Fact-checked by Grok 2 weeks ago

Plasminogen activator

Plasminogen activators are a class of serine proteases that catalyze the conversion of the plasminogen to the active enzyme , which is essential for —the breakdown of in blood clots to maintain vascular patency. This activation process is tightly regulated and plays a central role in , preventing excessive while enabling the dissolution of clots formed during injury. The primary plasminogen activators include tissue plasminogen activator (tPA), predominantly secreted by endothelial cells and highly efficient in fibrin-bound environments, and urokinase-type plasminogen activator (uPA), which operates mainly on cell surfaces to facilitate localized proteolysis. tPA binds to fibrin through its kringle domains, enhancing its activity up to 500-fold in the presence of clots, while uPA interacts with the urokinase plasminogen activator receptor (uPAR) to promote pericellular activation. Beyond fibrinolysis, the plasminogen activator-plasmin system contributes to extracellular matrix (ECM) remodeling by activating matrix metalloproteinases (MMPs) and releasing growth factors such as vascular endothelial growth factor (VEGF), supporting processes like wound healing, embryogenesis, and angiogenesis. In , dysregulation of this system is implicated in conditions ranging from thrombotic disorders to , where elevated and uPAR levels correlate with invasive tumor behavior and poor . Clinically, recombinant tPA is a of thrombolytic , approved for acute ischemic treatment within 3 to 4.5 hours of symptom onset, reducing by 10-30% when administered promptly, though it carries risks of hemorrhage. Ongoing research explores inhibitors of uPA/uPAR for anticancer applications and combination therapies to extend thrombolytic windows.

Molecular Biology

Structure and Mechanism

Plasminogen activators (PAs) belong to the family, specifically the chymotrypsin-like subgroup, which is characterized by a composed of , aspartate, and serine residues essential for hydrolyzing s. This triad facilitates nucleophilic attack by the serine hydroxyl group on the carbonyl carbon of the substrate , enabling the proteolytic activity central to PA function. The primary mechanism of PAs involves the specific of the Arg561-Val562 in plasminogen, converting the single-chain plasminogen into the active two-chain . This occurs within the activation loop of plasminogen and triggers a conformational change, releasing the from latency by inserting the newly formed N-terminal into a that stabilizes the of . The overall reaction catalyzed by PA can be summarized as: \text{Plasminogen} \xrightarrow{\text{PA}} \text{Plasmin} PAs themselves are often produced as inactive zymogen forms (single-chain) that undergo proteolytic activation to yield the mature two-chain enzymes, accompanied by conformational rearrangements that expose the active site. Structural features of PAs include modular domains that support substrate recognition and localization. For instance, tissue-type plasminogen activator (tPA) comprises a finger domain (fibronectin type II-like, residues 4-50), an epidermal growth factor (EGF)-like domain (residues 50-91), two kringle domains (residues 92-173 and 175-261) for fibrin binding, and a C-terminal serine protease domain (residues 276-527) containing the catalytic triad (His322, Asp371, Ser478). In contrast, urokinase-type plasminogen activator (uPA) features an EGF-like domain (residues 1-49) for receptor binding, a single kringle domain (residues 50-135), and a serine protease domain (residues 159-411) with its catalytic triad. These domains enable targeted activation of plasminogen at specific physiological sites.

Types

Plasminogen activators are primarily categorized into two endogenous mammalian isoforms: tissue plasminogen activator (tPA) and urokinase plasminogen activator (). tPA, encoded by the gene located on 8p11.21, is predominantly produced by endothelial cells and exhibits high fibrin specificity, meaning it efficiently activates plasminogen to only in the presence of clots. In contrast, uPA, encoded by the PLAU gene on 10q22.2, is synthesized mainly in the and secreted into , though it is also produced by various cell types including macrophages and epithelial cells; it lacks fibrin specificity and activates plasminogen independently of . Both isoforms share a common mechanism of cleaving plasminogen at the Arg561-Val562 bond to generate . uPA exists in two main forms: the inactive single-chain pro-uPA (scuPA), which binds to its specific receptor uPAR (encoded by PLAUR on 19q13), and the active two-chain uPA (tcuPA) generated upon proteolytic activation of scuPA. tPA is secreted as a single-chain that is rapidly converted to its two-chain active form. A less common variant is , a bacterial plasminogen activator derived from species, which indirectly activates plasminogen through complex formation rather than direct enzymatic cleavage and is not endogenous to humans. The discovery of these activators traces back to mid-20th-century research on ; was first identified in human urine in 1947, while tPA was purified in sufficient quantities from cell cultures in the early , enabling detailed characterization.
PropertytPA (Tissue-type)uPA (Urokinase-type)
SpecificityFibrin-specificNon-fibrin specific
Half-life~5 minutes~10 minutes (for active forms)
Production SitesEndothelial cells, macrophages, epithelial cells

Physiological Functions

In Fibrinolysis

Plasminogen activators (PAs) play a central role in the fibrinolytic system, which counterbalances the coagulation cascade to maintain vascular patency by degrading unwanted blood clots. This process was first linked to in the through studies on , a bacterial protein identified by Tillett and Sherry that indirectly activates plasminogen to , demonstrating the potential for enzymatic clot dissolution. In the coagulation-fibrinolysis balance, PAs localize to clots primarily through direct , concentrating their activity at sites of to promote targeted degradation without widespread proteolytic effects. (tPA), a key endogenous PA, exemplifies this by to specific residues on surfaces, which facilitates the recruitment and activation of plasminogen. The fibrin-dependent activation of tPA is markedly enhanced on clot surfaces, with catalytic for plasminogen increasing approximately 500-fold compared to solution-phase conditions, ensuring rapid and clot-specific generation. Downstream, cleaves into soluble degradation products, including D-dimers, which serve as markers of and exhibit properties by interfering with platelet aggregation and generation. This mechanism supports both local at the clot site and systemic surveillance to prevent , as tPA's affinity limits off-target activity, thereby preserving while enabling resolution.

