and requires complex care.

Severe economic and emotional problems occur as a result of the recurrent nature of the bleeding.

Complex psychiatric issues arise in the treatment of patients with HIV/AIDS.[126]

Prognosis

Prognosis depends on the types of complications that develop, on the type of replacement product the patient has received, and on the viral infections that the patient has accumulated over the years. Newly diagnosed patients should, whenever possible, receive purer products to ensure maximum safety.

The presence of inhibitors in patients poses a serious therapeutic challenge, and, currently, surgery should be considered only as a lifesaving measure.

Patient Education

Encourage patients to register with the local chapter of the National Hemophilia Foundation and to attend educational seminars. Provide one-on-one discussions of issues with patients and family members. Early and complete genetic testing can help families plan future pregnancies.

For patient education information, see Bleeding Disorders.

What is factor XIII (FXIII) deficiency?How does the site of bleeding affect the physical findings of factor XIII (FXIII) deficiency?What are the signs and symptoms of factor XIII (FXIII) deficiency?What are the signs and symptoms of joint bleeding in factor XIII (FXIII) deficiency?Which lab tests are performed in the workup of factor XIII (FXIII) deficiency?What is included in the qualitative screening test for severe factor XIII (FXIII) deficiency?What is the role of quantitative testing in the workup of factor XIII (FXIII) deficiency?What is included in inhibitor testing for factor XIII (FXIII) deficiency?How is factor XIII (FXIII) deficiency diagnosed prenatally?What is the role of FXIII replacement in factor XIII (FXIII) deficiency?Which replacement factor XIII concentrates are used in the treatment of factor XIII (FXIII) deficiency?How is factor XIII (FXIII) deficiency treated in patients with factor XIII inhibitors?How was factor XIII (FXIII) deficiency first recognized?What is the structure, production, and half-life of factor XIII (FXIII)?What is the role of activation in the pathophysiology of factor XIII (FXIII) deficiency?What is the role of factor XIII (FXIII) cross-linking and resistance to lysis in the pathophysiology of factor XIII (FXIII) deficiency?Which factors affect the level and activity of factor XIII (FXIII) in the pathophysiology of factor XIII (FXIII) deficiency?How does factor XIII (FXIII) deficiency affect pregnancy outcomes?How does factor XIII (FXIII) deficiency affect wound healing?What is the role of nitric oxide (NO) in the pathophysiology of factor XIII (FXIII) deficiency?What is the role of venom and toxins in the pathophysiology of factor XIII (FXIII) deficiency?What is the role of simvastatin in the pathophysiology of factor XIII (FXIII) deficiency?What is the role of synthetic inhibitors in the pathophysiology of factor XIII (FXIII) deficiency?What is the role of plasma and tissue transglutaminases in the pathophysiology of factor XIII (FXIII) deficiency?What is the role of factor XIII (FXIII) deficiency in the pathophysiology of degenerative brain disorders?What is the role of thrombin-activatable fibrinolytic inhibitor in the pathophysiology of factor XIII (FXIII) deficiency?What is the role of cell surface–directed hemostasis, in the pathophysiology of factor XIII (FXIII) deficiency?What is the prevalence of factor XIII (FXIII) deficiency in the US?What is the global prevalence of factor XIII (FXIII) deficiency?What is the mortality and morbidity associated with factor XIII (FXIII) deficiency?Which patient groups have the highest prevalence of factor XIII (FXIII) deficiency?Which clinical history findings are characteristic of factor XIII (FXIII) deficiency?Which physical findings are characteristic of factor XIII (FXIII) deficiency?What causes factor XIII (FXIII) deficiency?What is the role of the Val34Leu mutation in the etiology of factor XIII (FXIII) deficiency?What is the role of genetic polymorphisms in the etiology of factor XIII (FXIII) deficiency?What is the role of disorders of fibrin stabilization in the etiology of factor XIII (FXIII) deficiency?Which conditions are included in the differential diagnoses of factor XIII (FXIII) deficiency?Which disorders are included in the differential diagnoses of acquired factor XIII (FXIII) deficiency?Which factor XIII (FXII) inhibitors are included in the differential diagnoses of factor XIII (FXIII) deficiency?What are the differential diagnoses for Factor XIII Deficiency?What is included in the initial workup of factor XIII (FXIII) deficiency?Which findings on a qualitative screening test suggest factor XIII (FXIII) deficiency?What is the stepped approach to diagnostic testing of factor XIII (FXIII) deficiency?How is a diagnosis of acquired factor XIII (FXIII) deficiency confirmed?Which tests are performed in the prenatal workup of factor XIII (FXIII) deficiency?What is the role of liver function tests in the workup of factor XIII (FXIII) deficiency?Which imaging studies are included in the workup of factor XIII (FXIII) deficiency?When is ECG performed in the workup of factor XIII (FXIII) deficiency?What procedures are included in the prenatal workup of factor XIII (FXIII) deficiency?What is the role of colonoscopy and endoscopy in the workup of factor XIII (FXIII) deficiency?How is factor XIII (FXIII) deficiency treated?What are the reported uses of factor XIII (FXIII) in unusual circumstances?How is acquired factor XIII (FXIII) deficiency treated?What is the role of prophylactic factor replacement in the treatment of factor XIII (FXIII) deficiency?What advancements have been made in the storage and transport of factors used in factor XIII (FXIII) deficiency treatment?What is the role of PLAS+S in the treatment of factor XIII (FXIII) deficiency?What is the role of antifibrinolytic agents in the treatment of factor XIII (FXIII) deficiency?What is the role of NSAIDs in the treatment of factor XIII (FXIII) deficiency?Which routine preventive care should be provided to patients with factor XIII (FXIII) deficiency?What is the role of gene therapy in the treatment of factor XIII (FXIII) deficiency?What is included in perioperative management of patients with factor XIII (FXIII) deficiency undergoing routine surgical procedures?Which specialist consultations are beneficial to patients with factor XIII (FXIII) deficiency?Which dietary modifications are used in the treatment of factor XIII (FXIII) deficiency?Which activity modifications are used in the treatment of factor XIII (FXIII) deficiency?What is the role of medications in the treatment of factor XIII (FXIII) deficiency?Which medications in the drug class Antifibrinolytic agents are used in the treatment of Factor XIII Deficiency?Which medications in the drug class Blood Products are used in the treatment of Factor XIII Deficiency?Which medications in the drug class Clotting factors are used in the treatment of Factor XIII Deficiency?What is included in long-term monitoring of factor XIII (FXIII) deficiency?When is inpatient care indicated in the treatment of factor XIII (FXIII) deficiency?Which medications are contraindicated in factor XIII (FXIII) deficiency?When is patient transfer indicated for the treatment of factor XIII (FXIII) deficiency?How are exacerbations and complications of factor XIII (FXIII) deficiency prevented?What are the possible complications of factor XIII (FXIII) deficiency?What is the prognosis of factor XIII (FXIII) deficiency?What is included in the patient education about treatment of factor XIII (FXIII) deficiency?

