Your browser doesn't support javascript.
loading
In-depth structure-function profiling of the complex formation between clotting factor VIII and heme.
Hopp, Marie-T; Ugurlar, Deniz; Pezeshkpoor, Behnaz; Biswas, Arijit; Ramoji, Anuradha; Neugebauer, Ute; Oldenburg, Johannes; Imhof, Diana.
Afiliação
  • Hopp MT; Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, Bonn, Germany; Department of Chemistry, Institute for Integrated Natural Sciences, University of Koblenz, Koblenz, Germany. Electronic address: mhopp@uni-koblenz.de.
  • Ugurlar D; Center for Electron Microscopy, Thermo Fisher Scientific, Eindhoven, the Netherlands.
  • Pezeshkpoor B; Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany.
  • Biswas A; Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany.
  • Ramoji A; Leibniz Institute of Photonic Technology, Jena, Germany; Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany.
  • Neugebauer U; Leibniz Institute of Photonic Technology, Jena, Germany; Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.
  • Oldenburg J; Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany.
  • Imhof D; Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, Bonn, Germany. Electronic address: dimhof@uni-bonn.de.
Thromb Res ; 237: 184-195, 2024 May.
Article em En | MEDLINE | ID: mdl-38631156
ABSTRACT
BACKGROUND AND

AIMS:

Blood disorders, such as sickle cell disease, and other clinical conditions are often accompanied by intravascular hemolytic events along with the development of severe coagulopathies. Hemolysis, in turn, leads to the accumulation of Fe(II/III)-protoporphyrin IX (heme) in the intravascular compartment, which can trigger a variety of proinflammatory and prothrombotic reactions. As such, heme binding to the blood coagulation proteins factor VIII (FVIII), fibrinogen, and activated protein C with functional consequences has been demonstrated earlier.

METHODS:

We herein present an in-depth characterization of the FVIII-heme interaction at the molecular level and its (patho-)physiological relevance through the application of biochemical, biophysical, structural biology, bioinformatic, and diagnostic tools.

RESULTS:

FVIII has a great heme-binding capacity with seven heme molecules associating with the protein. The respective binding sites were identified by investigating heme binding to FVIII-derived peptides in combination with molecular docking and dynamic simulation studies of the complex as well as cryo-electron microscopy, revealing three high-affinity and four moderate heme-binding motifs (HBMs). Furthermore, the relevance of the FVIII-heme complex formation was characterized in physiologically relevant assay systems, revealing a ~ 50 % inhibition of the FVIII cofactor activity even in the protein-rich environment of blood plasma.

CONCLUSION:

Our study provides not only novel molecular insights into the FVIII-heme interaction and its physiological relevance, but also strongly suggests the reduction of the intrinsic pathway and the accentuation of the final clotting step (by, for example, fibrinogen crosslinking) in hemolytic conditions as well as a future perspective in the context of FVIII substitution therapy of hemorrhagic events in hemophilia A patients.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Fator VIII / Heme Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Fator VIII / Heme Idioma: En Ano de publicação: 2024 Tipo de documento: Article