In Tissue Remodeling and Development

Plasminogen activators, particularly urokinase-type plasminogen activator () and its receptor (uPAR), play a in facilitating and invasion during wound repair by promoting pericellular proteolysis of the (). In astrocytic wound healing models, uPA binding to uPAR triggers β-catenin signaling, detaching it from N-cadherin to enhance cell motility without stimulating proliferation, thereby accelerating repopulation of the wound site. This process is plasminogen-independent in some contexts but relies on uPA-mediated crosstalk for efficient closure, as evidenced by reduced in uPA-deficient cells. Similarly, in vitro scratch assays demonstrate that uPA supports into wounded areas through remodeling. In , generated by plasminogen activators degrades the , enabling endothelial cell and new vessel formation. and (tPA) activate matrix metalloproteinases (MMPs) that further break down ECM barriers, releasing proangiogenic factors such as (VEGF-A) to support vascular remodeling. directly targets components, including and , to facilitate tip cell invasion during . Plasminogen-deficient mice exhibit impaired angiogenic responses, underscoring the system's necessity for physiological vessel growth. During embryonic development, the uPA-uPAR system is essential for invasion at implantation, where uPAR expression on extravillous s promotes migration into the via localized proteolysis. Silencing uPAR reduces outgrowth and MMP-2 activity while increasing inhibitors of metalloproteinases (TIMPs), impairing invasion. models reveal the critical nature of this pathway; double uPA/tPA-deficient mice display retarded postnatal growth, reduced fertility, and shortened life expectancy due to defective remodeling and . specifically degrades key ECM proteins like and , enabling cellular reorganization during implantation and . uPA contributes to ovulation by supporting follicular wall degradation, with combined uPA/tPA deficiency reducing ovulation efficiency by 26% in gonadotropin-induced models, highlighting functional between activators. In mammary gland physiology, plasminogen activators ensure lobuloalveolar development and milk production; plasminogen deficiency causes milk stasis, , and premature involution due to impaired ECM turnover and epithelial . Activation of plasminogen by uPA or tPA on epithelial surfaces via receptors like Plg-RKT clears deposits and degrades , supporting competence.

Regulation

Activation Pathways

Plasminogen activators, primarily and , are activated through distinct upstream pathways that ensure targeted proteolytic activity. The intrinsic pathway predominantly involves the contact activation system, where factor XIIa and plasma proteolytically cleave the single-chain proenzyme form of uPA (pro-uPA) at the Lys158-Ile159 bond to generate active two-chain uPA. This process is slower with factor XIIa compared to , highlighting 's dominant role in initiating uPA during intrinsic . In contrast, the extrinsic pathway relies on -generated as a key cofactor that enhances tPA's catalytic efficiency in converting plasminogen to by over 500-fold through colocalization of tPA, plasminogen, and surfaces. further contributes by stimulating the rapid release of pre-stored tPA from endothelial cells, amplifying extrinsic in response to vascular injury. Receptor-mediated activation provides spatial control, particularly for uPA, where pro-uPA binds with high affinity to the glycosylphosphatidylinositol-anchored urokinase plasminogen activator receptor (uPAR) on surfaces, facilitating localized conversion to active uPA by nearby proteases such as . This uPAR-pro-uPA complex not only concentrates proteolytic activity at the pericellular environment but also promotes feedback amplification by enabling efficient generation in close proximity. Pathways differ notably between tPA and uPA, with tPA favoring soluble or fibrin-bound while uPA emphasizes cell-surface localization via uPAR. Positive feedback loops sustain PA activity, as plasmin produced from initial plasminogen activation reciprocally cleaves and activates pro-uPA, creating an amplification cascade that accelerates overall proteolysis without requiring additional external triggers. This mutual activation between plasmin and pro-uPA forms a robust autoregulatory mechanism essential for rapid responses in tissue remodeling. At the transcriptional level, genetic regulation modulates PA expression; for instance, the (PLAU) is upregulated by the complex, which binds to specific enhancer elements in the promoter region in response to proinflammatory such as tumor necrosis factor-alpha (TNF-α). AP-1 activation, often via c-Jun and c-Fos dimers, integrates signaling to enhance uPA transcription during inflammatory or invasive processes.