Author

Robert A Schwartz, MD, MPH, Professor and Head of Dermatology, Professor of Pathology, Professor of Pediatrics, Professor of Medicine, Rutgers New Jersey Medical School

Disclosure: Nothing to disclose.

Coauthor(s)

Elzbieta Klujszo, MD, Head of Department of Dermatology, Wojewodzki Szpital Zespolony, Kielce

Disclosure: Nothing to disclose.

Pere Gascon, MD, PhD, Professor and Director, Division of Medical Oncology, Institute of Hematology and Medical Oncology, IDIBAPS, University of Barcelona Faculty of Medicine, Spain

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Ronald A Sacher, MBBCh, FRCPC, DTM&H, Professor of Internal Medicine and Pathology, Director, Hoxworth Blood Center, University of Cincinnati Academic Health Center

Disclosure: Nothing to disclose.

Chief Editor

Perumal Thiagarajan, MD, Professor, Department of Pathology and Medicine, Baylor College of Medicine; Director, Transfusion Medicine and Hematology Laboratory, Michael E DeBakey Veterans Affairs Medical Center

Disclosure: Nothing to disclose.

Additional Contributors

Paul Schick, MD, Emeritus Professor, Department of Internal Medicine, Jefferson Medical College of Thomas Jefferson University; Research Professor, Department of Internal Medicine, Drexel University College of Medicine; Adjunct Professor of Medicine, Lankenau Hospital

Disclosure: Nothing to disclose.

Acknowledgements

Rajalaxmi McKenna, MD, FACP Southwest Medical Consultants, SC, Department of Medicine, Good Samaritan Hospital, Advocate Health Systems

Rajalaxmi McKenna, MD, FACP is a member of the following medical societies: American Society of Clinical Oncology, American Society of Hematology, and International Society on Thrombosis and Haemostasis

Disclosure: Nothing to disclose.

