Google Scholar citation report
Citations : 14978

Journal of Basic and Clinical Pharmacy received 14978 citations as per google scholar report

Indexed In

William Thore*
 
Department of Biomedical Sciences, Harvard University, Boston, USA
 
*Correspondence: William Thore, Department of Biomedical Sciences, Harvard University, Boston, USA, Email: william@thore.edu

Received: 29-Mar-2024, Manuscript No. Jbclinphar-24-134014; Editor assigned: 01-Apr-2024, Pre QC No. Jbclinphar-24-134014 (PQ); Reviewed: 15-Apr-2024 QC No. Jbclinphar-24-134014; Revised: 22-Apr-2024, Manuscript No. Jbclinphar-24-134014 (R); Published: 29-Apr-2024

Citation: Thore W. The Impact of Fibrinogen in Coagulation and Tissue Repair Mechanisms. J Basic Clin Pharma.2024,15(2):344.

This open-access article is distributed under the terms of the Creative Commons Attribution Non-Commercial License (CC BY-NC) (http://creativecommons.org/licenses/by-nc/4.0/), which permits reuse, distribution and reproduction of the article, provided that the original work is properly cited and the reuse is restricted to noncommercial purposes. For commercial reuse, contact reprints@jbclinpharm.org

Description

Fibrinogen, also known as coagulation factor I, stands as a foundation in the complex flow of blood coagulation. Beyond its conventional role in hemostasis, fibrinogen exerts multifaceted functions in various physiological and pathological processes. This overview searches into the essential role of fibrinogen in coagulation and its broader functioning, focusing on its diverse implications in health and disease. Fibrinogen, a crucial glycoprotein predominantly synthesized in the liver, consists of a three pairs of polypeptide chains: Two Aα, two Bβ, and two γ chains, intricately linked by disulfide bonds. Its assembly undergoes meticulous post-translational modifications and intracellular trafficking, ensuring proper structure and function. Upon release into the bloodstream, fibrinogen remains in an inactive state, ready to be activated upon encountering injury or pathological triggers. This inactive circulating form stands ready to fastly respond to vascular damage or aberrant stimuli, initiating the series of events leading to hemostasis and clot formation.

Fibrinogen stands as a crucial element in the culmination of the coagulation cascade, orchestrating the formation of durable fibrin clots. When vascular injury occurs, the sequential activation of coagulation factors causes thrombin’s enzymatic action, catalyzing the conversion of fibrinogen into fibrin. Subsequently, fibrin molecules undergo polymerization, interweaving into a complex mesh-like network. This fibrin mesh stabilizes platelet aggregates, effectively sealing off the site of injury and staunching blood loss. Through this meticulous process, fibrinogen ensures the swift and efficient formation of clots, essential for maintaining hemostasis and preventing excessive bleeding. Beyond its role in hemostasis, fibrinogen exhibits diverse functions in inflammation, wound healing, and tissue repair. Fibrinogen serves as a support system for cellular adhesion and migration, facilitating the recruitment of inflammatory cells to sites of tissue injury. Moreover, fibrinogen interacts with various cell surface receptors, modulating cell signaling pathways and immune responses.

Dysregulation of fibrinogen metabolism is associated with various pathological conditions, including thrombosis, hemorrhage, and inflammatory disorders. Elevated plasma fibrinogen levels are a risk factor for arterial and venous thrombosis, contributing to the pathogenesis of cardiovascular diseases. Conversely, congenital deficiencies or acquired disorders affecting fibrinogen synthesis or function can predispose individuals to bleeding disorders, such as afibrinogenemia or dysfibrinogenemia. Given its central role in coagulation and thrombosis, fibrinogen represents a potential target for therapeutic intervention in various clinical scenarios. Pharmacological agents targeting fibrinogen or its downstream mediators are under investigation for the prevention and treatment of thrombotic disorders. Additionally, fibrinogen supplementation therapy is utilized in the management of bleeding disorders associated with fibrinogen deficiency.

Advances in molecular biology and biotechnology offer new ways for studying fibrinogen structure-function relationships and developing novel therapeutic strategies. Elucidating the complex mechanisms managing the fibrinogen metabolism and its interactions with cellular components shows potential for revealing its more extensive physiological involvement and pathological implications. Furthermore, personalized approaches to fibrinogen-targeted therapy may enhance treatment efficacy and minimize adverse effects in individuals with coagulation disorders. Fibrinogen emerges as a central player in the complex network of blood coagulation, exerting diverse functions beyond its traditional role in hemostasis. Understanding the complexities of fibrinogen biology is crucial for understanding its implications in health and disease, paving the way for innovative therapeutic interventions and personalized medicine approaches in coagulation disorders.