Inhibitors and Modulation

Plasminogen activator inhibitor-1 (PAI-1), also known as family E member 1, serves as the primary physiological inhibitor of both (tPA) and urokinase-type plasminogen activator (), thereby regulating and pericellular to prevent excessive generation. Active PAI-1 is stabilized through high-affinity binding to the somatomedin B domain of , a in plasma and the , which extends its functional and modulates its localization in tissues. Elevated PAI-1 levels are commonly observed in , where adipocytes contribute significantly to its production, fostering a hypofibrinolytic state that heightens risk and contributes to progression. Plasminogen activator inhibitor-2 (PAI-2), or family B member 2, is predominantly produced by placental trophoblasts and exists mainly as a non-glycosylated intracellular form consisting of 415 , with limited under normal conditions. PAI-2 levels are undetectable in non-pregnant individuals but rise markedly during , peaking in the third (e.g., up to 100 ng/mL in certain cohorts), to inhibit and protect placental integrity by suppressing . Additional endogenous inhibitors modulate plasminogen activator activity indirectly by targeting downstream plasmin or related proteases. Alpha-2-antiplasmin (α2-antiplasmin) primarily neutralizes free plasmin in circulation by forming stable complexes, thereby limiting unbound proteolytic activity and localizing fibrinolysis to fibrin-bound sites. Protease nexin-1 (PN-1), a serpin also known as serpin family E member 2, inhibits tPA, uPA, and plasmin with high efficiency, particularly in extravascular contexts like tissue remodeling. Protein C inhibitor (PCI), alternatively termed plasminogen activator inhibitor-3 (PAI-3), further restrains the system by inhibiting uPA and activated protein C, balancing coagulation and fibrinolysis in plasma and reproductive fluids. Hormonal factors fine-tune PAI-1 expression to maintain fibrinolytic . , such as 17β-estradiol, decreases PAI-1 protein levels and activity in vascular cells through receptor-mediated suppression of and effects that reduce signaling, thereby enhancing . In contrast, acute and induces PAI-1 synthesis in a tissue-specific manner (e.g., up to 70-fold in of aged models), elevating plasma levels and promoting a prothrombotic via glucocorticoid-dependent pathways. Genetic variations in PAI-1 further influence its regulation and clinical outcomes. The 4G/5G polymorphism in the PAI-1 promoter (rs1799889) is associated with increased risk, particularly , as the 4G allele enhances transcriptional activity and PAI-1 expression; meta-analyses show an of 1.13 for CAD overall, with stronger effects in males (OR 1.15) and younger individuals (OR 1.19 for ≤50 years).

Clinical Applications

Thrombolytic Therapies

Thrombolytic therapies utilize recombinant forms of plasminogen activators to dissolve acute thrombi in conditions such as (MI), ischemic , and , thereby restoring blood flow and reducing tissue damage. These agents, primarily tissue plasminogen activator (tPA) variants and urokinase-type plasminogen activator (uPA), promote the conversion of plasminogen to , which degrades in clots, emulating the endogenous pathway. Administered intravenously or via catheter-directed methods, they are most effective when given early after symptom onset, ideally within hours, to maximize reperfusion benefits while minimizing risks. Alteplase (recombinant tPA, rtPA), the first recombinant plasminogen activator approved by the FDA in 1987 for acute , revolutionized treatment by enabling rapid clot lysis in . For , the accelerated regimen involves an initial 15 mg intravenous () bolus, followed by 0.75 mg/kg (maximum 50 mg) infused over 30 minutes, and then 0.5 mg/kg (maximum 35 mg) over the next , with a total dose not exceeding 100 mg. In 1996, the FDA expanded approval to acute ischemic , using a 0.9 mg/kg dose (maximum 90 mg), administered as a 10% bolus over 1 minute followed by the remainder infused over . This dosing targets rapid while limiting systemic exposure. In March 2025, the FDA approved , another tPA variant, for acute ischemic at a dose of 0.25 mg/kg as a single bolus (maximum 25 mg), offering a simpler compared to alteplase. Other tPA variants offer improved for easier administration. Reteplase, a deletion mutant of tPA with a longer plasma of 13-16 minutes compared to alteplase's 4-6 minutes, was approved for acute and is given as a fixed double-bolus regimen: 10 units over 2 minutes, repeated 30 minutes later. This bolus approach simplifies delivery in emergency settings without continuous infusion. , engineered for enhanced specificity and a of approximately 20 minutes, allows single-bolus administration for acute ST-elevation : 30 mg for patients under 60 kg, increasing in 5 mg increments to 50 mg for those 90 kg or more, injected over 5 seconds. Its cardiac-focused profile reduces non-target . Urokinase (uPA), a naturally derived , is FDA-approved for lysis of acute massive , often via systemic infusion: 4,400 international units/kg as a over 10 minutes, followed by 4,400 international units/kg/hour for 12-24 hours. Catheter-directed delivery enhances local efficacy for or peripheral thrombi, using lower doses (e.g., 100,000-250,000 units/hour) infused directly at the clot site to achieve faster resolution with reduced systemic risk. Large-scale trials have established the efficacy of these agents in acute . The GUSTO trial demonstrated that accelerated reduced 30-day mortality to 6.3% compared to 7.3% with , a relative reduction of 14%, building on prior evidence that thrombolytics overall decrease short-term mortality by 25-30% versus no reperfusion. Reteplase and showed comparable 30-day mortality rates to alteplase (around 7%) in pivotal trials like RAPID-II and ASSENT-2, with similar reperfusion success but potentially lower reocclusion rates due to their . For , achieves angiographic improvement in over 70% of cases within 24 hours, comparable to alteplase. Despite their benefits, thrombolytics carry significant risks, primarily hemorrhage due to plasmin's systemic fibrinolytic effects. Intracranial hemorrhage occurs in 0.5-1% of alteplase-treated MI or patients, with rates up to 6% in some stroke subgroups, while major bleeding affects 5-10% overall. Common side effects include gastrointestinal or genitourinary bleeding, , and allergic reactions (rare with recombinant agents). Contraindications include active , recent (within 3 months) ischemic or , recent major or (within 2-4 weeks), uncontrolled (>180/110 mmHg), and suspected , as these increase hemorrhagic transformation risk. Monitoring involves serial assessments for bleeding signs, with reversal using or if needed.