References

  1. Shanbhag S, Ghosh K, Shetty S. Genetic basis of severe factor XIII deficiency in a large cohort of Indian patients: Identification of fourteen novel mutations. Blood Cells Mol Dis. 2016 Mar. 57:81-4. [View Abstract]
  2. Muszbek L, Pénzes K, Katona É. Auto- and alloantibodies against factor XIII: laboratory diagnosis and clinical consequences. J Thromb Haemost. 2018 Feb 20. [View Abstract]
  3. Thakker S, McGehee W, Quismorio FP Jr. Arthropathy associated with factor XIII deficiency. Arthritis Rheum. 1986 Jun. 29(6):808-11. [View Abstract]
  4. McKenna R. Abnormal coagulation in the postoperative period contributing to excessive bleeding. Med Clin North Am. 2001 Sep. 85(5):1277-310, viii. [View Abstract]
  5. Wilmer M, Rudin K, Kolde H, et al. Evaluation of a sensitive colorimetric FXIII incorporation assay. Effects of FXIII Val34Leu, plasma fibrinogen concentration and congenital FXIII deficiency. Thromb Res. 2001 Apr 1. 102(1):81-91. [View Abstract]
  6. Laki K, Lóránd L. On the Solubility of Fibrin Clots. Science. 1948 Sep 10. 108(2802):280. [View Abstract]
  7. Duckert F, Jung E, Sherling DH. An undescribed congenital haemorrhagic diathesis probably due to fibrin stabilizing factor deficiency. Thromb Diath Haemorrh. 1960. 5:179.
  8. Duckert F. Documentation of the plasma factor XIII deficiency in man. Ann N Y Acad Sci. 1972 Dec 8. 202:190-9. [View Abstract]
  9. Laki K. Our ancient heritage in blood clotting and some of its consequences. Ann N Y Acad Sci. 1972 Dec 8. 202:297-307. [View Abstract]
  10. Reitsma PH. Genetic principles underlying disorders of procoagulant and anticoagulant proteins. Coleman RW, Hirsh J, Marder VJ, et al, eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. Philadelphia, Pa: Lippincott Williams & Wilkins; 2001. 59-87.
  11. Loewy AG, McDonagh J, Mikkola H, et al. Structure and function of F XIII. Coleman RW, Hirsh J, Marder VJ, et al, eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. Philadelphia, Pa: Lippincott Williams & Wilkins; 2001. 233-48.
  12. Cushman M, O'Meara ES, Folsom AR, Heckbert SR. Coagulation factors IX through XIII and the risk of future venous thrombosis: the Longitudinal Investigation of Thromboembolism Etiology. Blood. 2009 Oct 1. 114(14):2878-83. [View Abstract]
  13. Hoppe B, Tolou F, Dörner T, Kiesewetter H, Salama A. Gene polymorphisms implicated in influencing susceptibility to venous and arterial thromboembolism: frequency distribution in a healthy German population. Thromb Haemost. 2006 Oct. 96(4):465-70. [View Abstract]
  14. Wells PS, Anderson JL, Rodger MA, Carson N, Grimwood RL, Doucette SP. The factor XIII Val34Leu polymorphism: is it protective against idiopathic venous thromboembolism?. Blood Coagul Fibrinolysis. 2006 Oct. 17(7):533-8. [View Abstract]
  15. Shafey M, Anderson JL, Scarvelis D, Doucette SP, Gagnon F, Wells PS. Factor XIII Val34Leu variant and the risk of myocardial infarction: a meta-analysis. Thromb Haemost. 2007 Apr. 97(4):635-41. [View Abstract]
  16. Shin JI, Lee JS. Severe gastrointestinal vasculitis in Henoch-Schoenlein purpura: pathophysiologic mechanisms, the diagnostic value of factor XIII, and therapeutic options. Eur J Pediatr. 2007 Nov. 166(11):1199-200; author reply 1201. [View Abstract]
  17. Shin JI, Lee JS. Could measurement of factor XIII level detect the vasculitic process of Henoch-Schönlein purpura without skin rash?. Acta Paediatr. 2008 Apr. 97(4):395. [View Abstract]
  18. Kuroda K, Tajima S. Proliferation of HSP47-positive skin fibroblasts in dermatofibroma. J Cutan Pathol. 2008 Jan. 35(1):21-6. [View Abstract]
  19. Dorgalaleh A, Tabibian S, Shams M, Majid G, Naderi M, Casini A, et al. A Unique Factor XIII Mutation in Southeastern Iran with an Unexpectedly High Prevalence: Khash Factor XIII. Semin Thromb Hemost. 2019 Feb. 45 (1):43-49. [View Abstract]
  20. Ichinose A. Factor XIII is a key molecule at the intersection of coagulation and fibrinolysis as well as inflammation and infection control. Int J Hematol. 2012 Apr. 95(4):362-70. [View Abstract]
  21. Schroeder V, Kohler HP. Factor XIII Deficiency: An Update. Semin Thromb Hemost. 2013 Sep. 39(6):632-41. [View Abstract]
  22. Lorand L. Factor XIII: structure, activation, and interactions with fibrinogen and fibrin. Ann N Y Acad Sci. 2001. 936:291-311. [View Abstract]
  23. Siebenlist KR, Meh DA, Mosesson MW. Plasma factor XIII binds specifically to fibrinogen molecules containing gamma chains. Biochemistry. 1996 Aug 13. 35(32):10448-53. [View Abstract]
  24. Greenberg CS, Shuman MA. The zymogen forms of blood coagulation factor XIII bind specifically to fibrinogen. J Biol Chem. 1982 Jun 10. 257(11):6096-101. [View Abstract]
  25. Mosesson M. Hereditary abnormalities of fibrinogen. Beutler E, Lichtman M, Coller BS, et al, eds. Williams Hematology. New York, NY: McGraw-Hill; 2001. 1659-71.
  26. Jenner L, Husted L, Thirup S, Sottrup-Jensen L, Nyborg J. Crystal structure of the receptor-binding domain of alpha 2-macroglobulin. Structure. 1998 May 15. 6(5):595-604. [View Abstract]
  27. Nagy B Jr, Simon Z, Bagoly Z, Muszbek L, Kappelmayer J. Binding of plasma factor XIII to thrombin-receptor activated human platelets. Thromb Haemost. 2009 Jul. 102(1):83-9. [View Abstract]
