Your browser doesn't support javascript.
loading
Packaging of supplemented urokinase into alpha-granules of in vitro-grown megakaryocytes for targeted nascent clot lysis.
Poncz, Mortimer; Zaitsev, Sergei; Ahn, Hyunsook; Kowalska, M Anna; Bdeir, Khalil; Dergilev, Konstantin Vladimirovich; Ivanciu, Lacramioara; Camire, Rodney M; Cines, Douglas B; Stepanova, Victoria.
  • Poncz M; Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.
  • Zaitsev S; Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Philadelphia, Pennsylvania, United States.
  • Ahn H; CHOP, Philadelphia, Pennsylvania, United States.
  • Kowalska MA; Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland.
  • Bdeir K; University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States.
  • Dergilev KV; National Medical Research Center of Cardiology after academician E. Chazov, Moscow, Russian Federation.
  • Ivanciu L; Children's Hospital of Philadelphia / University of Pennsylvania, Philadelphia, Pennsylvania, United States.
  • Camire RM; Children's Hospital of Philadelphia / University of Pennsylvania, Philadelphia, Pennsylvania, United States.
  • Cines DB; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, Pennsylvania, United States.
  • Stepanova V; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, Pennsylvania, United States.
Blood Adv ; 2024 May 28.
Article en En | MEDLINE | ID: mdl-38805575
ABSTRACT
Fibrinolytics delivered into the general circulation lack selectivity for nascent thrombi, reducing efficacy and increasing the risk of bleeding. Urokinase-type plasminogen activator (uPA) transgenically expressed within murine platelets provided targeted thromboprophylaxis without causing bleeding, but is clinically infeasible. Recent advances in generating megakaryocytes prompted us to develop a potentially clinically relevant means to produce "anti-thrombotic" platelets from CD34+ hematopoietic stem cell-derived in vitro-grown megakaryocytes. CD34+-megakaryocytes internalize and store in -granules single-chain uPA (scuPA) and a plasmin-resistant thrombin-activatable variant (uPAT). Both uPAs co-localized with internalized factor V (FV), fibrinogen and plasminogen, low-density lipoprotein receptor-related protein 1 (LRP1), and interferon-induced transmembrane protein 3, but not with endogenous von Willebrand factor (VWF). Endocytosis of uPA by CD34+-megakaryocytes was mediated, in part, via LRP1 and IIb3. scuPA-containing megakaryocytes degraded endocytosed intragranular FV, but not endogenous VWF in the presence of internalized plasminogen, whereas uPAT-megakaryocytes did not significantly degrade either protein. We used a carotid-artery injury model in NOD-scid IL2rnull (NSG) mice homozygous for VWFR1326H (a mutation switching binding VWF specificity from mouse to human glycoprotein Ib) to test whether platelets derived from scuPA- or uPAT-megakaryocytes would prevent thrombus formation. NSG/VWFR1326H mice exhibited a lower thrombotic burden after carotid artery injury compared to NSG mice unless infused with human platelets or megakaryocytes, whereas intravenous injection of uPA-megakaryocytes generated sufficient uPA-containing human platelets to lyse nascent thrombi. These studies describe the use of in vitro-generated megakaryocytes as a potential platform for delivering uPA or other ectopic proteins within platelet -granules to sites of vascular injury.

Texto completo: 1 Banco de datos: MEDLINE Idioma: En Año: 2024 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Idioma: En Año: 2024 Tipo del documento: Article