Diagnostic and Emerging Uses

Elevated levels of (PAI-1) serve as a for assessing risk, with studies showing that higher concentrations are associated with an increased incidence of venous (VTE). Active PAI-1 levels have been independently linked to heightened VTE risk in patients with , highlighting its prognostic value in high-risk populations. Similarly, urokinase-type (uPA) and the uPA/PAI-1 complex in act as s for thrombosis-related complications in severe inflammatory conditions, such as , where they predict adverse outcomes including clotting events. In diagnostic imaging research, radiolabeled tissue plasminogen activator (tPA) enables non-invasive detection of thrombi. Enzymatically inactivated, radiolabeled tPA facilitates prompt scintigraphic visualization of thrombi , offering a targeted approach to identify clot locations without active . Technetium-99m-labeled recombinant tPA exhibits high for acute (DVT), with uptake patterns that distinguish fresh clots from those older than 30 days post-diagnosis. Emerging therapies leverage advanced delivery systems for plasminogen activators (PAs) to improve safety and efficacy. Preclinical studies from 2022 to 2025 have demonstrated nanoparticle-conjugated PAs for targeted , including plasminogen activator-coated nanobubbles that bind β2-glycoprotein I on activated platelets, enhancing clot-specific in thrombotic models. Systematic reviews of nanoparticle-based thrombolytics confirm improved fibrinolytic outcomes in ischemic preclinical models, reducing off-target effects compared to free tPA. Magnetic nanotherapeutics conjugated with recombinant tPA show strong biodistribution and thrombolytic efficiency in translational assessments, supporting their potential for clinical advancement. approaches for sustained PA delivery remain in early research stages, focusing on local expression to prevent rebound , though dedicated trials in the 2020s are not yet widespread. Non-thrombotic applications of PA modulation include inhibitors for treatment. Recent advances target the /uPAR system, which drives remodeling in fibrotic diseases, with preclinical evidence showing that uPA inhibition attenuates progression in liver and models.

Pathological Roles

In Cancer Progression

Plasminogen activators, particularly urokinase-type plasminogen activator (), play a critical role in cancer progression through the uPA-uPAR axis, which facilitates pericellular essential for () degradation and tumor cell in solid tumors. The binding of uPA to its receptor uPAR localizes plasmin generation at the tumor cell surface, enabling the breakdown of ECM components such as and , thereby promoting invasive behavior across various malignancies including , , and colorectal cancers. This localized not only supports expansion but also initiates the metastatic cascade by allowing cancer cells to breach membranes and enter surrounding . Elevated levels of and its inhibitor (PAI-1) serve as robust prognostic markers, correlating with adverse outcomes in multiple cancer types. In , high uPA and PAI-1 concentrations in tumor tissue independently predict reduced disease-free survival and overall survival, particularly in lymph node-negative cases, as validated in level-of-evidence-1 prospective studies involving thousands of patients. Similar associations hold for , where meta-analyses from the 2000s to 2010s demonstrate that increased uPA/PAI-1 expression links to higher recurrence rates and poorer . In , prospective analyses confirm that uPA/PAI-1 levels post-surgery forecast progression and survival, with high expression indicating aggressive disease. These biomarkers outperform traditional factors like tumor grade in stratifying risk, guiding decisions on . Beyond invasion, the uPA-uPAR system drives and , key mechanisms in . uPA stimulates the release of (VEGF) from ECM-bound stores, promoting endothelial cell proliferation and new vessel formation to sustain tumor and growth. In , uPAR-mediated signaling activates and G-protein-coupled receptors, enhancing chemotactic responses that direct cancer cell migration toward distant sites, as observed in preclinical models of and tumors. This multifaceted role underscores uPA-uPAR's contribution to systemic dissemination. Therapeutic strategies targeting have advanced to clinical stages, though challenges persist. The small-molecule inhibitor WX-UK1 (upamostat) reached phase II trials in the 2010s for advanced solid tumors, including pancreatic and cancers, demonstrating safety and preliminary efficacy in reducing but limited overall survival benefits in monotherapy; however, development has continued in combination therapies for cancer and other indications such as infectious s. More recently, antibody-based approaches, such as anti-uPAR VH domain antibodies and conjugates, have entered preclinical and early clinical evaluation as of 2023, showing promise in blocking uPAR signaling to inhibit in and pancreatic models. In specifically, stands as an independent prognostic factor, endorsed in the 2007 ASCO guidelines for node-negative risk . In the 2022 update, and PAI-1 are recommended only if locally validated and in conjunction with other parameters for patients without access to genomic tests, supporting limited use in personalized therapy decisions.