  28. Monteiro MR, Murphy EE, Galaria NA, Whitaker-Menezes D, Murphy GF. Cytological alterations in dermal dendrocytes in vitro: evidence for transformation to a non-dendritic phenotype. Br J Dermatol. 2000 Jul. 143(1):84-90. [View Abstract]
  29. Lorand L. Sol Sherry Lecture in Thrombosis : research on clot stabilization provides clues for improving thrombolytic therapies. Arterioscler Thromb Vasc Biol. 2000 Jan. 20(1):2-9. [View Abstract]
  30. Noguchi K, Ishikawa K, Yokoyama Ki, Ohtsuka T, Nio N, Suzuki E. Crystal structure of red sea bream transglutaminase. J Biol Chem. 2001 Apr 13. 276(15):12055-9. [View Abstract]
  31. Andersen MD, Kjalke M, Bang S, et al. Coagulation factor XIII variants with altered thrombin activation rates. Biol Chem. 2009 Dec. 390(12):1279-83. [View Abstract]
  32. Fox BA, Yee VC, Pedersen LC, et al. Identification of the calcium binding site and a novel ytterbium site in blood coagulation factor XIII by x-ray crystallography. J Biol Chem. 1999 Feb 19. 274(8):4917-23. [View Abstract]
  33. Casadio R, Polverini E, Mariani P, et al. The structural basis for the regulation of tissue transglutaminase by calcium ions. Eur J Biochem. 1999 Jun. 262(3):672-9. [View Abstract]
  34. Hevessy Z, Haramura G, Boda Z, Udvardy M, Muszbek L. Promotion of the crosslinking of fibrin and alpha 2-antiplasmin by platelets. Thromb Haemost. 1996 Jan. 75(1):161-7. [View Abstract]
  35. Moaddel M, Falls LA, Farrell DH. The role of gamma A/gamma ' fibrinogen in plasma factor XIII activation. J Biol Chem. 2000 Oct 13. 275(41):32135-40. [View Abstract]
  36. Weiss MS, Metzner HJ, Hilgenfeld R. Two non-proline cis peptide bonds may be important for factor XIII function. FEBS Lett. 1998 Feb 27. 423(3):291-6. [View Abstract]
  37. Jámbor C, Reul V, Schnider TW, Degiacomi P, Metzner H, Korte WC. In vitro inhibition of factor XIII retards clot formation, reduces clot firmness, and increases fibrinolytic effects in whole blood. Anesth Analg. 2009 Oct. 109(4):1023-8. [View Abstract]
  38. Lorand L, Urayama T, Atencio AC, Hsia DY. Inheritance of deficiency of fibrin-stabilizing factor (factor 13). Am J Hum Genet. 1970 Jan. 22(1):89-95. [View Abstract]
  39. Anwar R, Gallivan L, Edmonds SD, Markham AF. Genotype/phenotype correlations for coagulation factor XIII: specific normal polymorphisms are associated with high or low factor XIII specific activity. Blood. 1999 Feb 1. 93(3):897-905. [View Abstract]
  40. Kangsadalampai S, Board PG. The Val34Leu polymorphism in the A subunit of coagulation factor XIII contributes to the large normal range in activity and demonstrates that the activation peptide plays a role in catalytic activity. Blood. 1998 Oct 15. 92(8):2766-70. [View Abstract]
  41. Saha N, Aston CE, Low PS, Kamboh MI. Racial and genetic determinants of plasma factor XIII activity. Genet Epidemiol. 2000 Dec. 19(4):440-55. [View Abstract]
  42. van Wersch JW, Vooijs ME, Ubachs JM. Coagulation factor XIII in pregnant smokers and non-smokers. Int J Clin Lab Res. 1997. 27(1):68-71. [View Abstract]
  43. Asahina T, Kobayashi T, Okada Y, et al. Studies on the role of adhesive proteins in maintaining pregnancy. Horm Res. 1998. 50 Suppl 2:37-45. [View Abstract]
  44. Asahina T, Kobayashi T, Okada Y, Goto J, Terao T. Maternal blood coagulation factor XIII is associated with the development of cytotrophoblastic shell. Placenta. 2000 May. 21(4):388-93. [View Abstract]
  45. Anwar R, Miloszewski KJ. Factor XIII deficiency. Br J Haematol. 1999 Dec. 107(3):468-84. [View Abstract]
  46. Noll T, Wozniak G, McCarson K, et al. Effect of factor XIII on endothelial barrier function. J Exp Med. 1999 May 3. 189(9):1373-82. [View Abstract]
  47. Cario E, Goebell H, Dignass AU. Factor XIII modulates intestinal epithelial wound healing in vitro. Scand J Gastroenterol. 1999 May. 34(5):485-90. [View Abstract]
  48. Naito M, Nomura H, Iguchi A, Thompson WD, Smith EB. Effect of crosslinking by factor XIIIa on the migration of vascular smooth muscle cells into fibrin gels. Thromb Res. 1998 May 1. 90(3):111-6. [View Abstract]
  49. Wozniak G, Noll T, Brunner U, Hehrlein FW. Topical treatment of venous ulcer with fibrin stabilizing factor: experimental investigation of effects on vascular permeability. Vasa. 1999 Aug. 28(3):160-3. [View Abstract]
  50. Catani MV, Bernassola F, Rossi A, Melino G. Inhibition of clotting factor XIII activity by nitric oxide. Biochem Biophys Res Commun. 1998 Aug 10. 249(1):275-8. [View Abstract]
  51. Bernassola F, Rossi A, Melino G. Regulation of transglutaminases by nitric oxide. Ann N Y Acad Sci. 1999. 887:83-91. [View Abstract]
  52. Huang QQ, Teng MK, Niu LW. Purification and characterization of two fibrinogen-clotting enzymes from five-pace snake (Agkistrodon acutus) venom. Toxicon. 1999 Jul. 37(7):999-1013. [View Abstract]
  53. Arocha-Pinango CL, Marval E, Guerrero B. Lonomia genus caterpillar toxins: biochemical aspects. Biochimie. 2000 Sep-Oct. 82(9-10):937-42. [View Abstract]
  54. Zavalova L, Lukyanov S, Baskova I, et al. Genes from the medicinal leech (Hirudo medicinalis) coding for unusual enzymes that specifically cleave endo-epsilon (gamma-Glu)-Lys isopeptide bonds and help to dissolve blood clots. Mol Gen Genet. 1996 Nov 27. 253(1-2):20-5. [View Abstract]
