ER-to-Golgi transport of coagulation factors V and VIII Coagulation factor V (FV) and factor VIII (FVIII) are both secreted glycoproteins that share pivotal roles in both hemostasis and thrombosis. Genetic deficiency of FVIII results in hemophilia A, which affects ~1 in 5000 males. On the other hand, a gain-of-function mutation in FV (FV Leiden) and increased FVIII activity are major risk factors for venous thrombosis, which affects ~1:1,000 individuals in the US per year. Although many secreted proteins (referred to as cargo) are believed to require transport receptors for efficient endoplasmic reticulum (ER)-to-Golgi transport, only a limited number of such receptors have been described, mostly in yeast. Evidence for the existence of mammalian cargo receptors came unexpectedly from studies of the human genetic disorder combined deficiency of FV and FVIII (F5F8D), which identified mutations in LMAN1 and MCFD2 as the cause of the disorder. F5F8D is a rare bleeding disorder characterized by the reduction of both FV and FVIII to 5-30% of normal. LMAN1 and MCFD2 form a Ca2+-dependent protein complex in the ER- Golgi intermediate compartment that interacts with FV and FVIII, suggesting that the LMAN1-MCFD2 complex is a cargo receptor required for efficient transport of FV and FVIII from the ER to the Golgi. The requirement of both a transmembrane component (LMAN1) and a soluble cofactor (MCFD2) suggests a more sophisticated mechanism for cargo trafficking in higher eukaryotes, and could represent a paradigm for the organization of other mammalian cargo receptors. In this proposal, we will study the mechanism of receptor-mediated ER-to- Golgi transport of FV and FVIII.
In aim 1, we will identify N-linked glycosylation sites on FV/FVIII that interact with LMAN1, investigate Ca2+ in regulating the binding and release of cargo, and increase in vitro FVIII production by enhancing the LMAN1-MCFD2 secretion pathway.
In aim 2, we will identify sorting signals in FVIII that are recognized by MCFD2, investigate the ER-to-Golgi transport deficiency as a novel mechanism of hemophilia A, and identify other components/alternative pathways that control FV/FVIII trafficking.
In aim 3, we will test the LMAN1-MCFD2 secretion pathway as a therapeutic target for thrombophilia and create a mouse model to characterize the role of B domain in FVIII biosynthesis and LMAN1-MCFD2 mediated secretion in vivo. Results will not only answer fundamental questions regarding the mechanism of LMAN1-MCFD2 receptor-mediated secretion of FV and FVIII, but will also provide fundamental new insight into general mechanism of mammalian ER-to-Golgi protein transport. The findings will have practical importance for improving FVIII expression and may expedite the eventual goal of somatic cell gene therapy for hemophilia A, as well as new approaches to limiting FV and FVIII production in prothrombotic states.

Public Health Relevance

The project will study a protein transport receptor that is required for the secretion of blood clotting factor V and factor VIII. The goal is to understand how this transport receptor works and hopefully use the knowledge to develop better treatment for bleeding and thrombotic diseases.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL094505-10
Application #
9738782
Study Section
Hemostasis and Thrombosis Study Section (HT)
Program Officer
Sarkar, Rita
Project Start
2008-12-26
Project End
2023-06-30
Budget Start
2019-07-01
Budget End
2020-06-30
Support Year
10
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Cleveland Clinic Lerner
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
135781701
City
Cleveland
State
OH
Country
United States
Zip Code
44195
Khoriaty, Rami; Hesketh, Geoffrey G; Bernard, Amélie et al. (2018) Functions of the COPII gene paralogs SEC23A and SEC23B are interchangeable in vivo. Proc Natl Acad Sci U S A 115:E7748-E7757
Wei, Wei; Misra, Saurav; Cannon, Matthew V et al. (2018) Molecular mechanisms of missense mutations that generate ectopic N-glycosylation sites in coagulation factor VIII. Biochem J 475:873-886
Zhu, Min; Zheng, Chunlei; Wei, Wei et al. (2018) Analysis of MCFD2- and LMAN1-deficient mice demonstrates distinct functions in vivo. Blood Adv 2:1014-1021
Wei, Wei; Zheng, Chunlei; Zhu, Min et al. (2017) Missense mutations near the N-glycosylation site of the A2 domain lead to various intracellular trafficking defects in coagulation factor VIII. Sci Rep 7:45033
Khoriaty, Rami; Everett, Lesley; Chase, Jennifer et al. (2016) Pancreatic SEC23B deficiency is sufficient to explain the perinatal lethality of germline SEC23B deficiency in mice. Sci Rep 6:27802
Simeoni, Ilenia; Stephens, Jonathan C; Hu, Fengyuan et al. (2016) A high-throughput sequencing test for diagnosing inherited bleeding, thrombotic, and platelet disorders. Blood 127:2791-803
Yehia, Lamis; Niazi, Farshad; Ni, Ying et al. (2015) Germline Heterozygous Variants in SEC23B Are Associated with Cowden Syndrome and Enriched in Apparently Sporadic Thyroid Cancer. Am J Hum Genet 97:661-76
Zhu, Min; Tao, Jiayi; Vasievich, Matthew P et al. (2015) Neural tube opening and abnormal extraembryonic membrane development in SEC23A deficient mice. Sci Rep 5:15471
Hao, Hong; Gregorski, Janina; Qian, Haohua et al. (2014) In vivo function of the ER-Golgi transport protein LMAN1 in photoreceptor homeostasis. Adv Exp Med Biol 801:395-9
Khoriaty, Rami; Vasievich, Matthew P; Jones, Morgan et al. (2014) Absence of a red blood cell phenotype in mice with hematopoietic deficiency of SEC23B. Mol Cell Biol 34:3721-34

Showing the most recent 10 out of 22 publications