In Cardiovascular and Other Diseases

Plasminogen activator inhibitor-1 (PAI-1) overexpression impairs fibrinolysis by inhibiting tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA), leading to thrombus persistence and increased risk of thrombotic events such as myocardial infarction (MI) and stroke. In the Framingham Heart Study offspring cohort, elevated plasma PAI-1 levels independently predicted cardiovascular disease (CVD) events, including MI and stroke, even after adjusting for traditional risk factors like hypertension and diabetes. Elevated baseline PAI-1 levels are associated with a 2- to 10-fold excess risk of coronary artery disease, stroke, and CVD-related mortality. Serial increases in PAI-1 are also linked to modestly increased CVD risk (adjusted HR 1.3 for the highest quartile change). In , uPA contributes to plaque instability and rupture by promoting matrix degradation and inflammatory cell infiltration within vascular lesions. Elevated uPA and its receptor (uPAR) expression in macrophages correlates with features, such as thin fibrous caps and necrotic cores, facilitating plaque rupture and subsequent acute coronary syndromes. Studies in E-deficient mice demonstrate that uPA deficiency paradoxically accelerates plaque progression and luminal obstruction, underscoring its complex role in balancing plaque remodeling and stability. Beyond cardiovascular conditions, plasminogen activators exhibit pathological roles in several non-oncological diseases. In , tPA facilitates amyloid-β (Aβ) clearance through plasmin-mediated proteolysis, reducing plaque burden in mouse models of amyloid pathology; however, it also exerts a by triggering intracellular signaling pathways that promote phosphorylation and in response to Aβ exposure. In , drives joint destruction by enhancing synovial invasion and matrix breakdown via activation, acting as a pro-inflammatory mediator that exacerbates erosive lesions. involves hypofibrinolysis characterized by PAI-1 upregulation, which suppresses generation and promotes microvascular , contributing to (DIC) and multi-organ failure with elevated PAI-1 levels (>83 ng/mL) linked to poor prognosis. As of 2025, upamostat is under evaluation in phase II/III trials for early treatment of to mitigate and hospitalization risks. Genetic variations in PAI-1 further modulate thrombotic risk; the /4G promoter , which enhances PAI-1 transcription, is associated with increased venous thromboembolism (VTE) susceptibility, with meta-analyses reporting an of approximately 1.5 to 2.0 for homozygous carriers compared to 5G/5G individuals, particularly in Asian populations. Recent studies from 2021 to 2025 highlight tPA's neurotoxic effects in ischemic , particularly its disruption of the blood-brain barrier (). Exogenous tPA administration increases BBB permeability by mobilizing pro-inflammatory immune cells like neutrophils, exacerbating hemorrhagic transformation and brain edema in rodent models. Endogenous tPA knockdown post-recanalization mitigates BBB damage and improves neurological outcomes, suggesting a paradoxical role where tPA's thrombolytic benefits are offset by vascular toxicity in the subacute phase. Adjunctive therapies, such as mesenchymal stem cell-derived extracellular vesicles, have been shown to attenuate tPA-induced BBB disruption, reducing infarct expansion in experimental .