  55. Undas A, Brummel KE, Musial J, Mann KG, Szczeklik A. Simvastatin depresses blood clotting by inhibiting activation of prothrombin, factor V, and factor XIII and by enhancing factor Va inactivation. Circulation. 2001 May 8. 103(18):2248-53. [View Abstract]
  56. Finney S, Seale L, Sawyer RT, Wallis RB. Tridegin, a new peptidic inhibitor of factor XIIIa, from the blood- sucking leech Haementeria ghilianii. Biochem J. 1997 Jun 15. 324 ( Pt 3):797-805. [View Abstract]
  57. Lee SY, Chang SK, Lee IH, Kim YM, Chung SI. Depletion of plasma factor XIII prevents disseminated intravascular coagulation-induced organ damage. Thromb Haemost. 2001 Mar. 85(3):464-9. [View Abstract]
  58. Sidelmann JJ, Gram J, Jespersen J, Kluft C. Fibrin clot formation and lysis: basic mechanisms. Semin Thromb Hemost. 2000. 26(6):605-18. [View Abstract]
  59. Takahashi H, Isobe T, Horibe S, et al. Tissue transglutaminase, coagulation factor XIII, and the pro-polypeptide of von Willebrand factor are all ligands for the integrins alpha 9beta 1 and alpha 4beta 1. J Biol Chem. 2000 Aug 4. 275(31):23589-95. [View Abstract]
  60. Salge U, Daubner E, Heiden M, Sietz R. Factor XIII does not stimulate growth of human cultured tumor cells. Blood Coagul Fibrinolysis. 2000 Mar. 11(2):217-8. [View Abstract]
  61. Molnar P, Nemes Z. Hemangiopericytoma of the cerebello-pontine angle. Diagnostic pitfalls and the diagnostic value of the subunit A of factor XIII as a tumor marker. Clin Neuropathol. 1995 Jan-Feb. 14(1):19-24. [View Abstract]
  62. Adany R, Bardos H, Antal M, et al. Factor XIII of blood coagulation as a nuclear crosslinking enzyme. Thromb Haemost. 2001 May. 85(5):845-51. [View Abstract]
  63. Kallberg Y, Gustafsson M, Persson B, Thyberg J, Johansson J. Prediction of amyloid fibril-forming proteins. J Biol Chem. 2001 Apr 20. 276(16):12945-50. [View Abstract]
  64. Bajzar L, Manuel R, Nesheim ME. Purification and characterization of TAFI, a thrombin-activable fibrinolysis inhibitor. J Biol Chem. 1995 Jun 16. 270(24):14477-84. [View Abstract]
  65. Redlitz A, Tan AK, Eaton DL, Plow EF. Plasma carboxypeptidases as regulators of the plasminogen system. J Clin Invest. 1995 Nov. 96(5):2534-8. [View Abstract]
  66. Bajzar L, Nesheim ME, Tracy PB. The profibrinolytic effect of activated protein C in clots formed from plasma is TAFI-dependent. Blood. 1996 Sep 15. 88(6):2093-100. [View Abstract]
  67. Hoffman M, Monroe DM 3rd. A cell-based model of hemostasis. Thromb Haemost. 2001 Jun. 85(6):958-65. [View Abstract]
  68. HGMD. Human Gene Mutation Database at the Institute of Medical Genetics in Cardiff. Available at: http://www.uwcm.ac.uk//uwcm/mg/hgmd0.html. Accessed April 9, 2002.
  69. Dorgalaleh A, Assadollahi V, Tabibian S, Shamsizadeh M. Molecular Basis of Congenital Factor XIII Deficiency in Iran. Clin Appl Thromb Hemost. 2016 Nov 21. [View Abstract]
  70. Doncarli A, Demiguel V, Guseva Canu I, Goulet V, Bayart S, Calvez T, et al. FranceCoag: a 22-year prospective follow-up of the national French cohort of patients with inherited bleeding disorders. Eur J Epidemiol. 2019 May. 34 (5):521-532. [View Abstract]
  71. Attié-Castro FA, Zago MA, Lavinha J, et al. Ethnic heterogeneity of the factor XIII Val34Leu polymorphism. Thromb Haemost. 2000 Oct. 84(4):601-3. [View Abstract]
  72. FDA approves Tretten to treat rare genetic clotting disorder. U.S. Food & Drug Administration. Available at https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm379696.htm. December 23, 2013; Accessed: March 4, 2017.
  73. Sharief LA, Kadir RA. Congenital factor XIII deficiency in women: a systematic review of literature. Haemophilia. 2013 Aug 28. [View Abstract]
  74. Mikkola H, Muszbek L, Laiho E, et al. Molecular mechanism of a mild phenotype in coagulation factor XIII (FXIII) deficiency: a splicing mutation permitting partial correct splicing of FXIII A-subunit mRNA. Blood. 1997 Feb 15. 89(4):1279-87. [View Abstract]
  75. Greenberg DL, Davie EW. Blood coagulation factors: their complementary DNAs, genes, and expression. Coleman RW, Hirsh J, Marder VJ, et al, eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. Philadelphia, Pa: Lippincott Williams & Wilkins; 2001. 21-57.
  76. Kangsadalampai S, Yenchitsomanus P, Chelvanayagam G, Sawasdee N, Laosombat V, Board P. Identification of a new mutation (Gly420Ser), distal to the active site, that leads to factor XIII deficiency. Eur J Haematol. 2000 Oct. 65(4):279-84. [View Abstract]
  77. Koseki S, Souri M, Koga S, et al. Truncated mutant B subunit for factor XIII causes its deficiency due to impaired intracellular transportation. Blood. 2001 May 1. 97(9):2667-72. [View Abstract]
  78. Gomez Garcia EB, Poort SR, Stibbe J. Two novel and one recurrent missense mutation in the factor XIII A gene in two Dutch patients with factor XIII deficiency. Br J Haematol. 2001 Feb. 112(2):513-8. [View Abstract]
  79. Anwar R, Gallivan L, Trinh C, Hill F, Markham A. Identification of a new Leu354Pro mutation responsible for factor XIII deficiency. Eur J Haematol. 2001 Feb. 66(2):133-6. [View Abstract]
  80. Warner D, Mansfield MW, Grant PJ. Coagulation factor XIII and cardiovascular disease in UK Asian patients undergoing coronary angiography. Thromb Haemost. 2001 Mar. 85(3):408-11. [View Abstract]
  81. Canavy I, Henry M, Morange PE, et al. Genetic polymorphisms and coronary artery disease in the south of France. Thromb Haemost. 2000 Feb. 83(2):212-6. [View Abstract]
