Back to Search Start Over

Targeting STUB1–tissue factor axis normalizes hyperthrombotic uremic phenotype without increasing bleeding risk

Authors :
Institute for Medical Engineering and Science
Edelman, Elazer R.
Balcells-Camps, Mercedes
Kolandaivelu, Kumaran
Edelman, Elazer R
Shashar, Moshe
Belghasem, Mostafa E.
Matsuura, Shinobu
Walker, Joshua
Richards, Sean
Alousi, Faisal
Rijal, Keshab
Kolachalama, Vijaya B.
Odagi, Minami
Nagasawa, Kazuo
Henderson, Joel M.
Gautam, Amitabh
Rushmore, Richard
Francis, Jean
Kirchhofer, Daniel
Sherr, David H.
Ravid, Katya
Chitalia, Vipul C.
Institute for Medical Engineering and Science
Edelman, Elazer R.
Balcells-Camps, Mercedes
Kolandaivelu, Kumaran
Edelman, Elazer R
Shashar, Moshe
Belghasem, Mostafa E.
Matsuura, Shinobu
Walker, Joshua
Richards, Sean
Alousi, Faisal
Rijal, Keshab
Kolachalama, Vijaya B.
Odagi, Minami
Nagasawa, Kazuo
Henderson, Joel M.
Gautam, Amitabh
Rushmore, Richard
Francis, Jean
Kirchhofer, Daniel
Sherr, David H.
Ravid, Katya
Chitalia, Vipul C.
Source :
Edelman
Publication Year :
2018

Abstract

Chronic kidney disease (CKD/uremia) remains vexing because it increases the risk of atherothrombosis and is also associated with bleeding complications on standard antithrombotic/antiplatelet therapies. Although the associations of indolic uremic solutes and vascular wall proteins [such as tissue factor (TF) and aryl hydrocarbon receptor (AHR)] are being defined, the specific mechanisms that drive the thrombotic and bleeding risks are not fully understood. We now present an indolic solute-specific animal model, which focuses on solute-protein interactions and shows that indolic solutes mediate the hyperthrombotic phenotype across all CKD stages in an AHR- and TF-dependent manner. We further demonstrate that AHR regulates TF through STIP1 homology and U-box-containing protein 1 (STUB1). As a ubiquitin ligase, STUB1 dynamically interacts with and degrades TF through ubiquitination in the uremic milieu. TF regulation by STUB1 is supported in humans by an inverse relationship of STUB1 and TF expression and reduced STUB1-TF interaction in uremic vessels. Genetic or pharmacological manipulation of STUB1 in vascular smooth muscle cells inhibited thrombosis in flow loops. STUB1 perturbations reverted the uremic hyperthrombotic phenotype without prolonging the bleeding time, in contrast to heparin, the standard-of-care antithrombotic in CKD patients. Our work refines the thrombosis axis (STUB1 is a mediator of indolic solute-AHR-TF axis) and expands the understanding of the interconnected relationships driving the fragile thrombotic state in CKD. It also establishes a means of minimizing the uremic hyperthrombotic phenotype without altering the hemostatic balance, a long-sought-after combination in CKD patients.<br />National Institutes of Health (U.S.) (Grant R01HL132325)<br />National Institutes of Health (U.S.) (Grant R01CA175382)<br />National Institute of General Medical Sciences (U.S.) (Grant R01GM49039)<br />American Heart Association (Grant 12FTF12080241)

Details

Database :
OAIster
Journal :
Edelman
Notes :
application/pdf
Publication Type :
Electronic Resource
Accession number :
edsoai.on1141890928
Document Type :
Electronic Resource