References

  1. [1]
    Physiology, Plasminogen Activation - StatPearls - NCBI Bookshelf
    Sep 26, 2022 · Plasminogen activation results in increased conversion of plasminogen to plasmin, the latter an enzyme that breaks down the fibrinogen in blood clots.Missing: biology | Show results with:biology
  2. [2]
    The Plasminogen–Activator Plasmin System in Physiological and ...
    Dec 29, 2021 · The plasminogen activator–plasmin system plays a major role in the extracellular matrix remodeling process necessary for angiogenesis.Missing: definition | Show results with:definition
  3. [3]
    Serine‐proteases as plasminogen activators in terms of fibrinolysis
    Feb 7, 2012 · Plasminogen activators cleave the Arg561-Val562 peptide bond, whereby the two-chain plasmin occurs. The five kringle present the heavy chain, ...
  4. [4]
    of the serine protease plasmin - PNAS
    Asignifi- cant level of activity can be restored by adding imidazole, which can partially replace His-603 in the catalytic triad. We also directly demonstrate ...
  5. [5]
    Streptokinase triggers conformational activation of plasminogen ...
    Jul 13, 2001 · Cleavage of Arg(561)-Val(562) in plasminogen (Pg) generates plasmin (Pm) through a classical activation mechanism triggered by an insertion ...
  6. [6]
    New insights into the molecular mechanisms of the fibrinolytic system
    All plasminogen activators convert plasminogen to plasmin by cleavage of a single Arg561–Val562 peptide bond. The two-chain plasmin molecule is composed of ...
  7. [7]
    Crystal Structure of the Michaelis Complex between Tissue-type ...
    Human tPA contains a fibronectin type II domain (amino acids 1–50), a growth factor domain (amino acids 51–91), two kringle domains (amino acids 92–261), an ...
  8. [8]
    Domain structure and interactions of recombinant urokinase-type ...
    Feb 25, 1992 · Urokinase-type plasminogen activator (uPA) is a mosaic glycoprotein composed of an epidermal growth factor-like (EGF), a kringle and a serine protease (SP) ...
  9. [9]
    Tissue-type plasminogen activator: variants and crystal/solution ...
    NMR and crystal structures of many components of tissue-type plasminogen activator (t-PA) are now available: the finger-EGF pair and the kringle-2 domain ...
  10. [10]
    5327 - Gene ResultPLAT plasminogen activator, tissue type [ (human)]
    Aug 19, 2025 · This gene encodes tissue-type plasminogen activator, a secreted serine protease that converts the proenzyme plasminogen to plasmin, a fibrinolytic enzyme.
  11. [11]
    Gene: PLAU (ENSG00000122861) - Summary - Homo_sapiens
    Chromosome 10: 73,909,177-73,917,501 forward strand. This gene has 10 transcripts (splice variants), 442 orthologues, 3 paralogues and is associated with 3 ...
  12. [12]
    p00750 · tpa_human - UniProt
    Jul 21, 1986 · Tissue-type plasminogen activator (tPA) converts plasminogen to plasmin, playing a role in tissue remodeling, cell migration, and fibrinolysis.
  13. [13]
    p00749 · urok_human - UniProt
    Feb 23, 2022 · P00749, Urokinase-type plasminogen activator, cleaves plasminogen to form plasmin, and is involved in blood coagulation and fibrinolysis.
  14. [14]
    Urokinase-Type Plasminogen Activator Receptor (uPAR) in ...
    tcuPA and the single-chain form (scuPA) can both bind to uPAR; however, tcuPA is 250 times more potent at generating plasmin than scuPA [11]. A range of ...
  15. [15]
    The Mechanism of a Bacterial Plasminogen Activator ... - PubMed
    Unlike staphylokinase, a single domain protein, streptokinase, a 3-domain (alpha, beta, and gamma) molecule, nonproteolytically activates human (h)-plasminogen ...
  16. [16]
    Urokinase - Wikipedia
    The human urokinase protein was discovered, but not named, by McFarlane and Pilling in 1947. Urokinase was originally isolated from human urine, and it is also ...
  17. [17]
    The Tissue-Type Plasminogen Activator Story
    When we sought initially to purify the plasminogen activator from Bowes melanoma cell culture fluid in 1979, we observed that the activator, unlike urokinase, ...
  18. [18]
    History of drugs for thrombotic disease. Discovery, development ...
    Tillett and Sherry's group stumbled on the fibrinolytic properties of streptokinase in the 1930s and pioneered the therapeutic use of streptokinase in the 1940 ...
  19. [19]
    The Fibrinolytic System: Mysteries and Opportunities - PMC
    Jun 1, 2021 · ... fibrin, and markedly potentiates (~500-fold) the ability of tPA to activate plasminogen. Fibrin therefore initiates its own demise; an ...
  20. [20]
    Fibrinolysis and the control of blood coagulation - PMC - NIH
    Plasmin is generated from the zymogen plasminogen on the surface of the fibrin clot, or on cell surfaces, by either tissue plasminogen activator (tPA) or ...
  21. [21]
    Differences between binding of one-chain and two-chain tissue ...
    Aug 15, 1989 · Interaction of tissue plasminogen activator (t-PA) with fibrin plays a key role in regulation of plasminogen activation and clot dissolution.
  22. [22]
    Fibrin(ogen) as a Therapeutic Target: Opportunities and Challenges
    Jun 28, 2021 · The colocalization of tPA and plasminogen on fibrin leads to 500-fold increase in catalytic efficiency of plasminogen activation compared ...
  23. [23]
    D-Dimer Test - StatPearls - NCBI Bookshelf
    Jun 22, 2025 · ... plasmin, an enzyme that dissolves the clot through lytic action. Clot breakdown produces fibrin degradation products, one of which is D-dimer.
  24. [24]
    Anticoagulant properties of purified X-like fragments of human ...
    The anticoagulant properties of fibrinogen digestion products change with stage of digestion. On digestion with leukocyte elastase, in the presence of ...
  25. [25]
    Systemic versus localized coagulation activation contributing to ...
    The resulting effect on fibrinolysis is a complete inhibition and, as a consequence, inadequate fibrin removal, contributing to microvascular thrombosis.
  26. [26]
    Urokinase-type plasminogen activator-mediated crosstalk between ...
    Urokinase-type plasminogen activator (uPA) triggers a switch from N-cadherin-induced cell adhesion to β-catenin cell signaling that promotes astrocytic wound ...
  27. [27]
    uPA dependent and independent mechanisms of wound healing by ...
    In vitro wound healing and cell migration involves uPA. Fibroblast cell migration to wounded area is a hallmark of wound closure [21]. We used a standard in ...
  28. [28]
    Cell Surface Remodeling by Plasmin: A New Function for an Old ...