  82. Franco RF, Middeldorp S, Meinardi JR, van Pampus EC, Reitsma PH. Factor XIII Val34Leu and the risk of venous thromboembolism in factor V Leiden carriers. Br J Haematol. 2000 Oct. 111(1):118-21. [View Abstract]
  83. Catto AJ, Kohler HP, Coore J, Mansfield MW, Stickland MH, Grant PJ. Association of a common polymorphism in the factor XIII gene with venous thrombosis. Blood. 1999 Feb 1. 93(3):906-8. [View Abstract]
  84. Kohler HP. Role of blood coagulation factor XIII in vascular diseases. Swiss Med Wkly. 2001 Jan 27. 131(3-4):31-4. [View Abstract]
  85. Prata MJ, Miranda C, Rocha J, Amorim A. Allelic affinities between the F13A common gene products inferred by the analysis of an (AAAG)n STR polymorphism within the 5' untranslated region. Hum Hered. 2000 May-Jun. 50(3):189-93. [View Abstract]
  86. Chandler WL, Patel MA, Gravelle L, et al. Factor XIIIA and clot strength after cardiopulmonary bypass. Blood Coagul Fibrinolysis. 2001 Mar. 12(2):101-8. [View Abstract]
  87. Holst FG, Hemmer CJ, Foth C, Seitz R, Egbring R, Dietrich M. Low levels of fibrin-stabilizing factor (factor XIII) in human Plasmodium falciparum malaria: correlation with clinical severity. Am J Trop Med Hyg. 1999 Jan. 60(1):99-104. [View Abstract]
  88. Matayoshi T, Omi T, Sakai N, Kawana S. Clinical Significance of Blood Coagulation Factor XIII Activity in Adult Henoch-Schönlein Purpura. J Nippon Med Sch. 2013. 80(4):268-78. [View Abstract]
  89. Lorand L, Velasco PT, Murthy SN, Lefebvre P, Green D. Autoimmune antibody in a hemorrhagic patient interacts with thrombin-activated factor XIII in a unique manner. Blood. 1999 Feb 1. 93(3):909-17. [View Abstract]
  90. Feinstein DI. Immune coagulation disorders. Coleman RW, Hirsh J, Marder VJ, et al, eds. Hemostasis and Thrombosis. Basic Principles and Clinical Practice. Philadelphia, Pa: Lippincott Williams & Wilkins; 2001. 1003-20.
  91. Ahmad F, Solymoss S, Poon MC, Berube C, Sullivan AK. Characterization of an acquired IgG inhibitor of coagulation factor XIII in a patient with systemic lupus erythematosus. Br J Haematol. 1996 Jun. 93(3):700-3. [View Abstract]
  92. Tosetto A, Rodeghiero F, Gatto E, Manotti C, Poli T. An acquired hemorrhagic disorder of fibrin crosslinking due to IgG antibodies to FXIII, successfully treated with FXIII replacement and cyclophosphamide. Am J Hematol. 1995 Jan. 48(1):34-9. [View Abstract]
  93. Krumdieck R, Shaw DR, Huang ST, Poon MC, Rustagi PK. Hemorrhagic disorder due to an isoniazid-associated acquired factor XIII inhibitor in a patient with Waldenstrom's macroglobulinemia. Am J Med. 1991 May. 90(5):639-45. [View Abstract]
  94. Heinle K, Adam O, Rauh G. Factor XIII insufficiency in a patient with severe psoriasis vulgaris, arthritis, and infirmity. Clin Rheumatol. 1998. 17(4):346-8. [View Abstract]
  95. Lorand L. Acquired inhibitors of fibrin stabilization: a class of hemorrhagic disorders of diverse origins. Anticoagulants: Physiologic, Pathologic and Pharmacologic. CRC Press; 1994. 169-91.
  96. Dorgalaleh A, Kazemi A, Zaker F, Shamsizadeh M, Rashidpanah J, Mollaei M. Laboratory Diagnosis of Factor XIII Deficiency, Routine Coagulation Tests with Quantitative and Qualitative Methods. Clin Lab. 2016. 62 (4):491-8. [View Abstract]
  97. Glidden PF, Malaska C, Herring SW. Thromboelastograph assay for measuring the mechanical strength of fibrin sealant clots. Clin Appl Thromb Hemost. 2000 Oct. 6(4):226-33. [View Abstract]
  98. Karpati L, Penke B, Katona E, Balogh I, Vamosi G, Muszbek L. A modified, optimized kinetic photometric assay for the determination of blood coagulation factor XIII activity in plasma. Clin Chem. 2000 Dec. 46(12):1946-55. [View Abstract]
  99. Katona E, Haramura G, Karpati L, Fachet J, Muszbek L. A simple, quick one-step ELISA assay for the determination of complex plasma factor XIII (A2B2). Thromb Haemost. 2000 Feb. 83(2):268-73. [View Abstract]
  100. Killick CJ, Barton CJ, Aslam S, Standen G. Prenatal diagnosis in factor XIII-A deficiency. Arch Dis Child Fetal Neonatal Ed. 1999 May. 80(3):F238-9. [View Abstract]
  101. Orosz ZZ, Katona E, Facsko A, Berta A, Muszbek L. A highly sensitive chemiluminescence immunoassay for the measurement of coagulation factor XIII subunits and their complex in tears. J Immunol Methods. 2010 Feb 28. 353(1-2):87-92. [View Abstract]
  102. Brooks M. FDA Clears Tretten for Coagulation Factor XIII Deficiency. Available at http://www.medscape.com/viewarticle/818264. Accessed: January 6, 2014.
  103. Inbal A, Oldenburg J, Carcao M, Rosholm A, Tehranchi R, Nugent D. Recombinant factor XIII: a safe and novel treatment for congenital factor XIII deficiency. Blood. 2012 May 31. 119(22):5111-7. [View Abstract]
  104. Gootenberg JE. Factor concentrates for the treatment of factor XIII deficiency. Curr Opin Hematol. 1998 Nov. 5(6):372-5. [View Abstract]
  105. Green D. Spontaneous inhibitors to coagulation factors. Clin Lab Haematol. 2000 Oct. 22 Suppl 1:21-5; discussion 30-2. [View Abstract]
  106. Lorand L, Losowsky MS, Miloszewski KJM. Human factor XIII: fibrin stabilizing factor. Spaet T, ed. Progress in Hemostasis and Thrombosis. New York, NY: Grune & Stratton; 1980. Vol 5: 245-90.
