The overall goal of this grant has been to understand in molecular details the action of apolipoproteins (apos) and lipoproteins in health and disease. The current focus is on human apoA-I, the major protein of plasma high-density lipoproteins (HDL, a.k.a. good cholesterol) that remove cholesterol from cells and protect against cardiovascular disease. ApoA-I can be released from HDL in a labile lipid-poor/free state that is the precursor of amyloid, and can cause two forms of systemic human amyloidosis. In the acquired form, apoA-I deposits in the arteries as fibrils, which augments atherosclerosis. In the hereditary form, fragments of mutant apoA-I deposit in vital organs (kidney, liver, nerves, etc.) and damage them. There is no cure for this life-threatening disease and the only current treatment is organ transplant. To pinpoint therapeutic targets, we must understand what determines the dynamic equilibrium between the generation of the amyloid precursor, its clearance, and its misfolding, from the native helix-bundle structure in free apoA-I to the insoluble cross-?-sheet in amyloid. Unraveling the complex process of protein misfolding and proteostasis to block systemic amyloidosis has been very challenging but not impossible. To this end we have integrated high- and low-resolution structural and spectroscopic methods (circular dichroism, fluorescence, hydrogen-deuterium exchange, etc.) with biochemical and computational tools. Analysis of several disease-causing mutants enabled us to propose the first molecular mechanism of apoA-I misfolding. We postulated that perturbed packing in amyloid `hot spots' combined with the structural integrity of the native fold make the protein amyloidogenic. This idea can be extended to other proteins; it helps explain why structural destabilization of many globular proteins is neither necessary nor sufficient to cause amyloid disease. These and other new ideas will be rigorously tested in the next cycle of this grant.
Aim 1 will elucidate the molecular mechanism of apoA-I misfolding in hereditary amyloidosis. We will determine how the disease-causing mutations perturb the native protein conformation in sensitive segments to promote ?-aggregation vs. proteolysis. Cell-based studies will unveil why mutation carriers are at a low risk of atherosclerosis despite low levels of plasma HDL.
Aim 2 will identify the similarities and differences in the molecular basis for hereditary and acquired amyloidoses, and will establish the structure-toxicity relationship in aggregated forms of apoA-I.
Aim 3 will define the role of lipids in apoA-I misfolding.
This aim will test our hypothesis that lipid-lowering approaches hold therapeutic potential for apoA-I amyloidoses. This research opens a new frontier that goes beyond protein stability to identify key drivers of protein misfolding in vivo. The results will unveil the link between amyloidogenic and cardioprotective properties of apoA-I, help find therapies for apoA-I amyloidoses, yield sharper insights into the misfolding of other apos that are prominent in human amyloidoses, and have broad implications for misfolding diseases caused by other globular proteins.

Public Health Relevance

This research will determine how the functional soluble protein constituent of Good Cholesterol misfolds and forms fibrils that deposit in vital organs (kidneys, liver, heart, testes, nerves, skin and other organs), leading to organ damage. This research will also help determine how to delay or block this damaging process. The results will help find the much-needed therapies for devastating human diseases such as familial and acquired amyloidosis and atherosclerosis.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM067260-18
Application #
9999601
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Sakalian, Michael
Project Start
1998-12-15
Project End
2021-08-31
Budget Start
2020-09-01
Budget End
2021-08-31
Support Year
18
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Boston University
Department
Physiology
Type
Schools of Medicine
DUNS #
604483045
City
Boston
State
MA
Country
United States
Zip Code
02118
Klimtchuk, Elena S; Prokaeva, Tatiana; Frame, Nicholas M et al. (2018) Unusual duplication mutation in a surface loop of human transthyretin leads to an aggressive drug-resistant amyloid disease. Proc Natl Acad Sci U S A 115:E6428-E6436
Jayaraman, Shobini; Gantz, Donald L; Haupt, Christian et al. (2018) Serum amyloid A sequesters diverse phospholipids and their hydrolytic products, hampering fibril formation and proteolysis in a lipid-dependent manner. Chem Commun (Camb) 54:3532-3535
Wilson, Christopher J; Das, Madhurima; Jayaraman, Shobini et al. (2018) Effects of Disease-Causing Mutations on the Conformation of Human Apolipoprotein A-I in Model Lipoproteins. Biochemistry 57:4583-4596
Jayaraman, Shobini; Sánchez-Quesada, Jose Luis; Gursky, Olga (2017) Triglyceride increase in the core of high-density lipoproteins augments apolipoprotein dissociation from the surface: Potential implications for treatment of apolipoprotein deposition diseases. Biochim Biophys Acta Mol Basis Dis 1863:200-210
Jayaraman, Shobini; Gantz, Donald L; Haupt, Christian et al. (2017) Serum amyloid A forms stable oligomers that disrupt vesicles at lysosomal pH and contribute to the pathogenesis of reactive amyloidosis. Proc Natl Acad Sci U S A 114:E6507-E6515
Frame, Nicholas M; Jayaraman, Shobini; Gantz, Donald L et al. (2017) Serum amyloid A self-assembles with phospholipids to form stable protein-rich nanoparticles with a distinct structure: A hypothetical function of SAA as a ""molecular mop"" in immune response. J Struct Biol 200:293-302
Madico, Guillermo; Gursky, Olga; Fairman, Jeff et al. (2017) Structural and Immunological Characterization of Novel Recombinant MOMP-Based Chlamydial Antigens. Vaccines (Basel) 6:
Prokaeva, Tatiana; Akar, Harun; Spencer, Brian et al. (2017) Hereditary Renal Amyloidosis Associated With a Novel Apolipoprotein A-II Variant. Kidney Int Rep 2:1223-1232
Klimtchuk, Elena S; Prokaeva, Tatiana B; Spencer, Brian H et al. (2017) In vitro co-expression of human amyloidogenic immunoglobulin light and heavy chain proteins: a relevant cell-based model of AL amyloidosis. Amyloid 24:115-122
Frame, Nicholas M; Gursky, Olga (2017) Structure of serum amyloid A suggests a mechanism for selective lipoprotein binding and functions: SAA as a hub in macromolecular interaction networks. Amyloid 24:13-14

Showing the most recent 10 out of 53 publications