    The plasminogen activation (PA) system plays an important role in various pathophysiological processes, including vascular and tissue remodeling, tumor ...<|control11|><|separator|>
  29. [29]
    Expression of uPAR in human trophoblast and its role in ... - NIH
    uPAR promotes trophoblast migration and invasion during embryo implantation as well as plays a role in trophoblast differentiation into syntrophoblasts. Thus, ...
  30. [30]
    Characterizing the role of tissue-type plasminogen activator in a ...
    Apr 19, 2019 · Double-deficient uPA and tPA mice suffer from retarded postnatal growth, are significantly less fertile, and have a shortened life ...
  31. [31]
    Ovulation efficiency is reduced in mice that lack plasminogen ...
    ... plasminogen activator in ovulation. In vitro response of granulosa cells to gonadotropins, cyclic nucleotides, and prostaglandins. J Biol Chem. 1976 Sep 25 ...
  32. [32]
    Lack of plasminogen leads to milk stasis and premature mammary ...
    Lack of plasminogen leads to milk stasis and premature mammary gland involution during lactation ... (uPA and tPA respectively). One of the main plasmin ...Missing: production | Show results with:production
  33. [33]
    The Plasminogen Receptor, Plg-RKT, is Essential for Mammary ...
    uPA levels were not significantly different between the genotypes: 4.5±0.81 ng/mg and 5.5 ± 0.77 ng/mg of mammary tissue protein in Plg-RKT+/+ and Plg-RKT−/− ...
  34. [34]
    The activation of pro-urokinase by plasma kallikrein and ... - PubMed
    Mar 15, 1986 · The rate of activation of pro-urokinase by factor XIIa was approximately one-seventh of that by plasma kallikrein. The activation of the ...Missing: uPA | Show results with:uPA
  35. [35]
    Kinetics of the activation of plasminogen by human tissue ... - PubMed
    The Low Michaelis constant in the presence of fibrin allows efficient plasminogen activation on a fibrin clot, while its high value in the absence of fibrin ...
  36. [36]
    Thrombin induction of plasminogen activator-inhibitor in cultured ...
    We conclude that physiologic concentrations of thrombin increase both PA-I activity and tPA antigen in medium conditioned by human umbilical vein endothelial ...Missing: enhances | Show results with:enhances
  37. [37]
    Review The urokinase receptor: Focused cell surface proteolysis ...
    May 3, 2010 · uPAR is a GPI-anchored protein that binds with high-affinity and activates the serine protease uPA, thus regulating proteolytic activity at the cell surface.
  38. [38]
    Activation of Pro-uPA Is Critical for Initial Escape from the Primary ...
    Our findings demonstrate that, by generating plasmin, activated tumor-derived uPA facilitates early stages of PC-hi/diss dissemination.
  39. [39]
    The autoactivation of human single-chain urokinase-type ...
    The urokinase-type plasminogen activator (uPA) and plasmin cleave and activate each other, constituting a positive feedback loop. How this mutual activation ...
  40. [40]
    Transcriptional complexity and roles of Fra-1/AP-1 at the uPA/Plau ...
    Sep 8, 2014 · AP-1 is an ubiquitous family of dimeric transcription factors that controls virtually all important cell decisions due to binding to so-called ...
  41. [41]
    A Narrative Review on Plasminogen Activator Inhibitor-1 and Its ...
    Mar 8, 2021 · The plasminogen activator/plasmin system is also important for tissue remodeling and promoting wound healing, which is characterized by ...2. Pai-1 Synthesis And... · 3. Pai-1 Structure And... · 4. Role Of Pai-1 In Diverse...<|control11|><|separator|>
  42. [42]
    Plasminogen activator inhibitor-2 and impaired fibrinolysis in ... - NIH
    Jul 4, 2023 · PAI-2 is mainly produced by trophoblasts in the placenta and may thus contribute to the inhibition of fibrinolysis which occurs during ...
  43. [43]
    α2-Antiplasmin as a Potential Therapeutic Target for Systemic ... - NIH
    Mar 9, 2022 · α2-antiplasmin is known to be the main plasmin inhibitor and has various functions such as cell differentiation and cytokine production.
  44. [44]
    Recent Advances on Plasmin Inhibitors for the Treatment of ...
    Four physiologic inhibitors regulate fibrinolysis including plasminogen-activator inhibitor −1 and −2 (PAI-1 and PAI-2), which inhibit tPA and uPA, and α2- ...
  45. [45]
    Protein C Inhibitor - an overview | ScienceDirect Topics
    Protein C inhibitor is a member of the serine proteinase inhibitor family and is also known as plasminogen activator inhibitor-3 (PAI-3).
  46. [46]
    Effects of 17β-Estradiol on the Plasminogen Activator System ... - NIH
    Beneficial effects of estrogen on PAI-1 in VSMCs demonstrated in this study might contribute to attenuating atherosclerosis progression including neointima ...
  47. [47]
    Plasminogen activator inhibitor-1 is a major stress-regulated gene
    These observations indicate that the stress induces the PAI-1 gene expression in a tissue-specific and cell type-specific manner. The induction of PAI-1 mRNA by ...
  48. [48]
    PAI-1 4G/5G polymorphism and coronary artery disease risk
    Elevated PAI-1 levels appear to increase the risk of atherothrombotic events and may also promote the progression of vascular disease [4]. Renckens et al.
  49. [49]
    Association between plasminogen activator inhibitor-1 and ...
    Jun 5, 2018 · Ultimately, thrombosis risk is influenced by the balance between PAI-1 and t-PA. Thus, an increase in the PAI-1 levels in plasma can induce a ...
  50. [50]
    Plasminogen activator inhibitor 1 and venous thrombosis in ...
    Jan 21, 2021 · Levels of active PAI-1 were independently associated with increased risk of VTE in patients with pancreatic cancer.
  51. [51]
    Urokinase-type plasminogen activator and plasminogen ... - PubMed
    Jan 11, 2024 · These findings argue for using uPA and uPA/PAI-1 as novel biomarkers to detect patients at risk of developing severe COVID-19, including ARDS.Missing: plasma | Show results with:plasma
  52. [52]
    Visualization of thrombi in pulmonary arteries with radiolabeled ...
    The use of radiolabeled TPA in which thrombolytic activity is inactivated permits prompt scintigraphic detection of thrombi in pulmonary arteries in vivo.Missing: clot | Show results with:clot
  53. [53]
    Aging of Acute Deep Vein Thrombosis Measured by Radiolabeled ...
    99m Tc-recombinant tissue plasminogen activator (rt-PA) imaging has had a high sensitivity and specificity for the detection of deep vein thrombosis (DVT).
  54. [54]
    Plasminogen activator-coated nanobubbles targeting cellbound β2 ...