  107. Abbondanzo SL, Gootenberg JE, Lofts RS, McPherson RA. Intracranial hemorrhage in congenital deficiency of factor XIII. Am J Pediatr Hematol Oncol. 1988 Spring. 10(1):65-8. [View Abstract]
  108. Green D, Sanders J, Wong C, et al. Coronary revascularization in the presence of an inhibitory antibody to factor XIII. Bull Intensive Crit Care. 1996. 3(3):14-6.
  109. Wiel E, Marciniak B, Wibaut B. [Recurrent hematomas and normal standard hemostasis tests]. Ann Fr Anesth Reanim. 1998. 17(1):61-4. [View Abstract]
  110. Kawamura A, Tamaki N, Yonezawa K, Nakamura M, Asada M. [Effect of factor XIII on intractable CSF leakage after a transpetrosal-approach operation: a case report]. No Shinkei Geka. 1997 Jan. 25(1):53-6. [View Abstract]
  111. Chamouard P, Grunebaum L, Wiesel ML, et al. Significance of diminished factor XIII in Crohn's disease. Am J Gastroenterol. 1998 Apr. 93(4):610-4. [View Abstract]
  112. Linskens RK, van Bodegraven AA, Schoorl M, Tuynman HA, Bartels P. Predictive value of inflammatory and coagulation parameters in the course of severe ulcerative colitis. Dig Dis Sci. 2001 Mar. 46(3):644-8. [View Abstract]
  113. Helio T, Wartiovaara U, Halme L, et al. Arg506Gln factor V mutation and Val34Leu factor XIII polymorphism in Finnish patients with inflammatory bowel disease. Scand J Gastroenterol. 1999 Feb. 34(2):170-4. [View Abstract]
  114. Burrows RF, Ray JG, Burrows EA. Bleeding risk and reproductive capacity among patients with factor XIII deficiency: a case presentation and review of the literature. Obstet Gynecol Surv. 2000 Feb. 55(2):103-8. [View Abstract]
  115. Kreilgaard L, Jones LS, Randolph TW, et al. Effect of Tween 20 on freeze-thawing- and agitation-induced aggregation of recombinant human factor XIII. J Pharm Sci. 1998 Dec. 87(12):1597-603. [View Abstract]
  116. Kreilgaard L, Frokjaer S, Flink JM, Randolph TW, Carpenter JF. Effects of additives on the stability of recombinant human factor XIII during freeze-drying and storage in the dried solid. Arch Biochem Biophys. 1998 Dec 1. 360(1):121-34. [View Abstract]
  117. Becker PS. Gene Therapy for Blood Disorders. The Hematologist: ASH News and Reports. Available at http://www.hematology.org/Thehematologist/Years-Best/3601.aspx. January 13, 2015; Accessed: March 4, 2017.
  118. Dickneite G, Metzner H, Nicolay U. Prevention of suture hole bleeding using fibrin sealant: benefits of factor XIII. J Surg Res. 2000 Oct. 93(2):201-5. [View Abstract]
  119. ARC. PLAS+SD (pooled plasma, solvent-detergent treated). Monograph by the American Red Cross and V. I. Technologies, Inc. 1999. VIT-001A9/99:
  120. MediView Express. Recombinant therapy enhances safety and quality of life for hemophilia patients. Paper presented at: 53rd Annual Meeting of the National Hemophilia Foundation. November 16, 2001: Nashville, Tennessee.
  121. Rigas B, Hasan I, Rehman R, Donahue P, Wittkowski KM, Lebovics E. Effect on treatment outcome of coinfection with SEN viruses in patients with hepatitis C. Lancet. 2001 Dec 8. 358(9297):1961-2. [View Abstract]
  122. Azzi A, De Santis R, Morfini M, et al. TT virus contaminates first-generation recombinant factor VIII concentrates. Blood. 2001 Oct 15. 98(8):2571-3. [View Abstract]
  123. Bachmann F. Plasminogen-plasmin enzyme system. Colman RW, Hirsh J, George JN, et al, eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 4th ed. Lippincott Williams & Wilkins; 2001. 275-320.
  124. Fergusson DA, Hebert PC, Mazer CD, et al. A comparison of aprotinin and lysine analogues in high-risk cardiac surgery. N Engl J Med. 2008 May 29. 358(22):2319-31. [View Abstract]
  125. Di Bisceglie AM. SEN and sensibility: interactions between newly discovered and other hepatitis viruses?. Lancet. 2001 Dec 8. 358(9297):1925-6. [View Abstract]
  126. Treisman GJ, Angelino AF, Hutton HE. Psychiatric issues in the management of patients with HIV infection. JAMA. 2001 Dec 12. 286(22):2857-64. [View Abstract]
  127. Carcao M, Altisent C, Castaman G, Fukutake K, Kerlin BA, Kessler C, et al. Recombinant FXIII (rFXIII-A2) Prophylaxis Prevents Bleeding and Allows for Surgery in Patients with Congenital FXIII A-Subunit Deficiency. Thromb Haemost. 2018 Mar. 118 (3):451-460. [View Abstract]
  128. Urwin PJ, Mackenzie JM, Llewelyn CA, Will RG, Hewitt PE. Creutzfeldt-Jakob disease and blood transfusion: updated results of the UK Transfusion Medicine Epidemiology Review Study. Vox Sang. 2016 May. 110 (4):310-6. [View Abstract]
  129. Jackson GS, Burk-Rafel J, Edgeworth JA, Sicilia A, Abdilahi S, Korteweg J, et al. Population screening for variant Creutzfeldt-Jakob disease using a novel blood test: diagnostic accuracy and feasibility study. JAMA Neurol. 2014 Apr. 71 (4):421-8. [View Abstract]
  130. Ivaskevicius V, Biswas A, Loreth R, et al. Mutations affecting disulphide bonds contribute to a fairly common prevalence of F13B gene defects: results of a genetic study in 14 families with factor XIII B deficiency. Haemophilia. 2010 Mar 10. [View Abstract]
  131. Inbal A, Oldenburg J, Carcao M, Rosholm A, Tehranchi R, Nugent D. Recombinant factor XIII: a safe and novel treatment for congenital factor XIII deficiency. Blood. 2012 May 31. 119(22):5111-7. [View Abstract]
  132. ARC. FDA-approved product circular for Pooled Plasma, Solvent-Detergent-Treated (PLAS+SD) manufactured by the American Red Cross and V.I. Technologies, Inc. 2000.