    Sep 29, 2022 · We developed a new targeted thrombolytic agent consisting of nanobubbles (NB) coated with recombinant tissue plasminogen activator (rtPA) and a recombinant ...
  55. [55]
    Nanoparticles for Thrombolytic Therapy in Ischemic Stroke - NIH
    Feb 6, 2025 · Nanoparticles for Thrombolytic Therapy in Ischemic Stroke: A Systematic Review and Meta-Analysis of Preclinical Studies
  56. [56]
    (PDF) Targeted Thrombolysis with Magnetic Nanotherapeutics
    Oct 12, 2025 · This study evaluated the targeting efficiency, efficacy, biodistribution, and potential toxicity of a rtPA covalently attached to chitosan- ...
  57. [57]
    Gene therapy to promote thromboresistance - PNAS
    Gene therapy offers a method of local high-level tPA expression over a prolonged time period to avoid both systemic hemorrhage and local rebound thrombosis.Missing: sustained | Show results with:sustained
  58. [58]
    Recent Advances in Targeting the Urokinase Plasminogen Activator ...
    Apr 29, 2023 · We believe that the combination of uPA inhibitors with NDDS holds enormous potential to overcome existing challenges faced by traditional uPA ...
  59. [59]
    Clinical trials with uPA/uPAR system inhibitors. - ResearchGate
    Based on the phase I trial results, phase II studies were accomplished in order to assess the clinical efficacy and safety of Å6 (Table 1). Patients with ...
  60. [60]
    Multifaceted Role of the Urokinase-Type Plasminogen Activator ...
    Urokinase-Type Plasminogen Activator Receptor. The uPAR (CD87) has the ability to localize both pro-uPA and active uPA to the cell surface (39) (Figure 3) ...Missing: scuPA tcuPA
  61. [61]
    Urokinase type plasminogen activator receptor (uPAR) as a new ...
    Nov 1, 2016 · uPAR also regulates cell adhesion, migration and proliferation, protects from apoptosis and contributes to epithelial mesenchymal transition ( ...
  62. [62]
    uPA and PAI-1 as biomarkers in breast cancer: validated for clinical ...
    Aug 22, 2014 · uPA and PAI-1 are among the best validated prognostic biomarkers currently available for lymph node-negative breast cancer.
  63. [63]
    Phase II randomised proof-of-concept study of the urokinase ...
    Feb 14, 2013 · This randomised trial is the first to evaluate the efficacy and safety of an uPA inhibitor administered to cancer patients. ... inhibitor WX-UK1.
  64. [64]
    Human antibody VH domains targeting uPAR as ... - Frontiers
    Oct 8, 2023 · Currently, several antibody-based therapies targeting uPAR performed in preclinical showed promising effects in breast cancer (31, 32) but there ...
  65. [65]
    Plasminogen Activator Inhibitor and the Risk of Cardiovascular ... - NIH
    We investigated, in the Framingham Heart Study offspring cohort, whether PAI-1 predicted CVD risk among individuals without prior CVD. Methods. Plasma PAI-1 ...Missing: overexpression | Show results with:overexpression
  66. [66]
    Plasminogen activator inhibitor and the risk of cardiovascular disease
    We investigated, in the Framingham Heart Study offspring cohort, whether PAI-1 predicted CVD risk among individuals without prior CVD. Methods: Plasma PAI-1 ...Missing: overexpression thrombosis stroke
  67. [67]
    [PDF] Plasminogen activator inhibitor (PAI)-1 in vascular inflammation and ...
    The Framingham Heart Study has shown a 2- to 10-fold excess in risk of CAD, stroke, heart failure, and death from. Page 2. PAI-1 in thrombosis. 2958. CVD among ...<|separator|>
  68. [68]
    Soluble Urokinase Plasminogen Activator Receptor | Stroke
    Nov 19, 2013 · Recent studies indicate that the urokinase system could have an important role in atherogenesis and plaque rupture.Missing: instability | Show results with:instability
  69. [69]
    Lack of urokinase plasminogen activator promotes progression and ...
    A lack of uPA also increased the size of and the luminal obstruction from atherosclerotic plaques at the coronary and brachiocephalic arteries of apoE(-/-) mice ...Missing: review | Show results with:review
  70. [70]
    Tissue-Plasminogen Activator Attenuates Alzheimer's Disease ...
    This study unravels a novel role of Activase rt-PA in counteracting the progression of AD-like pathology in APPswe/PS1 mice. These effects include Aβ clearance ...Missing: dual | Show results with:dual
  71. [71]
    Enhanced clearance of Aβ in brain by sustaining the plasmin ...
    Our data demonstrate that PAI-1 inhibitors augment the activity of tPA and plasmin in hippocampus, significantly lower plasma and brain Aβ levels.
  72. [72]
    Tissue plasminogen activator mediates amyloid‐induced ...
    This is the first demonstration of an intracellular pathway by which tPA triggers kinase activation, tau phosphorylation and neurotoxicity.Missing: dual | Show results with:dual
  73. [73]
    The Urokinase Plasminogen Activation System in Rheumatoid Arthritis
    Urokinase Plasminogen Activator (uPA) in particular is a pro-inflammatory mediator that appears to play an important role in the bone and cartilage destruction ...
  74. [74]
    Plasminogen System in the Pathophysiology of Sepsis - MDPI
    Aug 3, 2023 · The main inhibitor of fibrinolysis PAI-1 has been extensively investigated as a major player in the hypercoagulable state associated with ...Plasminogen System In The... · 2.2. Coagulation... · 2.3. Endothelial Dysfunction...
  75. [75]
    Effect of hypofibrinolysis on clinical outcomes of patients with septic ...
    For septic patients, fibrinolysis suppression presenting as “hypofibrinolysis” (elevated LI60) is associated with poor prognosis and risk of higher organ ...
  76. [76]
    Association between the plasminogen activator inhibitor-1 4G/5G ...
    The association between the PAI-1 4G/5G variant and VTE risk is controversial. •. We performed an updated meta-analysis to further investigate the issue. •. A ...
  77. [77]
    Plasminogen activator inhibitor-1 (PAI-1) 4G/5G promoter ...
    Jan 10, 2020 · Our meta-analysis suggested that PAI-1 4G/5G polymorphism is associated with the risk of venous thromboembolism, especially in Asians.
  78. [78]
  79. [79]
    The paradox of tPA in ischemic stroke: tPA knockdown following ...
    Our results provide evidence that reducing endogenous tPA following recanalization improves functional outcomes and reduces post-stroke brain damage.
  80. [80]
    Mesenchymal stem cell-derived extracellular vesicles attenuate tPA ...
    Our study demonstrated that MSC-EVs can act as an attractive thrombolytic adjuvant to repair the BBB and improve thrombolysis in a mouse ischemic stroke model.Missing: neurotoxicity | Show results with:neurotoxicity