Final steps in clot formation (from article: Factor XIII).

Coagulation reactions leading to thrombin generation and activation of factor XIII.

Final steps in clot formation (from article: Factor XIII).

Activation of factor XIII and generation of insoluble cross-linked fibrin. Adapted from Lorand L. Ann N Y Acad Sci. 2001;936:291-311.

Postulated interaction between factor XIII and thrombin-activatable fibrinolytic inhibitor.

Cell surfaced–directed hemostasis. Initially, a small amount of thrombin is generated on the surface of the tissue factor–bearing (TF-bearing) cell. Following amplification, the second burst generates a larger amount of thrombin, leading to fibrin (clot) formation (from article: Factor XIII). Adapted from Hoffman and Monroe. Thromb Haemost. 2001;85(6):958-65.

Gene, messenger RNA, and protein for subunit A of factor XIII. Adapted from Reitsma PH. In: Hemostasis and Thrombosis: Basic Principles and Clinical Practice. Lippincott Williams & Wilkins; 2001:59-87 and from Roberts HR, Monroe DM III, Hoffman M. In: Williams Hematology. McGraw-Hill Professional; 2001:1409-34.

Coagulation reactions leading to thrombin generation and activation of factor XIII.

Final steps in clot formation (from article: Factor XIII).

Activation of factor XIII and generation of insoluble cross-linked fibrin. Adapted from Lorand L. Ann N Y Acad Sci. 2001;936:291-311.

Postulated interaction between factor XIII and thrombin-activatable fibrinolytic inhibitor.

Cell surfaced–directed hemostasis. Initially, a small amount of thrombin is generated on the surface of the tissue factor–bearing (TF-bearing) cell. Following amplification, the second burst generates a larger amount of thrombin, leading to fibrin (clot) formation (from article: Factor XIII). Adapted from Hoffman and Monroe. Thromb Haemost. 2001;85(6):958-65.

Gene, messenger RNA, and protein for subunit A of factor XIII. Adapted from Reitsma PH. In: Hemostasis and Thrombosis: Basic Principles and Clinical Practice. Lippincott Williams & Wilkins; 2001:59-87 and from Roberts HR, Monroe DM III, Hoffman M. In: Williams Hematology. McGraw-Hill Professional; 2001:1409-34.

Properties A Chain B Chain
Plasma FXIIIHas 2 A chainsHas 2 B chains
Plasma levelApproximately 15 mg/mLApproximately 21 mg/mL
Chains are free in plasmaNo. All bound to B chain and present as an A2 B2 tetramerYes. Excess B chain present in plasma as a B2 dimer
Chain contains the catalytic siteYesNo
Chain is the carrier proteinNoYes
Chain acts as a brake on FXIII activationNoYes
Cellular FXIIIHas 2 A chains (A2 dimer)Has no B chains
Mutations can lead to decreased FXIII activityYesYes